Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 365
Filter
1.
Nat Commun ; 15(1): 4484, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802340

ABSTRACT

Deciphering the intricate dynamic events governing type I interferon (IFN) signaling is critical to unravel key regulatory mechanisms in host antiviral defense. Here, we leverage TurboID-based proximity labeling coupled with affinity purification-mass spectrometry to comprehensively map the proximal human proteomes of all seven canonical type I IFN signaling cascade members under basal and IFN-stimulated conditions. This uncovers a network of 103 high-confidence proteins in close proximity to the core members IFNAR1, IFNAR2, JAK1, TYK2, STAT1, STAT2, and IRF9, and validates several known constitutive protein assemblies, while also revealing novel stimulus-dependent and -independent associations between key signaling molecules. Functional screening further identifies PJA2 as a negative regulator of IFN signaling via its E3 ubiquitin ligase activity. Mechanistically, PJA2 interacts with TYK2 and JAK1, promotes their non-degradative ubiquitination, and limits the activating phosphorylation of TYK2 thereby restraining downstream STAT signaling. Our high-resolution proximal protein landscapes provide global insights into the type I IFN signaling network, and serve as a valuable resource for future exploration of its functional complexities.


Subject(s)
Interferon Type I , Janus Kinase 1 , Receptor, Interferon alpha-beta , STAT2 Transcription Factor , Signal Transduction , TYK2 Kinase , Ubiquitination , Humans , HEK293 Cells , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Janus Kinase 1/metabolism , Phosphorylation , Proteome/metabolism , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , TYK2 Kinase/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
Fish Shellfish Immunol ; 150: 109656, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38801844

ABSTRACT

Interferon regulatory factor 11 (IRF11), a fish specific member of IRF family, is a transcription factor known for its positive role in teleost antiviral defense by regulating IFN expression. Despite its recognized function, the precise mechanism of IRF11 in type I IFNs production remains largely unknown. In this study, we identified IRF11 in Japanese eel, Anguilla japonica, (AjIRF11) and determined its involvement in the later phase of fish IFN production. Our results demonstrate that IRF11-induced IFN production operates through ISRE binding. Mutations in each ISRE site within the promoter of AjIFN2 or AjIFN4 abolished IRF11-mediated activation of IFN promoters. In addition, the overexpression of AjIRF11 does not significantly impact the activation of AjIFN promoters induced by RLR-related signaling pathway proteins. Furthermore, IRF11-knockdown in ZFLs (zebrafish liver cells) has no effect on the RLRs-induced expression of zebrafish IFN-φ1 and IFN-φ3, indicating that IRF11 is not involved in the RLR-mediated IFN production. However, AjIRF11 can form transcription complexes with AjSTAT1 or AjSTAT2, or form homo- or heterodimers with AjIRF1 to stimulate the transcription of type I IFNs. Overall, it is shown in this study that IRF11 can act synergistically with STAT1 and/or STAT2 for the induction of IFN.


Subject(s)
Anguilla , Fish Proteins , Interferon Type I , STAT1 Transcription Factor , STAT2 Transcription Factor , Animals , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Interferon Type I/genetics , Interferon Type I/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Anguilla/genetics , Anguilla/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/immunology , Fish Diseases/immunology , Immunity, Innate/genetics , Zebrafish/genetics , Zebrafish/immunology , Gene Expression Regulation/immunology
3.
Curr Opin Immunol ; 87: 102427, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38781720

ABSTRACT

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


Subject(s)
Interferon Lambda , Interferons , Virus Diseases , Humans , Interferons/metabolism , Interferons/immunology , Virus Diseases/immunology , Animals , Signal Transduction/immunology , STAT2 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Interleukin-10 Receptor beta Subunit/genetics , Interleukin-10 Receptor beta Subunit/immunology , Interleukin-10 Receptor beta Subunit/metabolism
4.
Am J Physiol Cell Physiol ; 326(6): C1697-C1709, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38586875

