Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 322
Filter
1.
Elife ; 132024 May 15.
Article in English | MEDLINE | ID: mdl-38747717

ABSTRACT

Invertebrates use the endoribonuclease Dicer to cleave viral dsRNA during antiviral defense, while vertebrates use RIG-I-like Receptors (RLRs), which bind viral dsRNA to trigger an interferon response. While some invertebrate Dicers act alone during antiviral defense, Caenorhabditis elegans Dicer acts in a complex with a dsRNA binding protein called RDE-4, and an RLR ortholog called DRH-1. We used biochemical and structural techniques to provide mechanistic insight into how these proteins function together. We found RDE-4 is important for ATP-independent and ATP-dependent cleavage reactions, while helicase domains of both DCR-1 and DRH-1 contribute to ATP-dependent cleavage. DRH-1 plays the dominant role in ATP hydrolysis, and like mammalian RLRs, has an N-terminal domain that functions in autoinhibition. A cryo-EM structure indicates DRH-1 interacts with DCR-1's helicase domain, suggesting this interaction relieves autoinhibition. Our study unravels the mechanistic basis of the collaboration between two helicases from typically distinct innate immune defense pathways.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , RNA, Double-Stranded , Ribonuclease III , Animals , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , RNA, Double-Stranded/metabolism , Ribonuclease III/metabolism , Ribonuclease III/chemistry , Ribonuclease III/genetics , Cryoelectron Microscopy , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , RNA Helicases/chemistry , Protein Binding , Adenosine Triphosphate/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/chemistry
2.
Biochem Biophys Res Commun ; 712-713: 149915, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38663038

ABSTRACT

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


Subject(s)
Escherichia coli , Levivirus , RNA, Double-Stranded , Escherichia coli/virology , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Double-Stranded/metabolism , Humans , Levivirus/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Immunity, Innate , Interferon-beta/metabolism , Interferon-beta/genetics , NF-kappa B/metabolism , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Signal Transduction , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Receptors, Immunologic , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics
3.
J Transl Med ; 22(1): 395, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685028

ABSTRACT

BACKGROUND: Current cancer therapies often fall short in addressing the complexities of malignancies, underscoring the urgent need for innovative treatment strategies. RNA interference technology, which specifically suppresses gene expression, offers a promising new approach in the fight against tumors. Recent studies have identified a novel immunostimulatory small-interfering RNA (siRNA) with a unique sequence (sense strand, 5'-C; antisense strand, 3'-GGG) capable of activating the RIG-I/IRF3 signaling pathway. This activation induces the release of type I and III interferons, leading to an effective antiviral immune response. However, this class of immunostimulatory siRNA has not yet been explored in cancer therapy. METHODS: IsiBCL-2, an innovative immunostimulatory siRNA designed to suppress the levels of B-cell lymphoma 2 (BCL-2), contains a distinctive motif (sense strand, 5'-C; antisense strand, 3'-GGG). Glioblastoma cells were subjected to 100 nM isiBCL-2 treatment in vitro for 48 h. Morphological changes, cell viability (CCK-8 assay), proliferation (colony formation assay), migration/invasion (scratch test and Transwell assay), apoptosis rate, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) were evaluated. Western blotting and immunofluorescence analyses were performed to assess RIG-I and MHC-I molecule levels, and ELISA was utilized to measure the levels of cytokines (IFN-ß and CXCL10). In vivo heterogeneous tumor models were established, and the anti-tumor effect of isiBCL-2 was confirmed through intratumoral injection. RESULTS: IsiBCL-2 exhibited significant inhibitory effects on glioblastoma cell growth and induced apoptosis. BCL-2 mRNA levels were significantly decreased by 67.52%. IsiBCL-2 treatment resulted in an apoptotic rate of approximately 51.96%, accompanied by a 71.76% reduction in MMP and a 41.87% increase in ROS accumulation. Western blotting and immunofluorescence analyses demonstrated increased levels of RIG-I, MAVS, and MHC-I following isiBCL-2 treatment. ELISA tests indicated a significant increase in IFN-ß and CXCL10 levels. In vivo studies using nude mice confirmed that isiBCL-2 effectively impeded the growth and progression of glioblastoma tumors. CONCLUSIONS: This study introduces an innovative method to induce innate signaling by incorporating an immunostimulatory sequence (sense strand, 5'-C; antisense strand, 3'-GGG) into siRNA, resulting in the formation of RNA dimers through Hoogsteen base-pairing. This activation triggers the RIG-I signaling pathway in tumor cells, causing further damage and inducing a potent immune response. This inventive design and application of immunostimulatory siRNA offer a novel perspective on tumor immunotherapy, holding significant implications for the field.


