Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 329
Filtrer
1.
mSphere ; 9(6): e0023624, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38757961

RÉSUMÉ

Mammalian orthoreovirus (MRV) outer capsid protein σ3 is a multifunctional protein containing a double-stranded RNA-binding domain, which facilitates viral entry and assembly. We reasoned that σ3 has an innate immune evasion function. Here, we show that σ3 protein localizes in the mitochondria and interacts with mitochondrial antiviral signaling protein (MAVS) to activate the intrinsic mitochondria-mediated apoptotic pathway. Consequently, σ3 protein promotes the degradation of MAVS through the intrinsic caspase-9/caspase-3 apoptotic pathway. Moreover, σ3 protein can also inhibit the expression of the components of the RNA-sensing retinoic acid-inducible gene (RIG)-like receptor (RLR) signaling pathway to block antiviral type I interferon responses. Mechanistically, σ3 inhibits RIG-I and melanoma differentiation-associated gene 5 expression is independent of its inhibitory effect on MAVS. Overall, we demonstrate that the MRV σ3 protein plays a vital role in negatively regulating the RLR signaling pathway to inhibit antiviral responses. This enables MRV to evade host defenses to facilitate its own replication providing a target for the development of effective antiviral drugs against MRV. IMPORTANCE: Mammalian orthoreovirus (MRV) is an important zoonotic pathogen, but the regulatory role of its viral proteins in retinoic acid-inducible gene-like receptor (RLR)-mediated antiviral responses is still poorly understood. Herein, we show that MRV σ3 protein co-localizes with mitochondrial antiviral signaling protein (MAVS) in the mitochondria and promotes the mitochondria-mediated intrinsic apoptotic pathway to cleave and consequently degrade MAVS. Furthermore, tryptophan at position 133 of σ3 protein plays a key role in the degradation of MAVS. Importantly, we show that MRV outer capsid protein σ3 is a key factor in antagonizing RLR-mediated antiviral responses, providing evidence to better unravel the infection and transmission mechanisms of MRV.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines de capside , Orthoréovirus mammalien , Transduction du signal , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines de capside/métabolisme , Protéines de capside/génétique , Humains , Orthoréovirus mammalien/génétique , Animaux , Apoptose , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Mitochondries/métabolisme , Immunité innée , Souris , Échappement immunitaire , Cellules HEK293 , Récepteurs immunologiques/métabolisme , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Hélicase IFIH1 inductrice de l'interféron/génétique , Lignée cellulaire , Interactions hôte-pathogène
2.
Redox Biol ; 73: 103196, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38772149

RÉSUMÉ

Hippocampal neural stem/progenitor cells (NSPCs) are highly vulnerable to different stress stimuli, resulting in adult neurogenesis decline and eventual cognitive defects. Our previous study demonstrated that NOD-like receptor family pyrin domain-containing 6 (Nlrp6) highly expressed in NSPCs played a critical role in sustaining hippocampal neurogenesis to resist stress-induced depression, but the underlying mechnistms are still unclear. Here, we found that Nlrp6 depletion led to cognitive defects and hippocampal NSPC loss in mice. RNA-sequencing analysis of the primary NSPCs revealed that Nlrp6 deficiency altered gene expression profiles of mitochondrial energy generation and ferroptotic process. Upon siNlrp6 transfection, as well as corticosterone (CORT) exposure, downregulation of Nlrp6 suppressed retinoic acid-inducible gene I (RIG-1)/mitochondrial antiviral signaling proteins (MAVS)-mediated autophagy, but drove NSPC ferroptotic death. More interesting, short chain fatty acids (SCFAs) upregulated Nlrp6 expression and promoted RIG-1/MAVS-mediated mitophagy, preventing CORT-induced NSPC ferroptosis. Our study further demonstrates that Nlrp6 should be a sensor for RIG-1/MAVS-mediated mitophagy and play a critical role in maintain mitochondrial homeostasis of hippocampal NSPCs. These results suggests that Nlrp6 should be a potential drug target to combat neurodegenerative diseases relative with chronic stress.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Corticostérone , Protéine-58 à domaine DEAD , Ferroptose , Mitophagie , Cellules souches neurales , Animaux , Souris , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Corticostérone/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Cellules souches neurales/métabolisme , Hippocampe/métabolisme , Mitochondries/métabolisme , Transduction du signal , Récepteurs de surface cellulaire
3.
J Virol ; 98(6): e0046124, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38780247

