Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 472
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.
Mol Immunol ; 170: 131-143, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663254

ABSTRACT

Mammalian reovirus (MRV) is a non-enveloped, gene segmented double-stranded RNA (dsRNA) virus. It is an important zoonotic pathogen that infects many mammals and vertebrates that act as natural hosts and causes respiratory and digestive tract diseases. Studies have reported that RIG-I and MDA5 in the innate immune cytoplasmic RNA-sensing RIG-like receptor (RLR) signaling pathway can recognize dsRNA from MRV and promote antiviral type I interferon (IFN) responses. However, the mechanism by which many MRV-encoded proteins evade the host innate immune response remains unclear. Here, we show that exogenous µ1 protein promoted the proliferation of MRV in vitro, while knockdown of MRV µ1 protein expression by shRNA could impair MRV proliferation. Specifically, µ1 protein inhibited MRV or poly(I:C)-induced IFN-ß expression, and attenuated RIG-I/MDA5-mediated signaling axis transduction during MRV infection. Importantly, we found that µ1 protein significantly decreased IFN-ß mRNA expression induced by MDA5, RIG-I, MAVS, TBK1, IRF3(5D), and degraded the protein expression of exogenous MDA5, RIG-I, MAVS, TBK1 and IRF3 via the proteasomal and lysosomal pathways. Additionally, we show that µ1 protein can physically interact with MDA5, RIG-I, MAVS, TBK1, and IRF3 and attenuate the RIG-I/MDA5-mediated signaling cascades by blocking the phosphorylation and nuclear translocation of IRF3. In conclusion, our findings reveal that MRV outer capsid protein µ1 is a key factor in antagonizing RLRs signaling cascades and provide new strategies for effective prevention and treatment of MRV infection.


Subject(s)
DEAD Box Protein 58 , Interferon Regulatory Factor-3 , Interferon-Induced Helicase, IFIH1 , Orthoreovirus, Mammalian , Receptors, Immunologic , Signal Transduction , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon Regulatory Factor-3/metabolism , DEAD Box Protein 58/metabolism , Signal Transduction/immunology , Humans , Phosphorylation , Orthoreovirus, Mammalian/immunology , Orthoreovirus, Mammalian/physiology , HEK293 Cells , Interferon-beta/metabolism , Interferon-beta/immunology , Animals , Cell Nucleus/metabolism , Reoviridae Infections/immunology , Viral Proteins/metabolism , Active Transport, Cell Nucleus , Immunity, Innate/immunology , Protein Serine-Threonine Kinases
3.
J Virol ; 98(5): e0157323, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38572974

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and characterized by dysregulated immune response. Studies have shown that the SARS-CoV-2 accessory protein ORF7b induces host cell apoptosis through the tumor necrosis factor alpha (TNF-α) pathway and blocks the production of interferon beta (IFN-ß). The underlying mechanism remains to be investigated. In this study, we found that ORF7b facilitated viral infection and production, and inhibited the RIG-I-like receptor (RLR) signaling pathway through selectively interacting with mitochondrial antiviral-signaling protein (MAVS). MAVS439-466 region and MAVS Lys461 were essential for the physical association between MAVS and ORF7b, and the inhibition of the RLR signaling pathway by ORF7b. MAVSK461/K63 ubiquitination was essential for the RLR signaling regulated by the MAVS-ORF7b complex. ORF7b interfered with the recruitment of tumor necrosis factor receptor-related factor 6 (TRAF6) and the activation of the RLR signaling pathway by MAVS. Furthermore, interfering peptides targeting the ORF7b complex reversed the ORF7b-suppressed MAVS-RLR signaling pathway. The most potent interfering peptide V disrupts the formation of ORF7b tetramers, reverses the levels of the ORF7b-inhibited physical association between MAVS and TRAF6, leading to the suppression of viral growth and infection. Overall, this study provides a mechanism for the suppression of innate immunity by SARS-CoV-2 infection and the mechanism-based approach via interfering peptides to potentially prevent SARS-CoV-2 infection.IMPORTANCEThe pandemic coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and continues to be a threat to public health. It is imperative to understand the biology of SARS-CoV-2 infection and find approaches to prevent SARS-CoV-2 infection and ameliorate COVID-19. Multiple SARS-CoV-2 proteins are known to function on the innate immune response, but the underlying mechanism remains unknown. This study shows that ORF7b inhibits the RIG-I-like receptor (RLR) signaling pathway through the physical association between ORF7b and mitochondrial antiviral-signaling protein (MAVS), impairing the K63-linked MAVS polyubiquitination and its recruitment of tumor necrosis factor receptor-related factor 6 (TRAF6) to MAVS. The most potent interfering peptide V targeting the ORF7b-MAVS complex may reverse the suppression of the MAVS-mediated RLR signaling pathway by ORF7b and prevent viral infection and production. This study may provide new insights into the pathogenic mechanism of SARS-CoV-2 and a strategy to develop new drugs to prevent SARS-CoV-2 infection.