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia and presents a considerable disease burden. Its pathology involves substantial neuronal loss, primarily attributed to neuronal apoptosis. Although sirtuin 4 (SIRT4) has been implicated in regulating apoptosis in various diseases, the role of SIRT4 in AD pathology remains unclear. The study used APP/PS1 mice as an animal model of AD and amyloid-ß (Aß)1-42-treated HT-22 cells as an AD cell model. SIRT4 expression was determined by quantitative real-time polymerase chain reaction, Western blot, and immunofluorescence. A Sirt4 knockdown model was established by intracranial injection of lentivirus-packaged sh-SIRT4 and cellular lentivirus transfection. Immunohistochemistry and flow cytometry were used to examine Aß deposition in mice and apoptosis, respectively. Protein expression was assessed by Western blot analysis. The UCSC and JASPAR databases were used to predict upstream transcription factors of Sirt4. Subsequently, the binding of transcription factors to Sirt4 was analyzed using a dual-luciferase assay and chromatin immunoprecipitation. SIRT4 expression was upregulated in both APP/PS1 mice and Aß-treated HT-22 cells compared with their respective control groups. Sirt4 knockdown in animal and cellular models of AD resulted in reduced apoptosis, decreased Aß deposition, and amelioration of learning and memory impairments in mice. Mechanistically, SIRT4 modulates apoptosis via the mTOR pathway and is negatively regulated by the transcription factor signal transducer and activator of transcription 2 (STAT2). Our study findings suggest that targeting the STAT2-SIRT4-mTOR axis may offer a new treatment approach for AD.NEW & NOTEWORTHY The study reveals that in Alzheimer's disease models, SIRT4 expression increases, contributing to neuronal apoptosis and amyloid-ß deposition. Reducing SIRT4 lessens apoptosis and amyloid-ß accumulation, improving memory in mice. This process involves the mTOR pathway, regulated by STAT2 transcription factor. These findings suggest targeting the STAT2-SIRT4-mTOR axis as a potential Alzheimer's treatment strategy.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Apoptosis , Disease Models, Animal , Mice, Transgenic , Neurons , STAT2 Transcription Factor , Signal Transduction , Sirtuins , TOR Serine-Threonine Kinases , Animals , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Sirtuins/metabolism , Sirtuins/genetics , TOR Serine-Threonine Kinases/metabolism , Mice , Neurons/metabolism , Neurons/pathology , STAT2 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , Amyloid beta-Peptides/metabolism , Humans , Male , Mice, Inbred C57BL , Cell Line , Mitochondrial Proteins
5.
Dev Cell ; 59(8): 1028-1042.e5, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38452758

ABSTRACT

The interferon signaling pathway is critical for host defense by serving diverse functions in both innate and adaptive immune responses. Here, we show that type I gamma phosphatidylinositol phosphate 5-kinase i5 (PIPKIγi5), an enzyme that synthesizes phosphatidylinositol-4,5-bisphosphate (PI4,5P2), controls the sensitivity to interferon in both human and mouse cells. PIPKIγi5 directly binds to the interferon-gamma (IFN-γ) downstream effector signal transducer and activator of transcription 1 (STAT1), which suppresses the STAT1 dimerization, IFN-γ-induced STAT1 nuclear translocation, and transcription of IFN-γ-responsive genes. Depletion of PIPKIγi5 significantly enhances IFN-γ signaling and strengthens an antiviral response. In addition, PIPKIγi5-synthesized PI4,5P2 can bind to STAT1 and promote the PIPKIγi5-STAT1 interaction. Similar to its interaction with STAT1, PIPKIγi5 is capable of interacting with other members of the STAT family, including STAT2 and STAT3, thereby suppressing the expression of genes mediated by these transcription factors. These findings identify the function of PIPKIγi5 in immune regulation.


Subject(s)
Interferon-gamma , Phosphotransferases (Alcohol Group Acceptor) , Signal Transduction , Animals , Humans , Mice , HEK293 Cells , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
6.
Immunology ; 172(3): 440-450, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38514903

ABSTRACT

Analysis of genetically defined immunodeficient patients allows study of the effect of the absence of specific proteins on human immune function in real-world conditions. Here we have addressed the importance of type I interferon signalling for human NK cell development by studying the phenotype and function of circulating NK cells isolated from patients suffering primary immunodeficiency disease due to mutation of either the human interferon regulatory factor 9 (IRF9) or the signal transducer and activator of transcription 2 (STAT2) genes. IRF9, together with phosphorylated STAT1 and STAT2, form a heterotrimer called interferon stimulated gene factor 3 (ISGF3) which promotes the expression of hundreds of IFN-stimulated genes that mediate antiviral function triggered by exposure to type I interferons. IRF9- and STAT2-deficient patients are unable to respond efficiently to stimulation by type I interferons and so our experiments provide insights into the importance of type I interferon signalling and the consequences of its impairment on human NK cell biology. Surprisingly, the NK cells of these patients display essentially normal phenotype and function.