RÉSUMÉ

Transmissible gastroenteritis virus (TGEV)-induced enteritis is characterized by watery diarrhea, vomiting, and dehydration, and has high mortality in newborn piglets, resulting in significant economic losses in the pig industry worldwide. Conventional cell lines have been used for many years to investigate inflammation induced by TGEV, but these cell lines may not mimic the actual intestinal environment, making it difficult to obtain accurate results. In this study, apical-out porcine intestinal organoids were employed to study TEGV-induced inflammation. We found that apical-out organoids were susceptible to TGEV infection, and the expression of representative inflammatory cytokines was significantly upregulated upon TGEV infection. In addition, retinoic acid-inducible gene I (RIG-I) and the nuclear factor-kappa B (NF-κB) pathway were responsible for the expression of inflammatory cytokines induced by TGEV infection. We also discovered that the transcription factor hypoxia-inducible factor-1α (HIF-1α) positively regulated TGEV-induced inflammation by activating glycolysis in apical-out organoids, and pig experiments identified the same molecular mechanism as the ex vivo results. Collectively, we unveiled that the inflammatory responses induced by TGEV were modulated via the RIG-I/NF-κB/HIF-1α/glycolysis axis ex vivo and in vivo. This study provides novel insights into TGEV-induced enteritis and verifies intestinal organoids as a reliable model for investigating virus-induced inflammation. IMPORTANCE: Intestinal organoids are a newly developed culture system for investigating immune responses to virus infection. This culture model better represents the physiological environment compared with well-established cell lines. In this study, we discovered that inflammatory responses induced by TGEV infection were regulated by the RIG-I/NF-κB/HIF-1α/glycolysis axis in apical-out porcine organoids and in pigs. Our findings contribute to understanding the mechanism of intestinal inflammation upon viral infection and highlight apical-out organoids as a physiological model to mimic virus-induced inflammation.


Sujet(s)
Gastroentérite transmissible du porc , Glycolyse , Inflammation , Organoïdes , Virus de la gastroentérite transmissible , Animaux , Cytokines/métabolisme , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Gastroentérite transmissible du porc/virologie , Gastroentérite transmissible du porc/métabolisme , Gastroentérite transmissible du porc/anatomopathologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Inflammation/métabolisme , Inflammation/virologie , Intestins/virologie , Intestins/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Organoïdes/virologie , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Transduction du signal , Suidae , Virus de la gastroentérite transmissible/physiologie
4.
Vet Microbiol ; 294: 110124, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38795403

RÉSUMÉ

PEDV, a single-stranded RNA virus, causes significant economic losses in the pig industry. Sin3-associated protein 18 (SAP18) is known for its role in transcriptional inhibition and RNA splicing. However, research on SAP18's involvement in PEDV infection is limited. Here, we identified an interaction between SAP18 and PEDV nonstructural protein 10 (Nsp10) using immunoprecipitation-mass spectrometry (IP-MS) and confirmed it through immunoprecipitation and laser confocal microscopy. Additionally, PEDV Nsp10 reduced SAP18 protein levels and induced its cytoplasmic accumulation. Overexpressing SAP18 suppressed PEDV replication, meanwhile its knockdown via short interfering RNA (siRNA) enhanced replication. SAP18 overexpression boosted IRF3 and NF-κB P65 phosphorylation, nuclear translocation, and IFN-ß antiviral response. Furthermore, SAP18 upregulated RIG-I expression and facilitated its dephosphorylation, while SAP18 knockdown had the opposite effect. Finally, SAP18 interacted with phosphatase 1 (PP1) catalytic subunit alpha (PPP1CA), promoting PPP1CA-RIG-I interaction during PEDV infection. These findings highlight SAP18's role in activating the type I interferon pathway and inhibiting viral replication by promoting RIG-I dephosphorylation through its interaction with PPP1CA.