Subject(s)
Adaptor Proteins, Signal Transducing , COVID-19 , DEAD Box Protein 58 , SARS-CoV-2 , Signal Transduction , TNF Receptor-Associated Factor 6 , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , TNF Receptor-Associated Factor 6/metabolism , DEAD Box Protein 58/metabolism , HEK293 Cells , COVID-19/virology , COVID-19/immunology , COVID-19/metabolism , Ubiquitination , Receptors, Immunologic/metabolism , Animals , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics , Interferon-beta/metabolism , Apoptosis , Immunity, Innate , Tumor Necrosis Factor-alpha/metabolism
4.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673764

ABSTRACT

The exacerbation of pneumonia in children with human adenovirus type 3 (HAdV-3E) is secondary to a Staphylococcus aureus (S. aureus) infection. The influence of host-pathogen interactions on disease progression remains unclear. It is important to note that S. aureus infections following an HAdV-3E infection are frequently observed in clinical settings, yet the underlying susceptibility mechanisms are not fully understood. This study utilized an A549 cell model to investigate secondary infection with S. aureus following an HAdV-3E infection. The findings suggest that HAdV-3E exacerbates the S. aureus infection by intensifying lung epithelial cell damage. The results highlight the role of HAdV-3E in enhancing the interferon signaling pathway through RIG-I (DDX58), resulting in the increased expression of interferon-stimulating factors like MX1, RSAD2, and USP18. The increase in interferon-stimulating factors inhibits the NF-κB and MAPK/P38 pro-inflammatory signaling pathways. These findings reveal new mechanisms of action for HAdV-3E and S. aureus in secondary infections, enhancing our comprehension of pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Adenovirus Infections, Human , Adenoviruses, Human , DEAD Box Protein 58 , Signal Transduction , Staphylococcal Infections , Staphylococcus aureus , Ubiquitin Thiolesterase , Humans , DEAD Box Protein 58/metabolism , Staphylococcus aureus/pathogenicity , A549 Cells , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Adenoviruses, Human/physiology , Adenoviruses, Human/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adenovirus Infections, Human/metabolism , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/virology , Host-Pathogen Interactions/immunology , Receptors, Immunologic/metabolism , Coinfection/microbiology , NF-kappa B/metabolism , Inflammation/metabolism
5.
Vet Res ; 55(1): 44, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589930

ABSTRACT

Swine acute diarrhea syndrome coronavirus (SADS-CoV), an emerging Alpha-coronavirus, brings huge economic loss in swine industry. Interferons (IFNs) participate in a frontline antiviral defense mechanism triggering the activation of numerous downstream antiviral genes. Here, we demonstrated that TRIM25 overexpression significantly inhibited SADS-CoV replication, whereas TRIM25 deficiency markedly increased viral yield. We found that SADS-CoV N protein suppressed interferon-beta (IFN-ß) production induced by Sendai virus (SeV) or poly(I:C). Moreover, we determined that SADS-CoV N protein interacted with RIG-I N-terminal two caspase activation and recruitment domains (2CARDs) and TRIM25 coiled-coil dimerization (CCD) domain. The interaction of SADS-CoV N protein with RIG-I and TRIM25 caused TRIM25 multimerization inhibition, the RIG-I-TRIM25 interaction disruption, and consequent the IRF3 and TBK1 phosphorylation impediment. Overexpression of SADS-CoV N protein facilitated the replication of VSV-GFP by suppressing IFN-ß production. Our results demonstrate that SADS-CoV N suppresses the host IFN response, thus highlighting the significant involvement of TRIM25 in regulating antiviral immune defenses.