Subject(s)
Apoptosis , Glioma , RNA, Small Interfering , Humans , Animals , Cell Line, Tumor , Glioma/therapy , Glioma/pathology , Glioma/genetics , RNA, Small Interfering/metabolism , Mice, Nude , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Cell Proliferation , Cell Movement , Xenograft Model Antitumor Assays , Mice , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Reactive Oxygen Species/metabolism , Neoplasm Invasiveness , Cell Survival
4.
J Agric Food Chem ; 72(17): 9782-9794, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38597360

ABSTRACT

Uncontrolled inflammation contributes significantly to the mortality in acute respiratory infections. Our previous research has demonstrated that maize bran feruloylated oligosaccharides (FOs) possess notable anti-inflammatory properties linked to the NF-kB pathway regulation. In this study, we clarified that the oral administration of FOs moderately inhibited H1N1 virus infection and reduced lung inflammation in influenza-infected mice by decreasing a wide spectrum of cytokines (IFN-α, IFN-ß, IL-6, IL-10, and IL-23) in the lungs. The mechanism involves FOs suppressing the transduction of the RIG-I/MAVS/TRAF3 signaling pathway, subsequently lowering the expression of NF-κB. In silico analysis suggests that FOs have a greater binding affinity for the RIG-I/MAVS signaling complex. This indicates that FOs have potential as promising targets for immune modulation. Moreover, in MAVS knockout mice, we confirmed that the anti-inflammatory function of FOs against influenza depends on MAVS. Comprehensive analysis using 16S rRNA gene sequencing and metabolite profiling techniques showed that FOs have the potential to restore immunity by modulating the gut microbiota. In conclusion, our study demonstrates that FOs are effective anti-inflammatory phytochemicals in inhibiting lung inflammation caused by influenza. This suggests that FOs could serve as a potential nutritional strategy for preventing the H1N1 virus infection and associated lung inflammation.


Subject(s)
DEAD Box Protein 58 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Mice, Knockout , Oligosaccharides , Orthomyxoviridae Infections , Signal Transduction , TNF Receptor-Associated Factor 3 , Animals , Mice , Oligosaccharides/administration & dosage , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Humans , Signal Transduction/drug effects , Signal Transduction/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 3/immunology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/immunology , Pneumonia/immunology , Pneumonia/prevention & control , Pneumonia/metabolism , Pneumonia/virology , Mice, Inbred C57BL , Lung/immunology , Lung/metabolism , Lung/drug effects , Lung/virology , Cytokines/metabolism , Cytokines/immunology , Cytokines/genetics , Female , NF-kappa B/immunology , NF-kappa B/genetics , NF-kappa B/metabolism , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology
5.
DNA Cell Biol ; 43(4): 197-205, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38466944

