Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 444
Filtrer
1.
Methods Mol Biol ; 2854: 265-282, 2025.
Article de Anglais | MEDLINE | ID: mdl-39192136

RÉSUMÉ

Protein kinase R (PKR), a key double-stranded RNA (dsRNA)-activated sensor, is pivotal for cellular responses to diverse stimuli. This protocol delineates a comprehensive methodological framework employing single luciferase assays, yeast assays, immunoblot assays, and quantitative PCR (qPCR) to discern and validate PKR activities and their downstream impacts on NF-κB-activating signaling pathways. These methodologies furnish a systematic approach to unraveling the role of PKR as a dsRNA sensor and effector in antiviral innate immunity, enabling in-depth analyses of dsRNA sensor activities.


Sujet(s)
Immunité innée , ARN double brin , eIF-2 Kinase , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , ARN double brin/immunologie , ARN double brin/génétique , Humains , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal , Animaux
2.
Sci Immunol ; 9(100): eado8758, 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39365875

RÉSUMÉ

The interferon (IFN) response is vital for the effectiveness of immune checkpoint inhibition (ICI) therapy. Our previous research showed that KRAS (Kirsten rat sarcoma viral) mutation impairs the IFN response in colorectal cancer (CRC), with an unclear mechanism. Here, we demonstrate that KRAS accelerates double-stranded RNA (dsRNA) degradation, impairing dsRNA sensing and IFN response by down-regulating DExD/H-box helicase 6 (DDX60). DDX60 was identified as a KRAS target here and could bind to dsRNAs to protect against RNA-induced silencing complex (RISC)-mediated degradation. Overexpressing DDX60 induced dsRNA accumulation, reactivated IFN signaling, and increased CRC sensitivity to ICI therapy. Mechanistically, KRAS engaged the AKT (also known as protein kinase B)-GSK3ß (glycogen synthase kinase-3 beta) pathway to suppress STAT3 phosphorylation, thereby inhibiting STAT3-driven DDX60 transcription. Our findings reveal a role for KRAS in dsRNA homeostasis, suggesting potential strategies to convert "cold" tumors to "hot" and to overcome ICI resistance in CRC with KRAS mutations.


Sujet(s)
Tumeurs colorectales , DEAD-box RNA helicases , Protéines proto-oncogènes p21(ras) , ARN double brin , Tumeurs colorectales/immunologie , Tumeurs colorectales/génétique , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/immunologie , ARN double brin/immunologie , Humains , DEAD-box RNA helicases/génétique , DEAD-box RNA helicases/métabolisme , Animaux , Souris , Lignée cellulaire tumorale , Femelle
3.
J Immunol ; 213(5): 700-717, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39058317

RÉSUMÉ

dsRNA-dependent protein kinase R (PKR) is a key factor of innate immunity. It is involved in translation inhibition, apoptosis, and enhancement of the proinflammatory and IFN responses. However, how these antiviral functions are conserved during evolution remains largely unknown. Overexpression and knockout studies in a Chinook salmon (Oncorhynchus tshawytscha) cell line were conducted to assess the role of salmonid PKR in the antiviral response. Three distinct mRNA isoforms from a unique pkr gene, named pkr-fl (full length), pkr-ml (medium length) and pkr-sl (short length), were cloned and a pkr-/- clonal fish cell line was developed using CRISPR/Cas9 genome editing. PKR-FL includes an N-terminal dsRNA-binding domain and a C-terminal kinase domain, whereas PKR-ML and PKR-SL display a truncated or absent kinase domain, respectively. PKR-FL is induced during IFNA2 stimulation but not during viral hemorrhagic septicemia virus (VHSV) infection. Overexpression experiments showed that only PKR-FL possesses antiviral functions, including activation of apoptosis and inhibition of de novo protein synthesis. Knockout experiments confirmed that PKR is involved in apoptosis activation during the late stage of VHSV infection. Endogenous PKR also plays a critical role in translation inhibition upon poly(I:C) transfection after IFNA2 treatment. It is, however, not involved in translational arrest during VHSV infection. Extra- and intracellular titrations showed that endogenous PKR does not directly inhibit viral replication but apparently favors virion release into the supernatant, likely by triggering late apoptosis. Altogether, our data confirm that salmonid PKR has conserved molecular functions that VHSV appears to bypass with subversion strategies.