Subject(s)
Interferon Type I , Interferon-Stimulated Gene Factor 3, gamma Subunit , Killer Cells, Natural , STAT2 Transcription Factor , Signal Transduction , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , STAT2 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon Type I/metabolism , Mutation , Cell Differentiation , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Cells, Cultured
7.
Fish Shellfish Immunol ; 148: 109510, 2024 May.
Article in English | MEDLINE | ID: mdl-38521143

ABSTRACT

The signal transducer and activator of transcription 2 (STAT2), a downstream factor of type I interferons (IFNs), is a key component of the cellular antiviral immunity response. However, the role of STAT2 in the upstream of IFN signaling, such as the regulation of pattern recognition receptors (PRRs), remains unknown. In this study, STAT2 homologue of black carp (Mylopharyngodon piceus) has been cloned and characterized. The open reading frame (ORF) of bcSTAT2 comprises 2523 nucleotides and encodes 841 amino acids, which presents the conserved structure to that of mammalian STAT2. The dual-luciferase reporter assay and the plaque assay showed that bcSTAT2 possessed certain IFN-inducing ability and antiviral ability against both spring viremia of carp virus (SVCV) and grass carp reovirus (GCRV). Interestingly, we detected the association between bcSTAT2 and bcRIG-I through co-immunoprecipitation (co-IP) assay. Moreover, when bcSTAT2 was co-expressed with bcRIG-I, bcSTAT2 obviously suppressed bcRIG-I-induced IFN expression and antiviral activity. The subsequent co-IP assay and immunoblotting (IB) assay further demonstrated that bcSTAT2 inhibited K63-linked polyubiquitination but not K48-linked polyubiquitination of bcRIG-I, however, did not affect the oligomerization of bcRIG-I. Thus, our data conclude that black carp STAT2 negatively regulates RIG-I through attenuates its K63-linked ubiquitination, which sheds a new light on the regulation of the antiviral innate immunity cascade in vertebrates.


Subject(s)
Carps , Fish Diseases , Reoviridae Infections , Reoviridae , Rhabdoviridae Infections , Animals , Carps/genetics , Carps/metabolism , Rhabdoviridae Infections/veterinary , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Reoviridae/physiology , Immunity, Innate/genetics , Fish Proteins , Mammals/metabolism
8.
Front Immunol ; 15: 1331731, 2024.
Article in English | MEDLINE | ID: mdl-38384473

ABSTRACT

The establishment of a virus infection is the result of the pathogen's ability to replicate in a hostile environment generated by the host's immune system. Here, we found that ISG15 restricts Dengue and Zika viruses' replication through the stabilization of its binding partner USP18. ISG15 expression was necessary to control DV replication driven by both autocrine and paracrine type one interferon (IFN-I) signaling. Moreover, USP18 competes with NS5-mediated STAT2 degradation, a major mechanism for establishment of flavivirus infection. Strikingly, reconstitution of USP18 in ISG15-deficient cells was sufficient to restore the STAT2's stability and restrict virus growth, suggesting that the IFNAR-mediated ISG15 activity is also antiviral. Our results add a novel layer of complexity in the virus/host interaction interface and suggest that NS5 has a narrow window of opportunity to degrade STAT2, therefore suppressing host's IFN-I mediated response and promoting virus replication.


Subject(s)
Dengue , Interferon Type I , Zika Virus Infection , Zika Virus , Humans , Interferon Type I/metabolism , Zika Virus Infection/genetics , Virus Replication , Dengue/genetics , Ubiquitins/metabolism , Cytokines/metabolism , Ubiquitin Thiolesterase/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism
9.
Commun Biol ; 7(1): 76, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38195857

ABSTRACT

Flaviviruses, including Zika virus (ZIKV) and Dengue virus (DENV), rely on their non-structural protein 5 (NS5) for both replication of viral genome and suppression of host IFN signaling. DENV and ZIKV NS5s were shown to facilitate proteosome-mediated protein degradation of human STAT2 (hSTAT2). However, how flavivirus NS5s have evolved for species-specific IFN-suppression remains unclear. Here we report structure-function characterization of the DENV serotype 2 (DENV2) NS5-hSTAT2 complex. The MTase and RdRP domains of DENV2 NS5 form an extended conformation to interact with the coiled-coil and N-terminal domains of hSTAT2, thereby promoting hSTAT2 degradation in cells. Disruption of the extended conformation of DENV2/ZIKV NS5, but not the alternative compact state, impaired their hSTAT2 binding. Our comparative structural analysis of flavivirus NS5s further reveals a conserved protein-interaction platform with subtle amino-acid variations likely underpinning diverse IFN-suppression mechanisms. Together, this study uncovers a conformational selection mechanism underlying species-specific hSTAT2 inhibition by flavivirus NS5.