ABSTRACT

Previous studies have shown that interferon gene-stimulating protein (STING) is essential for IFN-γ-inducible protein 16 (IFI16) as the DNA sensor and RNA sensor to induce transcription of type I interferon (IFN-I) and is essential for IFI16 to synergize with DNA sensor GMP-AMP (cGAMP) synthase (cGAS) in induction of IFN-I transcription. While other and our previous studies have shown that IFI16 enhanced retinoic acid-inducible gene I (RIG-I)-, which was an RNA sensor, and mitochondrial antiviral signaling (MAVS)-, which was the adaptor protein of RIG-I, induced production of IFN-I, so we wonder whether IFI16 regulates the signal pathway of RNA-RIG-I-MAVS-IFN-I in a STING-dependent manner. We used HEK 293T cells, which did not express endogenous STING and were unable to mount an innate immune response upon DNA transfection and found that IFI16 could enhance RIG-I- and MAVS-mediated induction of IFN-I in a STING-independent way. Furthermore, we found that upregulation of the expression of NF-kappa-B essential modulator (NEMO) by IFI16 was not the mechanism that IFI16 regulated the induction of IFN-I. In conclusion, we found that IFI16 regulated the signal pathway of RNA-RIG-I-MAVS-IFN-I in a STING-independent manner.


Subject(s)
Immunity, Innate , Interferon Type I , DEAD Box Protein 58/genetics , DNA , Interferon Type I/genetics , Receptors, Immunologic/genetics , RNA , Humans
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167092, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38382623

ABSTRACT

The production of type I interferon (IFN) is precisely modulated by host to protect against viral infection efficiently without obvious immune disorders. Elucidating the tight control towards type I IFN production would be helpful to get insight into natural immunity and inflammatory diseases. As yet, however, the mechanisms that regulate IFN-ß production, especially the epigenetic regulatory mechanisms, remain poorly explored. This study elucidated the potential function of Peptidylarginine deiminases (PADIs)-mediated citrullination in innate immunity. We identified PADI4, a PADIs family member that can act as an epigenetic coactivator, could repress IFN-ß production upon RNA virus infection. Detailed experiments showed that PADI4 deficiency increased IFN-ß production and promoted antiviral immune activities against RNA viruses. Mechanistically, the increased PADI4 following viral infection translocated to nucleus and recruited HDAC1 upon binding to Ifnb1 promoter, which then led to the deacetylation of histone H3 and histone H4 for repressing Ifnb1 transcription. Taken together, we identify a novel non-classical role for PADI4 in the regulation of IFN-ß production, suggesting its potential as treatment target in inflammatory or autoimmune diseases.


Subject(s)
Histones , Virus Diseases , DEAD Box Protein 58/genetics , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histones/metabolism , Immunity, Innate , Protein-Arginine Deiminases/genetics , Protein-Arginine Deiminases/metabolism , Receptors, Immunologic/metabolism
7.
Cancer Res Commun ; 4(2): 540-555, 2024 02 26.
Article in English | MEDLINE | ID: mdl-38358346

ABSTRACT

Type I IFN signaling is a crucial component of antiviral immunity that has been linked to promoting the efficacy of some chemotherapeutic drugs. We developed a reporter system in HCT116 cells that detects activation of the endogenous IFI27 locus, an IFN target gene. We screened a library of annotated compounds in these cells and discovered Aurora kinase inhibitors (AURKi) as strong hits. Type I IFN signaling was found to be the most enriched gene signature after AURKi treatment in HCT116, and this signature was also strongly enriched in other colorectal cancer cell lines. The ability of AURKi to activate IFN in HCT116 was dependent on MAVS and RIG-I, but independent of STING, whose signaling is deficient in these cells. MAVS dependence was recapitulated in other colorectal cancer lines with STING pathway deficiency, whereas in cells with intact STING signaling, the STING pathway was required for IFN induction by AURKi. AURKis were found to induce expression of endogenous retroviruses (ERV). These ERVs were distinct from those induced by the DNA methyltransferase inhibitors (DNMTi), which can induce IFN signaling via ERV induction, suggesting a novel mechanism of action. The antitumor effect of alisertib in mice was accompanied by an induction of IFN expression in HCT116 or CT26 tumors. CT26 tumor growth inhibition by alisertib was absent in NSG mice versus wildtype (WT) mice, and tumors from WT mice with alisertib treatment showed increased in CD8+ T-cell infiltration, suggesting that antitumor efficacy of AURKi depends, at least in part, on an intact immune response. SIGNIFICANCE: Some cancers deactivate STING signaling to avoid consequences of DNA damage from aberrant cell division. The surprising activation of MAVS/RIG-I signaling by AURKi might represent a vulnerability in STING signaling deficient cancers.