Sujet(s)
Apoptose , Biosynthèse des protéines , Saumon , eIF-2 Kinase , Animaux , Apoptose/immunologie , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , Saumon/immunologie , Lignée cellulaire , Biosynthèse des protéines/immunologie , Immunité innée , Novirhabdovirus/physiologie , Novirhabdovirus/immunologie , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/métabolisme , ARN double brin/immunologie , Maladies des poissons/immunologie , Systèmes CRISPR-Cas
4.
Proc Natl Acad Sci U S A ; 121(29): e2402126121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38980902

RÉSUMÉ

Upon sensing viral RNA, mammalian RIG-I-like receptors (RLRs) activate downstream signals using caspase activation and recruitment domains (CARDs), which ultimately promote transcriptional immune responses that have been well studied. In contrast, the downstream signaling mechanisms for invertebrate RLRs are much less clear. For example, the Caenorhabditis elegans RLR DRH-1 lacks annotated CARDs and up-regulates the distinct output of RNA interference. Here, we found that similar to mammal RLRs, DRH-1 signals through two tandem CARDs (2CARD) to induce a transcriptional immune response. Expression of DRH-1(2CARD) alone in the intestine was sufficient to induce immune gene expression, increase viral resistance, and promote thermotolerance, a phenotype previously associated with immune activation in C. elegans. We also found that DRH-1 is required in the intestine to induce immune gene expression, and we demonstrate subcellular colocalization of DRH-1 puncta with double-stranded RNA inside the cytoplasm of intestinal cells upon viral infection. Altogether, our results reveal mechanistic and spatial insights into antiviral signaling in C. elegans, highlighting unexpected parallels in RLR signaling between C. elegans and mammals.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Transduction du signal , Animaux , Caenorhabditis elegans/immunologie , Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/immunologie , Transduction du signal/immunologie , Intestins/immunologie , Intestins/virologie , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , ARN double brin/métabolisme , ARN double brin/immunologie , Immunité innée , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , ARN viral/immunologie , ARN viral/métabolisme , ARN viral/génétique
5.
Clin Cancer Res ; 30(16): 3355-3357, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38869441

RÉSUMÉ

The presence of moieties denoting viral infection is crucial to mount powerful cytotoxic T-cell immune responses acting through innate receptors such as Toll-like receptor 3. For cancer immunotherapy, several safe analogues of viral double-stranded RNA are under clinical development following compelling evidence for efficacy in mouse models. See related article by van Eijck et al., p. 3447.


Sujet(s)
Immunothérapie , Tumeurs , ARN double brin , Maladies virales , Humains , Tumeurs/thérapie , Tumeurs/immunologie , ARN double brin/immunologie , Immunothérapie/méthodes , Animaux , Maladies virales/immunologie , Récepteur de type Toll-3/génétique , Souris , Lymphocytes T cytotoxiques/immunologie , ARN viral
6.
Mol Immunol ; 170: 156-169, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38692097

RÉSUMÉ

Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer.


Sujet(s)
Tumeurs du sein , Protéines de liaison à l'ADN , Facteurs de transcription , Animaux , Femelle , Humains , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Régulation de l'expression des gènes tumoraux , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Facteur-1 de régulation d'interféron/immunologie , Facteur-3 de régulation d'interféron/métabolisme , Facteur-3 de régulation d'interféron/génétique , Interférons/métabolisme , Interférons/immunologie , Interférons/génétique , Récidive tumorale locale/immunologie , ARN double brin/immunologie , Transduction du signal/immunologie , Facteur de transcription RelA/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/immunologie
7.
Clin Rheumatol ; 43(3): 1111-1126, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38182800