Subject(s)
Flavivirus , STAT2 Transcription Factor , Viral Nonstructural Proteins , Zika Virus Infection , Zika Virus , Humans , Proteolysis , Species Specificity , STAT2 Transcription Factor/metabolism , Viral Nonstructural Proteins/metabolism
10.
Gut ; 73(2): 282-297, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37884352

ABSTRACT

OBJECTIVE: We sought to investigate the role of interleukin (IL)-20 in IBD and experimental colitis. DESIGN: Experimental colitis was induced in mice deficient in components of the IL-20 and signal transducer and activator of transcription (STAT)2 signalling pathways. In vivo imaging, high-resolution mini-endoscopy and histology were used to assess intestinal inflammation. We further used RNA-sequencing (RNA-Seq), RNAScope and Gene Ontology analysis, western blot analysis and co-immunoprecipitation, confocal microscopy and intestinal epithelial cell (IEC)-derived three-dimensional organoids to investigate the underlying molecular mechanisms. Results were validated using samples from patients with IBD and non-IBD control subjects by a combination of RNA-Seq, organoids and immunostainings. RESULTS: In IBD, IL20 levels were induced during remission and were significantly higher in antitumour necrosis factor responders versus non-responders. IL-20RA and IL-20RB were present on IECs from patients with IBD and IL-20-induced STAT3 and suppressed interferon (IFN)-STAT2 signalling in these cells. In IBD, experimental dextran sulfate sodium (DSS)-induced colitis and mucosal healing, IECs were the main producers of IL-20. Compared with wildtype controls, Il20-/-, Il20ra-/- and Il20rb-/- mice were more susceptible to experimental DSS-induced colitis. IL-20 deficiency was associated with increased IFN/STAT2 activity in mice and IFN/STAT2-induced necroptotic cell death in IEC-derived organoids could be markedly blocked by IL-20. Moreover, newly generated Stat2ΔIEC mice, lacking STAT2 in IECs, were less susceptible to experimental colitis compared with wildtype controls and the administration of IL-20 suppressed colitis activity in wildtype animals. CONCLUSION: IL-20 controls colitis and mucosal healing by interfering with the IFN/STAT2 death signalling pathway in IECs. These results indicate new directions for suppressing gut inflammation by modulating IL-20-controlled STAT2 signals.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Animals , Mice , Intestinal Mucosa/metabolism , Colitis/metabolism , Interleukins/metabolism , Inflammation/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/genetics , Dextran Sulfate/pharmacology , Mice, Inbred C57BL , STAT2 Transcription Factor/metabolism
11.
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
12.
PLoS One ; 18(11): e0289863, 2023.
Article in English | MEDLINE | ID: mdl-37939052

ABSTRACT

Because pigs are intermediate or amplifying hosts for several zoonotic viruses, the pig-derived PK-15 cell line is an indispensable tool for studying viral pathogenicity and developing treatments, vaccines, and preventive measures to mitigate the risk of disease outbreaks. However, we must consider the possibility of contamination by type I interferons (IFNs), such as IFNα and IFNß, or IFN-inducing substances, such as virus-derived double-stranded RNA or bacterial lipopolysaccharides, in clinical samples, leading to lower rates of viral isolation. In this study, we aimed to generate a PK-15 cell line that can be used to isolate viruses from clinical samples carrying a risk of contamination by IFN-inducing substances. To this end, we depleted the IFN alpha and beta receptor subunit 1 (Ifnar1) gene or signal transducer and activator of transcription 2 (Stat2) gene in PK-15 cells using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 method. Treatment of PK-15 cells lacking Ifnar1 or Stat2 with IFNß or poly (I:C) resulted in no inhibitory effects on viral infection by a lentiviral vector, influenza virus, and Akabane virus. These results demonstrate that PK-15 cells lacking Ifnar1 or Stat2 could represent a valuable and promising tool for viral isolation, vaccine production, and virological investigations.