Subject(s)
Alphacoronavirus , Nucleocapsid Proteins , Animals , Swine , Alphacoronavirus/metabolism , Interferons/genetics , DEAD Box Protein 58/metabolism
6.
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
7.
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
8.
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
9.
Cell Rep ; 43(3): 113945, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38483900

ABSTRACT

U1 small nuclear RNA (snRNA) is an abundant and evolutionarily conserved 164-nucleotide RNA species that functions in pre-mRNA splicing, and it is considered to be a housekeeping non-coding RNA. However, the role of U1 snRNA in regulating host antiviral immunity remains largely unexplored. Here, we find that RNVU1-18, a U1 pseudogene, is significantly upregulated in the host infected with RNA viruses, including influenza and respiratory syncytial virus. Overexpression of U1 snRNA protects cells against RNA viruses, while knockdown of U1 snRNA leads to more viral burden in vitro and in vivo. Knockout of RNVU1-18 is sufficient to impair the type I interferon-dependent antiviral innate immunity. U1 snRNA is required to fully activate the retinoic acid-inducible gene I (RIG-I)-dependent antiviral signaling, since it interacts with tripartite motif 25 (TRIM25) and enhances the RIG-I-TRIM25 interaction to trigger K63-linked ubiquitination of RIG-I. Our study reveals the important role of housekeeping U1 snRNA in regulating host antiviral innate immunity and restricting RNA virus infection.


Subject(s)
Transcription Factors , Ubiquitin-Protein Ligases , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , DEAD Box Protein 58/metabolism , Immunity, Innate , RNA, Small Nuclear , Ubiquitination , Tripartite Motif Proteins/metabolism
10.
J Virol ; 98(4): e0014624, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38440983

ABSTRACT

Peste des petits ruminants is an acute and highly contagious disease caused by the Peste des petits ruminants virus (PPRV). Host proteins play a crucial role in viral replication. However, the effect of fusion (F) protein-interacting partners on PPRV infection is poorly understood. In this study, we found that the expression of goat plasminogen activator urokinase (PLAU) gradually decreased in a time- and dose-dependent manner in PPRV-infected goat alveolar macrophages (GAMs). Goat PLAU was subsequently identified using co-immunoprecipitation and confocal microscopy as an F protein binding partner. The overexpression of goat PLAU inhibited PPRV growth and replication, whereas silencing goat PLAU promoted viral growth and replication. Additionally, we confirmed that goat PLAU interacted with a virus-induced signaling adapter (VISA) to antagonize F-mediated VISA degradation, increasing the production of type I interferon. We also found that goat PLAU reduced the inhibition of PPRV replication in VISA-knockdown GAMs. Our results show that the host protein PLAU inhibits the growth and replication of PPRV by VISA-triggering RIG-I-like receptors and provides insight into the host protein that antagonizes PPRV immunosuppression.IMPORTANCEThe role of host proteins that interact with Peste des petits ruminants virus (PPRV) fusion (F) protein in PPRV replication is poorly understood. This study confirmed that goat plasminogen activator urokinase (PLAU) interacts with the PPRV F protein. We further discovered that goat PLAU inhibited PPRV replication by enhancing virus-induced signaling adapter (VISA) expression and reducing the ability of the F protein to degrade VISA. These findings offer insights into host resistance to viral invasion and suggest new strategies and directions for developing PPR vaccines.


Subject(s)
Goat Diseases , Goats , Host-Pathogen Interactions , Peste-des-Petits-Ruminants , Peste-des-petits-ruminants virus , Urokinase-Type Plasminogen Activator , Viral Fusion Proteins , Animals , Adaptor Proteins, Signal Transducing/metabolism , DEAD Box Protein 58/metabolism , Goat Diseases/immunology , Goat Diseases/metabolism , Goat Diseases/virology , Goats/immunology , Goats/virology , Macrophages, Alveolar , Peste-des-Petits-Ruminants/immunology , Peste-des-Petits-Ruminants/metabolism , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/growth & development , Peste-des-petits-ruminants virus/immunology , Peste-des-petits-ruminants virus/metabolism , Protein Binding , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Viral Fusion Proteins/metabolism
11.
PLoS One ; 19(2): e0281564, 2024.
Article in English | MEDLINE | ID: mdl-38394154