RÉSUMÉ

BACKGROUND: Melanoma differentiation-associated gene 5 (MDA5), as a cytoplasmic sensor for viral double-stranded RNAs, has received increasing attention in recent years. Although considerable headway has been made on the functional role of MDA5 in antiviral immunity and autoimmune disease, the available literature is insufficient to assess the vast field. METHODS: This study performed a bibliometric analysis to investigate current hotspots in the global scientific output of MDA5 over the past two decades. Related publications and recorded information from 2002 to 2022 in the Web of Science Core Collection (WoSCC) database were retrieved. VOSviewer and CiteSpace were used for quantitative evaluation and visualization. RESULTS: A total of 2267 original articles and reviews were obtained, and the annual number of publications related to MDA5 was increasing rapidly. China has published the most papers, while the USA was the most influential country with the most citations and the highest H-index. The Chinese Academy of Sciences, the United States Department of Health and Human Services, and the Journal of Virology were the most prolific research affiliation, funding source, and journal, respectively. Fujita T (Kyoto University) was the most productive author with the highest H-index and had close cooperation with Kato H and Yoneyama M. The keywords "RIG-I," "MDA5," "innate immunity," "double-stranded-RNA," and "recognition" had the highest frequency, while "dermatomyositis" as well as "autoantibody" seemed to be the emerging hotspots. CONCLUSION: This study comprehensively demonstrated the research frontiers of MDA5 and will provide a useful resource for scholars to conduct future decisions. KEY POINTS: We conducted the first in-depth survey of the research frontiers on melanoma differentiation-associated gene 5 (MDA5) over the past two decades via bibliometric analysis. We found that many early breakthroughs have been made in the mechanism of MDA5-mediated antiviral immune responses, and the role of MDA5 in autoimmune and autoinflammatory diseases has raised the recent concern. We identified that the virus infection-associated pathogenesis and effective therapeutic strategy of anti-MDA5 antibody-positive dermatomyositis will remain the hotspots in the future.


Sujet(s)
Maladies auto-immunes , Hélicase IFIH1 inductrice de l'interféron , ARN viral , Humains , Autoanticorps/immunologie , Maladies auto-immunes/immunologie , Maladies auto-immunes/virologie , Bibliométrie , Chine , Virus à ARN double brin/immunologie , Hélicase IFIH1 inductrice de l'interféron/immunologie , ARN double brin/immunologie , ARN viral/immunologie , États-Unis
8.
Nature ; 617(7962): 818-826, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-37198486

RÉSUMÉ

Cancer cells rewire metabolism to favour the generation of specialized metabolites that support tumour growth and reshape the tumour microenvironment1,2. Lysine functions as a biosynthetic molecule, energy source and antioxidant3-5, but little is known about its pathological role in cancer. Here we show that glioblastoma stem cells (GSCs) reprogram lysine catabolism through the upregulation of lysine transporter SLC7A2 and crotonyl-coenzyme A (crotonyl-CoA)-producing enzyme glutaryl-CoA dehydrogenase (GCDH) with downregulation of the crotonyl-CoA hydratase enoyl-CoA hydratase short chain 1 (ECHS1), leading to accumulation of intracellular crotonyl-CoA and histone H4 lysine crotonylation. A reduction in histone lysine crotonylation by either genetic manipulation or lysine restriction impaired tumour growth. In the nucleus, GCDH interacts with the crotonyltransferase CBP to promote histone lysine crotonylation. Loss of histone lysine crotonylation promotes immunogenic cytosolic double-stranded RNA (dsRNA) and dsDNA generation through enhanced H3K27ac, which stimulates the RNA sensor MDA5 and DNA sensor cyclic GMP-AMP synthase (cGAS) to boost type I interferon signalling, leading to compromised GSC tumorigenic potential and elevated CD8+ T cell infiltration. A lysine-restricted diet synergized with MYC inhibition or anti-PD-1 therapy to slow tumour growth. Collectively, GSCs co-opt lysine uptake and degradation to shunt the production of crotonyl-CoA, remodelling the chromatin landscape to evade interferon-induced intrinsic effects on GSC maintenance and extrinsic effects on immune response.


Sujet(s)
Histone , Lysine , Tumeurs , Maturation post-traductionnelle des protéines , Chromatine/composition chimique , Chromatine/génétique , Chromatine/métabolisme , Glutaryl-CoA dehydrogenase/métabolisme , Histone/composition chimique , Histone/métabolisme , Lysine/déficit , Lysine/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , ARN double brin/immunologie , Humains , Animaux , Souris , Interféron de type I/immunologie
9.
J Immunol ; 210(3): 335-347, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36525065