Subject(s)
Colorectal Neoplasms , Interferon Type I , Animals , Mice , Retroelements , Interferon Lambda , Aurora Kinases/metabolism , Interferon Type I/metabolism , DEAD Box Protein 58/genetics , Receptors, Immunologic
8.
Sci Rep ; 14(1): 1455, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38228690

ABSTRACT

Influenza virus infection alters the promoter DNA methylation of key immune response-related genes, including type-1 interferons and proinflammatory cytokines. However, less is known about the effect of the influenza vaccine on the epigenome. We utilized a targeted DNA methylation approach to study the longitudinal effects (day 0 pre-vaccination and day 28 post-vaccination) on influenza vaccination responses in peripheral blood mononuclear cells. We found that baseline, pre-vaccination methylation profiles are associated with pre-existing, protective serological immunity. Additionally, we identified 481 sites that were differentially methylated between baseline and day 28 post-vaccination. These were enriched for genes involved in the regulation of the RIG-I signaling pathway, an important regulator of viral responses. Our results suggest that DNA methylation changes to components of the RIG-I pathway are associated with vaccine effectiveness. Therefore, immunization strategies that target this pathway may improve serological responses to influenza vaccination.


Subject(s)
Influenza Vaccines , Influenza, Human , Humans , DNA Methylation , Influenza, Human/prevention & control , Leukocytes, Mononuclear , Vaccination/methods , DEAD Box Protein 58/genetics , Signal Transduction , Antibodies, Viral
9.
Nat Commun ; 15(1): 780, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38278841

ABSTRACT

The Retinoic acid-Inducible Gene I (RIG-I) like receptors (RLRs) are the major viral RNA sensors essential for the initiation of antiviral immune responses. RLRs are subjected to stringent transcriptional and posttranslational regulations, of which ubiquitination is one of the most important. However, the role of ubiquitination in RLR transcription is unknown. Here, we screen 375 definite ubiquitin ligase knockout cell lines and identify Ubiquitin Protein Ligase E3 Component N-Recognin 5 (UBR5) as a positive regulator of RLR transcription. UBR5 deficiency reduces antiviral immune responses to RNA viruses, while increases viral replication in primary cells and mice. Ubr5 knockout mice are more susceptible to lethal RNA virus infection than wild type littermates. Mechanistically, UBR5 mediates the Lysine 63-linked ubiquitination of Tripartite Motif Protein 28 (TRIM28), an epigenetic repressor of RLRs. This modification prevents intramolecular SUMOylation of TRIM28, thus disengages the TRIM28-imposed brake on RLR transcription. In sum, UBR5 enables rapid upregulation of RLR expression to boost antiviral immune responses by ubiquitinating and de-SUMOylating TRIM28.


Subject(s)
RNA Viruses , Mice , Animals , Ubiquitination , Cell Line , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Immunity, Innate , Ubiquitin-Protein Ligases/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism
10.
Nucleic Acids Res ; 52(1): 355-369, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38015453

ABSTRACT

The RIG-I family helicases, comprising RIG-I, MDA5 and LGP2, are cytoplasmic RNA sensors that trigger an antiviral immune response by specifically recognizing foreign RNAs. While LGP2 lacks the signaling domain necessary for immune activation, it plays a vital role in regulating the RIG-I/MDA5 signaling pathway. In this study, we investigate the mechanisms underlying this regulation by examining the oligomeric state, RNA binding specificity, and translocation activity of human LGP2 and the impact of ATPase activity. We show that LGP2, like RIG-I, prefers binding blunt-ended double-stranded (ds) RNAs over internal dsRNA regions or RNA overhangs and associates with blunt-ends faster than with overhangs. Unlike RIG-I, a 5'-triphosphate (5'ppp), Cap0, or Cap1 RNA-end does not influence LGP2's RNA binding affinity. LGP2 hydrolyzes ATP in the presence of RNA but at a 5-10 fold slower rate than RIG-I. Nevertheless, LGP2 uses its ATPase activity to translocate and displace biotin-streptavidin interactions. This activity is significantly hindered by a methylated RNA patch, particularly on the 3'-strand, suggesting a 3'-strand tracking mechanism like RIG-I. The preference of LGP2 for blunt-end RNA binding, its insensitivity to Cap0/Cap1 modification, and its translocation/protein displacement ability have substantial implications for how LGP2 regulates the RNA sensing process by MDA5/RIG-I.