Sujet(s)
Virus de la diarrhée porcine épidémique , Protéines virales non structurales , Réplication virale , Animaux , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Virus de la diarrhée porcine épidémique/physiologie , Virus de la diarrhée porcine épidémique/génétique , Phosphorylation , Suidae , Lignée cellulaire , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Chlorocebus aethiops
5.
Vet Microbiol ; 294: 110127, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38797057

RÉSUMÉ

Glaesserella parasuis (G. parasuis) is a common Gram-negative commensal bacterium in the upper respiratory tract of swine that can cause Glässer's disease under stress conditions. Pyroptosis is an important immune defence mechanism of the body that plays a crucial role in clearing pathogen infections and endogenous danger signals. This study aimed to investigate the mechanism of G. parasuis serotype 5 SQ (GPS5-SQ)-induced pyroptosis in swine tracheal epithelial cells (STECs). The results of the present study demonstrated that GPS5-SQ infection induces pyroptosis in STECs by enhancing the protein level of the N-terminal domain of gasdermin D (GSDMD-N) and activating the NOD-like receptor protein 3 (NLRP3) inflammasome. Furthermore, the levels of pyroptosis-related proteins, including GSDMD-N and cleaved caspase-1 were considerably decreased in STECs after the knockdown of retinoic acid inducible gene-I (RIG-I) and mitochondrial antiviral signaling protein (MAVS). These results indicated that GPS5-SQ might trigger pyroptosis through the activation of the RIG-I/MAVS/NLRP3 signaling pathway. More importantly, the reactive oxygen species (ROS) scavenger N-acetylcysteine (NAC) repressed the activation of the RIG-I/MAVS/NLRP3 signaling and rescued the decrease in Occludin and zonula occludens-1 (ZO-1) after GPS5-SQ infection. Overall, our findings show that GPS5-SQ can activate RIG-I/MAVS/NLRP3 signaling and destroy the integrity of the epithelial barrier by inducing ROS generation in STECs, shedding new light on G. parasuis pathogenesis.


Sujet(s)
Cellules épithéliales , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Transduction du signal , Animaux , Cellules épithéliales/microbiologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Suidae , Haemophilus parasuis/pathogénicité , Haemophilus parasuis/génétique , Trachée/microbiologie , Trachée/cytologie , Maladies des porcs/microbiologie , Sérogroupe , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Inflammasomes/métabolisme , Inflammasomes/génétique , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/métabolisme , Infections à Haemophilus/médecine vétérinaire , Infections à Haemophilus/microbiologie
6.
FASEB J ; 38(10): e23651, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38752537

RÉSUMÉ

Singleton-Merten syndrome (SMS) is a rare immunogenetic disorder affecting multiple systems, characterized by dental dysplasia, aortic calcification, glaucoma, skeletal abnormalities, and psoriasis. Glaucoma, a key feature of both classical and atypical SMS, remains poorly understood in terms of its molecular mechanism caused by DDX58 mutation. This study presented a novel DDX58 variant (c.1649A>C [p.Asp550Ala]) in a family with childhood glaucoma. Functional analysis showed that DDX58 variant caused an increase in IFN-stimulated gene expression and high IFN-ß-based type-I IFN. As the trabecular meshwork (TM) is responsible for controlling intraocular pressure (IOP), we examine the effect of IFN-ß on TM cells. Our study is the first to demonstrate that IFN-ß significantly reduced TM cell viability and function by activating autophagy. In addition, anterior chamber injection of IFN-ß remarkably increased IOP level in mice, which can be attenuated by treatments with autophagy inhibitor chloroquine. To uncover the specific mechanism underlying IFN-ß-induced autophagy in TM cells, we performed microarray analysis in IFN-ß-treated and DDX58 p.Asp550Ala TM cells. It showed that RSAD2 is necessary for IFN-ß-induced autophagy. Knockdown of RSAD2 by siRNA significantly decreased autophagy flux induced by IFN-ß. Our findings suggest that DDX58 mutation leads to the overproduction of IFN-ß, which elevates IOP by modulating autophagy through RSAD2 in TM cells.