ABSTRACT

Retinoic acid inducible gene I (Rig-I) is a cytosolic pattern recognition receptor canonically described for its important role in sensing viral RNAs. Increasingly, bacterially-derived RNA from intracellular bacteria such as Mycobacterium tuberculosis, have been shown to activate the same host Rig-I/Mitochondrial antiviral sensing protein (MAVS) signaling pathway to drive a type-I interferon response that contributes to bacterial pathogenesis in vivo. In M. tuberculosis, this response is mediated by the protein secretion system SecA2, but little is known about whether this process is conserved in other pathogenic mycobacteria or the mechanism by which these nucleic acids gain access to the host cytoplasm. Because the M. tuberculosis and M. marinum SecA2 protein secretion systems share a high degree of genetic and functional conservation, we hypothesized that Rig-I/MAVS activation and subsequent induction of IFN-ß secretion by host macrophages will also be conserved between these two mycobacterial species. To test this, we generated a ΔsecA2 M. marinum strain along with complementation strains expressing either the M. marinum or M. tuberculosis secA2 genes. Our results suggest that the ΔsecA2 strain has a growth defect in vitro but not in host macrophages. These intracellular growth curves also suggested that the calculation applied to estimate the number of bacteria added to macrophage monolayers in infection assays underestimates bacterial inputs for the ΔsecA2 strain. Therefore, to better examine secreted IFN-ß levels when bacterial infection levels are equal across strains we plated bacterial CFUs at 2hpi alongside our ELISA based infections. This enabled us to normalize secreted levels of IFN-ß to a standard number of bacteria. Applying this approach to both WT and MAVS-/- bone marrow derived macrophages we observed equal or higher levels of secreted IFN-ß from macrophages infected with the ΔsecA2 M. marinum strain as compared to WT. Together our findings suggest that activation of host Rig-I/MAVS cytosolic sensors and subsequent induction of IFN-ß response in a SecA2-dependent manner is not conserved in M. marinum under the conditions tested.


Subject(s)
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Humans , Mycobacterium tuberculosis/genetics , Mycobacterium marinum/genetics , Signal Transduction , Macrophages/metabolism , DEAD Box Protein 58/metabolism , Tuberculosis/pathology
12.
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
13.
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
14.
Microbiol Spectr ; 12(1): e0312423, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38019020

ABSTRACT

IMPORTANCE: miR-26a serves as a potent positive regulator of type I interferon (IFN) responses. By inhibiting USP15 expression, miR-26a promotes RIG-I K63-ubiquitination to enhance type I IFN responses, resulting in an active antiviral state against viruses. Being an intricate regulatory network, the activation of type I IFN responses could in turn suppress miR-26a expression to avoid the disordered activation that might result in the so-called "type I interferonopathy." The knowledge gained would be essential for the development of novel antiviral strategies against viral infection.


Subject(s)
Interferon Type I , MicroRNAs , DEAD Box Protein 58/metabolism , Signal Transduction , MicroRNAs/genetics , Antiviral Agents/pharmacology , Immunity, Innate
15.
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
16.
Nat Commun ; 14(1): 7308, 2023 11 11.
Article in English | MEDLINE | ID: mdl-37951994

ABSTRACT

RIG-I is an essential innate immune receptor that responds to infection by RNA viruses. The RIG-I signaling cascade is mediated by a series of post-translational modifications, the most important of which is ubiquitination of the RIG-I Caspase Recruitment Domains (CARDs) by E3 ligase Riplet. This is required for interaction between RIG-I and its downstream adapter protein MAVS, but the mechanism of action remains unclear. Here we show that Riplet is required for RIG-I signaling in the presence of both short and long dsRNAs, establishing that Riplet activation does not depend upon RIG-I filament formation on long dsRNAs. Likewise, quantitative Riplet-RIG-I affinity measurements establish that Riplet interacts with RIG-I regardless of whether the receptor is bound to RNA. To understand this, we solved high-resolution cryo-EM structures of RIG-I/RNA/Riplet complexes, revealing molecular interfaces that control Riplet-mediated activation and enabling the formulation of a unified model for the role of Riplet in signaling.