Subject(s)
DEAD-box RNA Helicases , RNA Helicases , Humans , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , DEAD-box RNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Protein Binding/physiology , Receptors, Immunologic/genetics , RNA Helicases/metabolism , RNA, Double-Stranded , RNA, Viral/metabolism
11.
Cell Death Differ ; 31(1): 28-39, 2024 01.
Article in English | MEDLINE | ID: mdl-38001254

ABSTRACT

The ability of cells to mount an interferon response to virus infections depends on intracellular nucleic acid sensing pattern recognition receptors (PRRs). RIG-I is an intracellular PRR that binds short double-stranded viral RNAs to trigger MAVS-dependent signalling. The RIG-I/MAVS signalling complex requires the coordinated activity of multiple kinases and E3 ubiquitin ligases to activate the transcription factors that drive type I and type III interferon production from infected cells. The linear ubiquitin chain assembly complex (LUBAC) regulates the activity of multiple receptor signalling pathways in both ligase-dependent and -independent ways. Here, we show that the three proteins that constitute LUBAC have separate functions in regulating RIG-I signalling. Both HOIP, the E3 ligase capable of generating M1-ubiquitin chains, and LUBAC accessory protein HOIL-1 are required for viral RNA sensing by RIG-I. The third LUBAC component, SHARPIN, is not required for RIG-I signalling. These data cement the role of LUBAC as a positive regulator of RIG-I signalling and as an important component of antiviral innate immune responses.


Subject(s)
RNA Viruses , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Signal Transduction , DEAD Box Protein 58/genetics , RNA Viruses/metabolism
12.
Virology ; 589: 109942, 2024 01.
Article in English | MEDLINE | ID: mdl-38048647

ABSTRACT

Hantaan virus (HTNV) is responsible for hemorrhagic fever with renal syndrome (HFRS), primarily due to its ability to inhibit host innate immune responses, such as type I interferon (IFN-I). In this study, we conducted a transcriptome analysis to identify host factors regulated by HTNV nucleocapsid protein (NP) and glycoprotein. Our findings demonstrate that NP and Gc proteins inhibit host IFN-I production by manipulating the retinoic acid-induced gene I (RIG-I)-like receptor (RLR) pathways. Further analysis reveals that HTNV NP and Gc proteins target upstream molecules of MAVS, such as RIG-I and MDA-5, with Gc exhibiting stronger inhibition of IFN-I responses than NP. Mechanistically, NP and Gc proteins interact with tripartite motif protein 25 (TRIM25) to competitively inhibit its interaction with RIG-I/MDA5, suppressing RLR signaling pathways. Our study unveils a cross-talk between HTNV NP/Gc proteins and host immune response, providing valuable insights into the pathogenic mechanism of HTNV.


Subject(s)
Hantaan virus , Interferon Type I , Interferon Type I/metabolism , Hantaan virus/genetics , Hantaan virus/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Signal Transduction , Immunity, Innate , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism
13.
Int J Biol Macromol ; 253(Pt 7): 127351, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37839600