Subject(s)
Interferon Type I , Viruses , Animals , Swine , STAT2 Transcription Factor/metabolism , Cell Line , Interferon-alpha/metabolism , Interferon Type I/metabolism
13.
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
14.
Birth Defects Res ; 115(16): 1500-1512, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37526179

ABSTRACT

INTRODUCTION: Zika virus (ZIKV) is a human teratogen that causes congenital Zika syndrome (CZS). AXL, TLR3, and STAT2 are proteins involved in the ZIKV's entry into cells (AXL) and host's immune response (TLR3 and STAT2). In this study, we evaluated the role of genetic polymorphisms in these three genes as risk factors to CZS, and highlighted which proteins that interact with them could be important for ZIKV infection and teratogenesis. MATERIALS AND METHODS: We evaluate eighty-eight children exposed to ZIKV during the pregnancy, 40 with CZS and 48 without congenital anomalies. The evaluated polymorphisms in AXL (rs1051008), TLR3 (rs3775291), and STAT2 (rs2066811) were genotyped using TaqMan® Genotyping Assays. A protein-protein interaction network was created in STRING database and analyzed in Cytoscape software. RESULTS: We did not find any statistical significant association among the polymorphisms and the occurrence of CZS. Through the analyses of the network composed by AXL, TLR3, STAT2 and their interactions targets, we found that EGFR and SRC could be important proteins for the ZIKV infection and its teratogenesis. CONCLUSION: In summary, our results demonstrated that the evaluated polymorphisms do not seem to represent risk factors for CZS; however, EGFR and SRC appear to be important proteins that should be investigated in future studies.


Subject(s)
Teratogenesis , Zika Virus Infection , Zika Virus , Pregnancy , Child , Female , Humans , Zika Virus Infection/genetics , Zika Virus/physiology , Axl Receptor Tyrosine Kinase , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Protein Interaction Maps/genetics , ErbB Receptors/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism
15.
Cell Mol Life Sci ; 80(7): 187, 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37347298

ABSTRACT

To understand in detail the transcriptional and functional overlap of IFN-I- and IFN-II-activated responses, we used an integrative RNAseq-ChIPseq approach in Huh7.5 cells and characterized the genome-wide role of pSTAT1, pSTAT2, IRF9 and IRF1 in time-dependent ISG expression. For the first time, our results provide detailed insight in the timely steps of IFNα- and IFNγ-induced transcription, in which pSTAT1- and pSTAT2-containing ISGF3 and GAF-like complexes and IRF1 are recruited to individual or combined ISRE and GAS composite sites in a phosphorylation- and time-dependent manner. Interestingly, composite genes displayed a more heterogeneous expression pattern, as compared to GAS (early) and ISRE genes (late), with the time- and phosphorylation-dependent recruitment of GAF, ISGF3 and IRF1 after IFNα stimulation and GAF and IRF1 after IFNγ. Moreover, functional composite genes shared features of GAS and ISRE genes through transcription factor co-binding to closely located sites, and were able to sustain IFN responsiveness in STAT1-, STAT2-, IRF9-, IRF1- and IRF9/IRF1-mutant Huh7.5 cells compared to Wt cells. Thus, the ISRE + GAS composite site acted as a molecular switch, depending on the timely available components and transcription factor complexes. Consequently, STAT1, STAT2 and IRF9 were identified as functional composite genes that are part of a positive feedback loop controlling long-term IFNα and IFNγ responses. More important, in the absence of any one of the components, the positive feedback regulation of the ISGF3 and GAF components appeared to be preserved. Together, these findings provide further insight in the existence of a novel ISRE + GAS composite-dependent intracellular amplifier circuit prolonging ISG expression and controlling cellular responsiveness to different types of IFNs and subsequent antiviral activity. It also offers an explanation for the existing molecular and functional overlap between IFN-I- and IFN-II-activated ISG expression.