RÉSUMÉ

Melanoma differentiation-associated gene 5 (MDA5), a member of the retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), has pivotal roles in innate immune responses against many positive-stranded RNA viruses, including picornavirus and coronavirus. Upon engagement with dsRNA derived from viral infection, MDA5 initiates coordinated signal transduction leading to type I IFN induction to restrict viral replication. In this study, we describe a targeted cleavage events of MDA5 by the 3C protease from Theilovirus. Upon ectopic expression of theilovirus 3C protease from Saffold virus or Theiler's murine encephalomyelitis virus but not encephalomyocarditis virus, fragments of cleaved MDA5 were observed in a dose-dependent manner. When enzymatically inactive Theilovirus 3C protease was expressed, MDA5 cleavage was completely abrogated. Mass spectrometric analysis identified two cleavage sites at the C terminus of MDA5, cleaving off one of the RNA-binding domains. The same cleavage pattern was observed during Theilovirus infection. The cleavage of MDA5 by Theilovirus protease impaired ATP hydrolysis, RNA binding, and filament assembly on RNA, resulting in dysfunction of MDA5 as an innate immune RNA sensor for IFN induction. Furthermore, the cleavage-resistant MDA5 mutant against the 3C protease showed an enhanced IFN response during Saffold virus infection, indicating that Theilovirus has a strategy to circumvent the antiviral immune response by cleaving MDA5 using 3C protease. In summary, these data suggest MDA5 cleavage by 3C protease as a novel immune evasive strategy of Theilovirus.


Sujet(s)
Hélicase IFIH1 inductrice de l'interféron , ARN double brin , Theilovirus , Animaux , Souris , Cysteine endopeptidases/génétique , Interactions hôte-pathogène , Immunité innée , Interféron de type I/métabolisme , Hélicase IFIH1 inductrice de l'interféron/génétique , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Peptide hydrolases/métabolisme , ARN double brin/immunologie , ARN double brin/métabolisme , Protéases virales 3C
10.
Proc Natl Acad Sci U S A ; 119(33): e2204235119, 2022 08 16.
Article de Anglais | MEDLINE | ID: mdl-35939694

RÉSUMÉ

Mammalian cells respond to dsRNA in multiple manners. One key response to dsRNA is the activation of PKR, an eIF2α kinase, which triggers translational arrest and the formation of stress granules. However, the process of PKR activation in cells is not fully understood. In response to increased endogenous or exogenous dsRNA, we observed that PKR forms novel cytosolic condensates, referred to as dsRNA-induced foci (dRIFs). dRIFs contain dsRNA, form in proportion to dsRNA, and are enhanced by longer dsRNAs. dRIFs enrich several other dsRNA-binding proteins, including ADAR1, Stau1, NLRP1, and PACT. Strikingly, dRIFs correlate with and form before translation repression by PKR and localize to regions of cells where PKR activation is initiated. We hypothesize that dRIF formation is a mechanism that cells use to enhance the sensitivity of PKR activation in response to low levels of dsRNA or to overcome viral inhibitors of PKR activation.


Sujet(s)
ARN double brin , ARN viral , Maladies virales , eIF-2 Kinase , Activation enzymatique , Humains , Immunité innée , Phosphorylation , Biosynthèse des protéines , ARN double brin/composition chimique , ARN double brin/immunologie , ARN viral/composition chimique , ARN viral/immunologie , Protéines de liaison à l'ARN/composition chimique , Granules de stress , Maladies virales/enzymologie , Maladies virales/immunologie , eIF-2 Kinase/composition chimique
11.
Nature ; 606(7914): 594-602, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35614224

RÉSUMÉ

Only a small proportion of patients with cancer show lasting responses to immune checkpoint blockade (ICB)-based monotherapies. The RNA-editing enzyme ADAR1 is an emerging determinant of resistance to ICB therapy and prevents ICB responsiveness by repressing immunogenic double-stranded RNAs (dsRNAs), such as those arising from the dysregulated expression of endogenous retroviral elements (EREs)1-4. These dsRNAs trigger an interferon-dependent antitumour response by activating A-form dsRNA (A-RNA)-sensing proteins such as MDA-5 and PKR5. Here we show that ADAR1 also prevents the accrual of endogenous Z-form dsRNA elements (Z-RNAs), which were enriched in the 3' untranslated regions of interferon-stimulated mRNAs. Depletion or mutation of ADAR1 resulted in Z-RNA accumulation and activation of the Z-RNA sensor ZBP1, which culminated in RIPK3-mediated necroptosis. As no clinically viable ADAR1 inhibitors currently exist, we searched for a compound that can override the requirement for ADAR1 inhibition and directly activate ZBP1. We identified a small molecule, the curaxin CBL0137, which potently activates ZBP1 by triggering Z-DNA formation in cells. CBL0137 induced ZBP1-dependent necroptosis in cancer-associated fibroblasts and reversed ICB unresponsiveness in mouse models of melanoma. Collectively, these results demonstrate that ADAR1 represses endogenous Z-RNAs and identifies ZBP1-mediated necroptosis as a new determinant of tumour immunogenicity masked by ADAR1. Therapeutic activation of ZBP1-induced necroptosis provides a readily translatable avenue for rekindling the immune responsiveness of ICB-resistant human cancers.