ABSTRACT

Bovine viral diarrhea virus (BVDV) is one of the most important pathogens of cattle, causing numerous economic losses to the cattle industry. To date, many potential mechanisms of BVDV evading or subverting innate immunity are still unknown. In this study, an lnc-CYLD/miR-2383/CYLD axis involved in BVDV-host interactions was screened from RNA-seq-based co-expression networks analysis of long noncoding RNAs, microRNAs and mRNAs in BVDV-infected bovine cells, and underlying mechanisms of lnc-CYLD/miR-2383/CYLD axis regulating BVDV replication were explored. Results showed that BVDV-induced up-regulation of the lnc-CYLD competed for binding to the miR-2383, and then promoted CYLD expression, thereby inhibiting RIG-I-mediated type-I interferon (IFN) production, which was subsequently confirmed by treatment with lnc-CYLD overexpression and miR-2383 inhibitor. However, miR-2383 transfection and small interfering RNA-mediated lnc-CYLD knockdown inhibited CYLD expression and enhanced RIG-I-mediated type-I IFN production, inhibiting BVDV replication. In addition, interaction relationship between lnc-CYLD and miR-2383, and colocalization relationship of lnc-CYLD, miR-2383 and CYLD were confirmed by dual-luciferase assay and in situ hybridization assay. Conclusively, up-regulation of the lnc-CYLD as a competing endogenous RNA binds to the miR-2383 to reduce inhibitory effect of the miR-2383 on the CYLD expression, playing an important role in counteracting type-I IFN-dependent antiviral immunity to facilitate BVDV replication.


Subject(s)
Diarrhea Virus 1, Bovine Viral , Diarrhea Viruses, Bovine Viral , Interferon Type I , MicroRNAs , RNA, Long Noncoding , Animals , Cattle , DEAD Box Protein 58/genetics , RNA, Long Noncoding/metabolism , Virus Replication/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Diarrhea Viruses, Bovine Viral/genetics , Interferon Type I/genetics , Diarrhea , Diarrhea Virus 1, Bovine Viral/genetics
14.
EMBO Rep ; 24(12): e57137, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37870297

ABSTRACT

Most SARS-CoV-2 proteins are translated from subgenomic RNAs (sgRNAs). While the majority of these sgRNAs are monocistronic, some viral mRNAs encode more than one protein. One example is the ORF3a sgRNA that also encodes ORF3c, an enigmatic 41-amino-acid peptide. Here, we show that ORF3c is expressed in SARS-CoV-2-infected cells and suppresses RIG-I- and MDA5-mediated IFN-ß induction. ORF3c interacts with the signaling adaptor MAVS, induces its C-terminal cleavage, and inhibits the interaction of RIG-I with MAVS. The immunosuppressive activity of ORF3c is conserved among members of the subgenus sarbecovirus, including SARS-CoV and coronaviruses isolated from bats. Notably, however, the SARS-CoV-2 delta and kappa variants harbor premature stop codons in ORF3c, demonstrating that this reading frame is not essential for efficient viral replication in vivo and is likely compensated by other viral proteins. In agreement with this, disruption of ORF3c does not significantly affect SARS-CoV-2 replication in CaCo-2, CaLu-3, or Rhinolophus alcyone cells. In summary, we here identify ORF3c as an immune evasion factor of SARS-CoV-2 that suppresses innate sensing in infected cells.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Caco-2 Cells , COVID-19/genetics , Signal Transduction , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Immunity, Innate/genetics
15.
Nucleic Acids Res ; 51(21): 11893-11910, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37831086

ABSTRACT

RIG-I is a cytosolic receptor of viral RNA essential for the immune response to numerous RNA viruses. Accordingly, RIG-I must sensitively detect viral RNA yet tolerate abundant self-RNA species. The basic binding cleft and an aromatic amino acid of the RIG-I C-terminal domain(CTD) mediate high-affinity recognition of 5'triphosphorylated and 5'base-paired RNA(dsRNA). Here, we found that, while 5'unmodified hydroxyl(OH)-dsRNA demonstrated residual activation potential, 5'-monophosphate(5'p)-termini, present on most cellular RNAs, prevented RIG-I activation. Determination of CTD/dsRNA co-crystal structures and mutant activation studies revealed that the evolutionarily conserved I875 within the CTD sterically inhibits 5'p-dsRNA binding. RIG-I(I875A) was activated by both synthetic 5'p-dsRNA and endogenous long dsRNA within the polyA-rich fraction of total cellular RNA. RIG-I(I875A) specifically interacted with long, polyA-bearing, mitochondrial(mt) RNA, and depletion of mtRNA from total RNA abolished its activation. Altogether, our study demonstrates that avoidance of 5'p-RNA recognition is crucial to prevent mtRNA-triggered RIG-I-mediated autoinflammation.