Subject(s)
Interferon Type I , Interferon-alpha , Interferon-alpha/pharmacology , Interferon-alpha/genetics , Interferon-gamma/pharmacology , Interferon-gamma/metabolism , Gene Expression Regulation , Antiviral Agents , Interferon Type I/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism
16.
J Biol Chem ; 299(5): 104703, 2023 05.
Article in English | MEDLINE | ID: mdl-37059181

ABSTRACT

The conversion of signal transducer and activator of transcription (STAT) proteins from latent to active transcription factors is central to cytokine signaling. Triggered by their signal-induced tyrosine phosphorylation, it is the assembly of a range of cytokine-specific STAT homo- and heterodimers that marks a key step in the transition of hitherto latent proteins to transcription activators. In contrast, the constitutive self-assembly of latent STATs and how it relates to the functioning of activated STATs is understood less well. To provide a more complete picture, we developed a co-localization-based assay and tested all 28 possible combinations of the seven unphosphorylated STAT (U-STAT) proteins in living cells. We identified five U-STAT homodimers-STAT1, STAT3, STAT4, STAT5A, and STAT5B-and two heterodimers-STAT1:STAT2 and STAT5A:STAT5B-and performed semi-quantitative assessments of the forces and characterizations of binding interfaces that support them. One STAT protein-STAT6-was found to be monomeric. This comprehensive analysis of latent STAT self-assembly lays bare considerable structural and functional diversity in the ways that link STAT dimerization before and after activation.


Subject(s)
Gene Expression Regulation , STAT Transcription Factors , Trans-Activators , Cytokines/metabolism , Phosphorylation , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Trans-Activators/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Protein Multimerization
17.
Proc Natl Acad Sci U S A ; 120(16): e2216953120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37036972

ABSTRACT

In cancer cells, endogenous or therapy-induced DNA damage leads to the abnormal presence of DNA in the cytoplasm, which triggers the activation of cGAS (cyclic GMP-AMP synthase) and STING (stimulator of interferon genes). STAT2 suppresses the cGAMP-induced expression of IRF3-dependent genes by binding to STING, blocking its intracellular trafficking, which is essential for the full response to STING activation. STAT2 reshapes STING signaling by inhibiting the induction of IRF3-dependent, but not NF-κB-dependent genes. This noncanonical activity of STAT2 is regulated independently of its tyrosine phosphorylation but does depend on the phosphorylation of threonine 404, which promotes the formation of a STAT2:STING complex that keeps STING bound to the endoplasmic reticulum (ER) and increases resistance to DNA damage. We conclude that STAT2 is a key negative intracellular regulator of STING, a function that is quite distinct from its function as a transcription factor.


Subject(s)
Membrane Proteins , Nucleotidyltransferases , Protein Serine-Threonine Kinases , STAT2 Transcription Factor , DNA/metabolism , DNA Damage , Nucleotidyltransferases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , STAT2 Transcription Factor/metabolism , Membrane Proteins/metabolism
18.
Virology ; 583: 1-13, 2023 06.
Article in English | MEDLINE | ID: mdl-37060797

ABSTRACT

Type I interferon (IFN-I) evasion by Dengue virus (DENV) is key in DENV pathogenesis. The non-structural protein 5 (NS5) antagonizes IFN-I response through the degradation of the signal transducer and activator of transcription 2 (STAT2). We developed a K562 cell-based platform, for high throughput screening of compounds potentially counteracting the NS5-mediated antagonism of IFN-I signaling. Upon a screening with a library of 1220 approved drugs, 3 compounds previously linked to DENV inhibition (Apigenin, Chrysin, and Luteolin) were identified. Luteolin and Apigenin determined a significant inhibition of DENV2 replication in Huh7 cells and the restoration of STAT2 phosphorylation in both cell systems. Apigenin and Luteolin were able to stimulate STAT2 even in the absence of infection. Despite the "promiscuous" and "pan-assay-interfering" nature of Luteolin, Apigenin promotes STAT2 Tyr 689 phosphorylation and activation, highlighting the importance of screening for compounds able to interact with host factors, to counteract viral proteins capable of dampening innate immune responses.