Sujet(s)
Adenosine deaminase , Nécroptose , Tumeurs , Protéines de liaison à l'ARN , Régions 3' non traduites , Adenosine deaminase/métabolisme , Animaux , Fibroblastes associés au cancer , Carbazoles/pharmacologie , Humains , Immunothérapie/tendances , Interférons/métabolisme , Mélanome , Souris , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , ARN double brin/immunologie , Protéines de liaison à l'ARN/métabolisme
12.
Front Immunol ; 13: 818023, 2022.
Article de Anglais | MEDLINE | ID: mdl-35126398

RÉSUMÉ

Alu retrotransposons belong to the class of short interspersed nuclear elements (SINEs). Alu RNA is abundant in cells and its repetitive structure forms double-stranded RNAs (dsRNA) that activate dsRNA sensors and trigger innate immune responses with significant pathological consequences. Mechanisms to prevent innate immune activation include deamination of adenosines to inosines in dsRNAs, referred to as A-to-I editing, degradation of Alu RNAs by endoribonucleases, and sequestration of Alu RNAs by RNA binding proteins. We have previously demonstrated that widespread loss of Alu RNA A-to-I editing is associated with diverse human diseases including viral (COVID-19, influenza) and autoimmune diseases (multiple sclerosis). Here we demonstrate loss of A-to-I editing in leukocytes is also associated with inflammatory bowel diseases. Our structure-function analysis demonstrates that ability to activate innate immune responses resides in the left arm of Alu RNA, requires a 5'-PPP, RIG-I is the major Alu dsRNA sensor, and A-to-I editing disrupts both structure and function. Further, edited Alu RNAs inhibit activity of unedited Alu RNAs. Altering Alu RNA nucleotide sequence increases biological activity. Two classes of Alu RNAs exist, one class stimulates both IRF and NF-kB transcriptional activity and a second class only stimulates IRF transcriptional activity. Thus, Alu RNAs play important roles in human disease but may also have therapeutic potential.


Sujet(s)
Séquences Alu/génétique , Séquences Alu/immunologie , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/immunologie , Adénosine , COVID-19 , Humains , Inosine , ARN double brin/génétique , ARN double brin/immunologie , SARS-CoV-2
13.
Fish Shellfish Immunol ; 121: 215-222, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-34999226

RÉSUMÉ

Salmonids are one of the most farmed fish species worldwide. These aquatic vertebrates rely heavily on their innate immune responses as the first line of defense to defend themselves against invading pathogens. Although commercial vaccines are available against some viral and bacterial pathogens affecting salmonids, their protective efficacy varies. Using a prophylactic inducer of local and systemic innate immune responses to limit infection could have significant implications in salmonid aquaculture. A potent inducer of innate immune responses in fish is double-stranded RNA (dsRNA), a molecule that all viruses make during their replicative cycle. Polyinosinic: polycytidylic acid (polyI:C) is a synthetic dsRNA commonly used to induce type I interferons (IFNs), interferon stimulated genes (ISGs) as well as an antiviral state in vertebrate species. Based on in vitro data it was hypothesized that both local and systemic innate immune responses, in salmonids, would be enhanced by orally delivering high molecular weight polyI:C (HMW polyI:C) using cationic phytoglycogen nanoparticles (NPs) as a delivery method. The present study investigates this hypothesis using two feed delivery methods. In the first in vivo study, to ensure an equal distribution of dose, individual rainbow trout (Oncorhynchus mykiss) were orally gavaged with feed moistened with a solution containing HMW-NP (polyI:C complexed with cationic phytoglycogen nanoparticles) or HMW polyI:C alone. In a second in vivo experiment, to better mimic a more realistic feeding scenario, rainbow trout were fed feed pellets to which HMW, or HMW-NP was added. The expression of IFN1 and ISGs (vig-3, Mx1) were quantified using real-time PCR in the intestine (local response) and head kidney (systemic response). The results of these studies indicate that HMW-NP induced a higher level of IFN1 and ISG expression in the intestine and head kidney compared to the HMW fed fish. The results of this study could lead to new advances in therapeutics for the aquaculture industry by utilizing the innate immune response against invading pathogens using an orally delivered stimulant.