Sujet(s)
Autophagie , Interféron bêta , Pression intraoculaire , Réseau trabéculaire de la sclère , Autophagie/effets des médicaments et des substances chimiques , Réseau trabéculaire de la sclère/métabolisme , Réseau trabéculaire de la sclère/effets des médicaments et des substances chimiques , Humains , Animaux , Souris , Pression intraoculaire/physiologie , Interféron bêta/métabolisme , Mâle , Femelle , Glaucome/anatomopathologie , Glaucome/métabolisme , Glaucome/génétique , Surdité neurosensorielle/génétique , Surdité neurosensorielle/anatomopathologie , Surdité neurosensorielle/métabolisme , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Souris de lignée C57BL , Mutation , Atrophie optique/génétique , Atrophie optique/métabolisme , Atrophie optique/anatomopathologie , Pedigree , Odontodysplasie , Calcification vasculaire , Hypoplasie de l'émail dentaire , Métacarpe/malformations , Ostéoporose , Maladies musculaires , Maladies de l'aorte , Récepteurs immunologiques
7.
Elife ; 132024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38747717

RÉSUMÉ

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.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , ARN double brin , Ribonuclease III , Animaux , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/composition chimique , Caenorhabditis elegans/génétique , Caenorhabditis elegans/métabolisme , ARN double brin/métabolisme , Ribonuclease III/métabolisme , Ribonuclease III/composition chimique , Ribonuclease III/génétique , Cryomicroscopie électronique , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/composition chimique , DEAD-box RNA helicases/génétique , RNA helicases/métabolisme , RNA helicases/génétique , RNA helicases/composition chimique , Liaison aux protéines , Adénosine triphosphate/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/composition chimique
8.
Biochem Biophys Res Commun ; 712-713: 149915, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38663038

RÉSUMÉ

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.


Sujet(s)
Escherichia coli , Levivirus , ARN double brin , Escherichia coli/virologie , Escherichia coli/génétique , Escherichia coli/métabolisme , ARN double brin/métabolisme , Humains , Levivirus/génétique , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Immunité innée , Interféron bêta/métabolisme , Interféron bêta/génétique , Facteur de transcription NF-kappa B/métabolisme , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Transduction du signal , Facteur-7 de régulation d'interféron/métabolisme , Facteur-7 de régulation d'interféron/génétique , Récepteurs immunologiques , Facteur-3 de régulation d'interféron/métabolisme , Facteur-3 de régulation d'interféron/génétique
9.
J Transl Med ; 22(1): 395, 2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38685028

RÉSUMÉ

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.


Sujet(s)
Apoptose , Gliome , Petit ARN interférent , Humains , Animaux , Lignée cellulaire tumorale , Gliome/thérapie , Gliome/anatomopathologie , Gliome/génétique , Petit ARN interférent/métabolisme , Souris nude , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Prolifération cellulaire , Mouvement cellulaire , Tests d'activité antitumorale sur modèle de xénogreffe , Souris , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Espèces réactives de l'oxygène/métabolisme , Invasion tumorale , Survie cellulaire
10.
J Agric Food Chem ; 72(17): 9782-9794, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38597360

RÉSUMÉ

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.


Sujet(s)
Protéine-58 à domaine DEAD , Sous-type H1N1 du virus de la grippe A , Grippe humaine , Souris knockout , Oligosaccharides , Infections à Orthomyxoviridae , Transduction du signal , Facteur-3 associé aux récepteurs de TNF , Animaux , Souris , Oligosaccharides/administration et posologie , Oligosaccharides/composition chimique , Oligosaccharides/pharmacologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/métabolisme , Sous-type H1N1 du virus de la grippe A/immunologie , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie , Grippe humaine/immunologie , Grippe humaine/prévention et contrôle , Grippe humaine/métabolisme , Facteur-3 associé aux récepteurs de TNF/génétique , Facteur-3 associé aux récepteurs de TNF/métabolisme , Facteur-3 associé aux récepteurs de TNF/immunologie , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/immunologie , Pneumopathie infectieuse/immunologie , Pneumopathie infectieuse/prévention et contrôle , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/virologie , Souris de lignée C57BL , Poumon/immunologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/virologie , Cytokines/métabolisme , Cytokines/immunologie , Cytokines/génétique , Femelle , Facteur de transcription NF-kappa B/immunologie , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Anti-inflammatoires/administration et posologie , Anti-inflammatoires/pharmacologie
11.
DNA Cell Biol ; 43(4): 197-205, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38466944

RÉSUMÉ

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.