Subject(s)
Dengue Virus , Apigenin/pharmacology , Dengue Virus/physiology , Luteolin/pharmacology , Signal Transduction , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Humans
19.
J Virol ; 97(3): e0194222, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36856422

ABSTRACT

African swine fever virus (ASFV) is a large DNA virus that causes African swine fever (ASF), an acute and hemorrhagic disease in pigs with lethality rates of up to 100%. To date, how ASFV efficiently suppress the innate immune response remains enigmatic. In this study, we identified ASFV cysteine protease pS273R as an antagonist of type I interferon (IFN). Overexpression of pS273R inhibited JAK-STAT signaling triggered by type I IFNs. Mechanistically, pS273R interacted with STAT2 and recruited the E3 ubiquitin ligase DCST1, resulting in K48-linked polyubiquitination at K55 of STAT2 and subsequent proteasome-dependent degradation of STAT2. Furthermore, such a function of pS273R in JAK-STAT signaling is not dependent on its protease activity. These findings suggest that ASFV pS273R is important to evade host innate immunity. IMPORTANCE ASF is an acute disease in domestic pigs caused by infection with ASFV. ASF has become a global threat with devastating economic and ecological consequences. To date, there are no commercially available, safe, and efficacious vaccines to prevent ASFV infection. ASFV has evolved a series of strategies to evade host immune responses, facilitating its replication and transmission. Therefore, understanding the immune evasion mechanism of ASFV is helpful for the development of prevention and control measures for ASF. Here, we identified ASFV cysteine protease pS273R as an antagonist of type I IFNs. ASFV pS273R interacted with STAT2 and mediated degradation of STAT2, a transcription factor downstream of type I IFNs that is responsible for induction of various IFN-stimulated genes. pS273R recruited the E3 ubiquitin ligase DCST1 to enhance K48-linked polyubiquitination of STAT2 at K55 in a manner independent of its protease activity. These findings suggest that pS273R is important for ASFV to escape host innate immunity, which sheds new light on the mechanisms of ASFV immune evasion.


Subject(s)
African Swine Fever Virus , African Swine Fever , Cysteine Proteases , Interferon Type I , Animals , Cysteine Proteases/genetics , Cysteine Proteases/metabolism , Immunity, Innate/genetics , Interferon Type I/metabolism , Sus scrofa , Swine , Ubiquitin-Protein Ligases/metabolism , STAT2 Transcription Factor/metabolism , Signal Transduction
20.
J Gene Med ; 25(8): e3506, 2023 08.
Article in English | MEDLINE | ID: mdl-36994700

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) play a critical role in regulating various human diseases including cancer. In colorectal cancer (CRC), there are still some undervalued lncRNAs with potential functions and mechanisms that need to be clarified. The present study aimed to investigate the role of linc02231 in the progression of CRC. METHODS: The proliferation of CRC cells was evaluated using Cell Counting Kit-8, colony formation, and 5-ethynyl-2'-deoxyuridine (EdU) assays. Cell migration was examined through wound healing and Transwell analyses. The impact of linc02231 on angiogenesis was determined through a tube formation assay. Western blotting was used to detect the expression of specific proteins. A mouse xenograft model is established to observe the effect of linc02231 on the in vivo growth of CRC cells. Target genes of linc02231 are screened using high-throughput sequencing. The transcriptional activity of STAT2 on linc02231 and the binding activity between linc02231/miR-939-5p/hnRNPA1 were analyzed by a luciferase assay. RESULTS: Based on public databases and comprehensive bioinformatics analysis, we found that lncRNA linc02231 was upregulated in CRC tumor tissues, which is consistent with our clinical results. linc02231 promoted the proliferation and migration of CRC cells in vitro and their tumorigenicity in vivo. Furthermore, linc02231 promotes the angiogenic ability of human umbilical vein endothelial cells. Mechanistically, the transcription factor STAT2 binds to the promoter region of linc02231 and activates its transcription. linc02231 also competes with miR-939-5p for binding to the pro-oncogenic target gene hnRNPA1, preventing its degradation. hnRNPA1 prevents the maturation of angiopoietin-like protein 4 (ANGPTL4) messenger RNA, leading to impaired tumor angiogenesis and increased metastasis of CRC. CONCLUSIONS: The expression of linc02231, which is induced by STAT2, has been found to enhance the proliferation, metastasis, and angiogenesis of CRC by binding to miR-939-5p and increasing the expression of hnNRPA1 at the same time as suppressing ANGPTL4. These findings suggest that linc02231 could serve as a potential biomarker and therapeutic target for CRC.


Subject(s)
Colorectal Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Angiopoietin-Like Protein 4/genetics , Angiopoietin-Like Protein 4/metabolism , Cell Line, Tumor , RNA, Long Noncoding/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/pathology , Cell Proliferation/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic
SELECTION OF CITATIONS
SEARCH DETAIL
...