Subject(s)
DEAD Box Protein 58 , Isoleucine , Receptors, Immunologic , DEAD Box Protein 58/chemistry , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Immune Tolerance , Isoleucine/genetics , RNA, Double-Stranded/genetics , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
16.
Commun Biol ; 6(1): 818, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37543704

ABSTRACT

Radiotherapy (RT) plus immunotherapy is a promising modality; however, the therapeutic effects are insufficient, and the molecular mechanism requires clarification to further develop combination therapies. Here, we found that the RNA virus sensor pathway dominantly regulates the cellular immune response in NSCLC and ESCC cell lines. Notably, transposable elements (TEs), especially long terminal repeats (LTRs), functioned as key ligands for the RNA virus sensor RIG-I, and the mTOR-LTR-RIG-I axis induced the cellular immune response and dendritic cell and macrophage infiltration after irradiation. Moreover, RIG-I-dependent immune activation was observed in ESCC patient tissue. scRNA sequencing and spatial transcriptome analysis revealed that radiotherapy induced the expression of LTRs, and the RNA virus sensor pathway in immune and cancer cells; this pathway was also found to mediate tumour conversion to an immunological hot state. Here, we report the upstream and ligand of the RNA virus sensor pathway functions in irradiated cancer tissues.


Subject(s)
DNA Transposable Elements , Macrophages , Humans , Cell Line , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Macrophages/metabolism
17.
Acta Biochim Biophys Sin (Shanghai) ; 55(10): 1582-1591, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37580950

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is a cytosolic viral RNA receptor. Upon viral infection, the protein recognizes and then recruits adapter protein mitochondrial antiviral signaling (MAVS) protein, initiating the production of interferons and proinflammatory cytokines to establish an antiviral state. In the present study, we identify zinc finger protein 205 (ZNF205) which associates with RIG-I and promotes the Sendai virus (SeV)-induced antiviral innate immune response. Overexpression of ZNF205 facilitates interferon-beta (IFN-ß) introduction, whereas ZNF205 deficiency restricts its introduction. Mechanistically, the C-terminal zinc finger domain of ZNF205 interacts with the N-terminal tandem caspase recruitment domains (CARDs) of RIG-I; this interaction markedly promotes K63 ubiquitin-linked polyubiquitination of RIG-I, which is crucial for RIG-I activation. Thus, our results demonstrate that ZNF205 is a positive regulator of the RIG-I-mediated innate antiviral immune signaling pathway.


Subject(s)
Immunity, Innate , Signal Transduction , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Immunity, Innate/genetics , Interferon-beta/genetics , Interferons/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Sendai virus/immunology
18.
Cell Mol Immunol ; 20(10): 1186-1202, 2023 10.
Article in English | MEDLINE | ID: mdl-37582970

ABSTRACT

The adaptor molecule MAVS forms prion-like aggregates to govern the RIG-I-like receptor (RLR) signaling cascade. Lys63 (K63)-linked polyubiquitination is critical for MAVS aggregation, yet the underlying mechanism and the corresponding E3 ligases and deubiquitinating enzymes (DUBs) remain elusive. Here, we found that the K63-linked polyubiquitin chains loaded on MAVS can be directly recognized by RIG-I to initiate RIG-I-mediated MAVS aggregation with the prerequisite of the CARDRIG-I-CARDMAVS interaction. Interestingly, many K63-linked polyubiquitin chains attach to MAVS via an unanchored linkage. We identified Ube2N as a major ubiquitin-conjugating enzyme for MAVS and revealed that Ube2N cooperates with the E3 ligase Riplet and TRIM31 to promote the unanchored K63-linked polyubiquitination of MAVS. In addition, we identified USP10 as a direct DUB that removes unanchored K63-linked polyubiquitin chains from MAVS. Consistently, USP10 attenuates RIG-I-mediated MAVS aggregation and the production of type I interferon. Mice with a deficiency in USP10 show more potent resistance to RNA virus infection. Our work proposes a previously unknown mechanism for the activation of the RLR signaling cascade triggered by MAVS-attached unanchored K63-linked polyubiquitin chains and establishes the DUB USP10 and the E2:E3 pair Ube2N-Riplet/TRIM31 as a specific regulatory system for the unanchored K63-linked ubiquitination and aggregation of MAVS upon viral infection.