Sujet(s)
Immunité innée , Interféron de type I , Protéine-58 à domaine DEAD/génétique , ADN , Interféron de type I/génétique , Récepteurs immunologiques/génétique , ARN , Humains
12.
Microbes Infect ; 26(4): 105321, 2024.
Article de Anglais | MEDLINE | ID: mdl-38461968

RÉSUMÉ

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.


Sujet(s)
Protéine-58 à domaine DEAD , Virus de la rage , Humains , Lignée cellulaire , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie , Rage (maladie)/immunologie , Rage (maladie)/virologie , Vaccins antirabiques/immunologie , Virus de la rage/immunologie , Virus de la rage/génétique , Virus de la rage/pathogénicité , Récepteurs immunologiques/métabolisme , ARN viral/génétique , Transduction du signal , Réplication virale
13.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167092, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38382623

RÉSUMÉ

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.


Sujet(s)
Histone , Maladies virales , Protéine-58 à domaine DEAD/génétique , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Histone/métabolisme , Immunité innée , Protein-arginine deiminases/génétique , Protein-arginine deiminases/métabolisme , Récepteurs immunologiques/métabolisme
14.
Cancer Res Commun ; 4(2): 540-555, 2024 02 26.
Article de Anglais | MEDLINE | ID: mdl-38358346

RÉSUMÉ

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.


Sujet(s)
Tumeurs colorectales , Interféron de type I , Animaux , Souris , Rétroéléments , Interféron lambda , Aurora kinases/métabolisme , Interféron de type I/métabolisme , Protéine-58 à domaine DEAD/génétique , Récepteurs immunologiques
15.
Nat Commun ; 15(1): 780, 2024 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-38278841

RÉSUMÉ

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.


Sujet(s)
Virus à ARN , Souris , Animaux , Ubiquitination , Lignée cellulaire , Protéines à motif tripartite/génétique , Protéines à motif tripartite/métabolisme , Immunité innée , Ubiquitin-protein ligases/métabolisme , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/métabolisme
16.
Sci Rep ; 14(1): 1455, 2024 01 17.
Article de Anglais | MEDLINE | ID: mdl-38228690

RÉSUMÉ

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.


Sujet(s)
Vaccins antigrippaux , Grippe humaine , Humains , Méthylation de l'ADN , Grippe humaine/prévention et contrôle , Agranulocytes , Vaccination/méthodes , Protéine-58 à domaine DEAD/génétique , Transduction du signal , Anticorps antiviraux
17.
Nucleic Acids Res ; 52(1): 355-369, 2024 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-38015453

RÉSUMÉ

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.


Sujet(s)
DEAD-box RNA helicases , RNA helicases , Humains , Adenosine triphosphatases/génétique , Adenosine triphosphatases/métabolisme , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/métabolisme , DEAD-box RNA helicases/métabolisme , Helicase/génétique , Helicase/métabolisme , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Liaison aux protéines/physiologie , Récepteurs immunologiques/génétique , RNA helicases/métabolisme , ARN double brin , ARN viral/métabolisme
18.
Cell Death Differ ; 31(1): 28-39, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-38001254

RÉSUMÉ

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.


Sujet(s)
Virus à ARN , Ubiquitin-protein ligases , Ubiquitination , Ubiquitin-protein ligases/métabolisme , Ubiquitine/métabolisme , Transduction du signal , Protéine-58 à domaine DEAD/génétique , Virus à ARN/métabolisme
19.
Virology ; 589: 109942, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-38048647

RÉSUMÉ

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.


Sujet(s)
Virus Hantaan , Interféron de type I , Interféron de type I/métabolisme , Virus Hantaan/génétique , Virus Hantaan/métabolisme , Protéines à motif tripartite/génétique , Protéines à motif tripartite/métabolisme , Transduction du signal , Immunité innée , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/métabolisme
20.
EMBO Rep ; 24(12): e57137, 2023 Dec 06.
Article de Anglais | MEDLINE | ID: mdl-37870297

RÉSUMÉ

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.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , Cellules Caco-2 , COVID-19/génétique , Transduction du signal , Protéine-58 à domaine DEAD/génétique , Protéine-58 à domaine DEAD/métabolisme , Immunité innée/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...