Subject(s)
Immunity, Innate , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , DEAD Box Protein 58/metabolism , Signal Transduction , Ubiquitination , RNA, Double-Stranded
17.
Viruses ; 15(11)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-38005825

ABSTRACT

Nipah virus (NiV; genus: Henipavirus; family: Paramyxoviridae) naturally infects Old World fruit bats (family Pteropodidae) without causing overt disease. Conversely, NiV infection in humans and other mammals can be lethal. Comparing bat antiviral responses with those of humans may illuminate the mechanisms that facilitate bats' tolerance. Tripartite motif proteins (TRIMs), a large family of E3-ubiquitin ligases, fine-tune innate antiviral immune responses, and two human TRIMs interact with Henipavirus proteins. We hypothesize that NiV infection induces the expression of an immunosuppressive TRIM in bat, but not human cells, to promote tolerance. Here, we show that TRIM40 is an interferon-stimulated gene (ISG) in pteropodid but not human cells. Knockdown of bat TRIM40 increases gene expression of IFNß, ISGs, and pro-inflammatory cytokines following poly(I:C) transfection. In Pteropus vampyrus, but not human cells, NiV induces TRIM40 expression within 16 h after infection, and knockdown of TRIM40 correlates with reduced NiV titers as compared to control cells. Bats may have evolved to express TRIM40 in response to viral infections to control immunopathogenesis.


Subject(s)
Chiroptera , DEAD Box Protein 58 , Henipavirus Infections , Tripartite Motif Proteins , Animals , Humans , Chiroptera/immunology , Chiroptera/virology , Immunity, Innate , Interferons/genetics , Nipah Virus/genetics , Tripartite Motif Proteins/metabolism , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism
18.
Cell Mol Immunol ; 20(12): 1457-1471, 2023 12.
Article in English | MEDLINE | ID: mdl-37978243

ABSTRACT

The G protein-coupled receptor ADGRE5 (CD97) binds to various metabolites that play crucial regulatory roles in metabolism. However, its function in the antiviral innate immune response remains to be determined. In this study, we report that CD97 inhibits virus-induced type-I interferon (IFN-I) release and enhances RNA virus replication in cells and mice. CD97 was identified as a new negative regulator of the innate immune receptor RIG-I, and RIG-1 degradation led to the suppression of the IFN-I signaling pathway. Furthermore, overexpression of CD97 promoted the ubiquitination of RIG-I, resulting in its degradation, but did not impact its mRNA expression. Mechanistically, CD97 upregulates RNF125 expression to induce RNF125-mediated RIG-I degradation via K48-linked ubiquitination at Lys181 after RNA virus infection. Most importantly, CD97-deficient mice are more resistant than wild-type mice to RNA virus infection. We also found that sanguinarine-mediated inhibition of CD97 effectively blocks VSV and SARS-CoV-2 replication. These findings elucidate a previously unknown mechanism through which CD97 negatively regulates RIG-I in the antiviral innate immune response and provide a molecular basis for the development of new therapeutic strategies and the design of targeted antiviral agents.


Subject(s)
RNA Virus Infections , RNA Viruses , Animals , Mice , Antiviral Agents/pharmacology , DEAD Box Protein 58/metabolism , Immunity, Innate , Receptors, G-Protein-Coupled/metabolism , RNA Virus Infections/genetics , RNA Viruses/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
19.
Cell Rep ; 42(11): 113388, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37934668

ABSTRACT

Cancer evades host immune surveillance by virtue of poor immunogenicity. Here, we report an immune suppressor, designated as PTIR1, that acts as a promotor of tumor immune resistance. PTIR1 is selectively induced in human cancers via alternative splicing of DDX58 (RIG-I), and its induction is closely related to poor outcome in patients with cancer. Through blocking the recruitment of leukocytes, PTIR1 facilitates cancer immune escape and tumor-intrinsic resistance to immunotherapeutic treatments. Unlike RIG-I, PTIR1 is capable of binding to the C terminus of UCHL5 and activates its ubiquitinating function, which in turn inhibits immunoproteasome activity and limits neoantigen processing and presentation, consequently blocking T cell recognition and attack against cancer. Moreover, we find that the adenosine deaminase ADAR1 induces A-to-I RNA editing on DDX58 transcript, thus triggering PTIR1 production. Collectively, our data uncover the immunosuppressive role of PTIR1 in tumorigenesis and propose that ADAR1-PTIR1-UCHL5 signaling is a potential cancer immunotherapeutic target.


Subject(s)
Cell Communication , Signal Transduction , Humans , Carcinogenesis/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , DEAD Box Protein 58/metabolism , Receptors, Immunologic , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...