Subject(s)
Immunity, Innate , Polyubiquitin , Animals , Mice , DEAD Box Protein 58/genetics , Polyubiquitin/metabolism , Immunity, Innate/genetics , Signal Transduction/genetics , Ubiquitination , Ubiquitin-Protein Ligases/metabolism
19.
J Gen Virol ; 104(8)2023 08.
Article in English | MEDLINE | ID: mdl-37584553

ABSTRACT

Type I interferons (IFNs) are the major host defence against viral infection and are induced following activation of cell surface or intracellular pattern recognition receptors, including retinoic-acid-inducible gene I (RIG-I)-like receptors (RLRs). All cellular processes are shaped by the microenvironment and one important factor is the local oxygen tension. The majority of published studies on IFN signalling are conducted under laboratory conditions of 18% oxygen (O2), that do not reflect the oxygen levels in most organs (1-5 % O2). We studied the effect of low oxygen on IFN induction and signalling in induced Pluripotent Stem Cell (iPSC)-derived macrophages as a model for tissue-resident macrophages and assessed the consequence for Zika virus (ZIKV) infection. Hypoxic conditions dampened the expression of interferon-stimulated genes (ISGs) following RLR stimulation or IFN treatment at early time points. RNA-sequencing and bio-informatic analysis uncovered several pathways including changes in transcription factor availability, the presence of HIF binding sites in promoter regions, and CpG content that may contribute to the reduced ISG expression. Hypoxic conditions increased the abundance of ZIKV RNA highlighting the importance of understanding how low oxygen conditions in the local microenvironment affect pathogen sensing and host defences.


Subject(s)
Induced Pluripotent Stem Cells , Interferon Type I , Zika Virus Infection , Zika Virus , Humans , Zika Virus/genetics , Induced Pluripotent Stem Cells/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Receptors, Immunologic , Interferon Type I/metabolism , Macrophages/metabolism , Immunity, Innate , RNA , Hypoxia , Oxygen/pharmacology
20.
Nucleic Acids Res ; 51(17): 9356-9368, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37486777

ABSTRACT

RIG-I (retinoic acid inducible gene-I) can sense subtle differences between endogenous and viral RNA in the cytoplasm, triggering an anti-viral immune response through induction of type I interferons (IFN) and other inflammatory mediators. Multiple crystal and cryo-EM structures of RIG-I suggested a mechanism in which the C-terminal domain (CTD) is responsible for the recognition of viral RNA with a 5'-triphoshate modification, while the CARD domains serve as a trigger for downstream signaling, leading to the induction of type I IFN. However, to date contradicting conclusions have been reached around the role of ATP in the mechanism of the CARD domains ejection from RIG-I's autoinhibited state. Here we present an application of NMR spectroscopy to investigate changes induced by the binding of 5'-triphosphate and 5'-OH dsRNA, both in the presence and absence of nucleotides, to full length RIG-I with all its methionine residues selectively labeled (Met-[ϵ-13CH3]). With this approach we were able to identify residues on the CTD, helicase domain, and CARDs that served as probes to sense RNA-induced conformational changes in those respective regions. Our results were analyzed in the context of either agonistic or antagonistic RNAs, by and large supporting a mechanism proposed by the Pyle Lab in which CARD release is primarily dependent on the RNA binding event.


Subject(s)
Trans-Activators , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Interferon Type I/genetics , Protein Structure, Tertiary , RNA, Double-Stranded , RNA, Viral/genetics , RNA, Viral/metabolism , Signal Transduction , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...