Sujet(s)
Immunité innée , Interféron de type I , Nanoparticules , Oncorhynchus mykiss , ARN double brin/immunologie , Animaux , Maladies des poissons/prévention et contrôle , Interféron de type I/immunologie , Oncorhynchus mykiss/immunologie
14.
PLoS Pathog ; 18(1): e1010250, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-35041722

RÉSUMÉ

ADARs (adenosine deaminases acting on RNA) are known for their adenosine-to-inosine RNA editing activity, and most recently, for their role in preventing aberrant dsRNA-response by activation of dsRNA sensors (i.e., RIG-I-like receptor homologs). However, it is still unclear whether suppressing spurious dsRNA-response represents the ancestral role of ADARs in bilaterians. As a first step to address this question, we identified ADAR1 and ADAR2 homologs in the planarian Schmidtea mediterranea, which is evolutionarily distant from canonical lab models (e.g., flies and nematodes). Our results indicate that knockdown of either planarian adar1 or adar2 by RNA interference (RNAi) resulted in upregulation of dsRNA-response genes, including three planarian rig-I-like receptor (prlr) homologs. Furthermore, independent knockdown of adar1 and adar2 reduced the number of infected cells with a dsRNA virus, suggesting they suppress a bona fide anti-viral dsRNA-response activity. Knockdown of adar1 also resulted in lesion formation and animal lethality, thus attesting to its essentiality. Simultaneous knockdown of adar1 and prlr1 rescued adar1(RNAi)-dependent animal lethality and rescued the dsRNA-response, suggesting that it contributes to the deleterious effect of adar1 knockdown. Finally, we found that ADAR2, but not ADAR1, mediates mRNA editing in planarians, suggesting at least in part non-redundant activities for planarians ADARs. Our results underline the essential role of ADARs in suppressing activation of harmful dsRNA-response in planarians, thus supporting it as their ancestral role in bilaterians. Our work also set the stage to study further and better understand the regulatory mechanisms governing anti-viral dsRNA-responses from an evolutionary standpoint using planarians as a model.


Sujet(s)
Adenosine deaminase/immunologie , Adenosine deaminase/métabolisme , Planaires/physiologie , ARN double brin/immunologie , Animaux , Maladies virales/immunologie
15.
Signal Transduct Target Ther ; 7(1): 22, 2022 01 24.
Article de Anglais | MEDLINE | ID: mdl-35075101

RÉSUMÉ

As a highly pathogenic human coronavirus, SARS-CoV-2 has to counteract an intricate network of antiviral host responses to establish infection and spread. The nucleic acid-induced stress response is an essential component of antiviral defense and is closely related to antiviral innate immunity. However, whether SARS-CoV-2 regulates the stress response pathway to achieve immune evasion remains elusive. In this study, SARS-CoV-2 NSP5 and N protein were found to attenuate antiviral stress granule (avSG) formation. Moreover, NSP5 and N suppressed IFN expression induced by infection of Sendai virus or transfection of a synthetic mimic of dsRNA, poly (I:C), inhibiting TBK1 and IRF3 phosphorylation, and restraining the nuclear translocalization of IRF3. Furthermore, HEK293T cells with ectopic expression of NSP5 or N protein were less resistant to vesicular stomatitis virus infection. Mechanistically, NSP5 suppressed avSG formation and disrupted RIG-I-MAVS complex to attenuate the RIG-I-mediated antiviral immunity. In contrast to the multiple targets of NSP5, the N protein specifically targeted cofactors upstream of RIG-I. The N protein interacted with G3BP1 to prevent avSG formation and to keep the cofactors G3BP1 and PACT from activating RIG-I. Additionally, the N protein also affected the recognition of dsRNA by RIG-I. This study revealed the intimate correlation between SARS-CoV-2, the stress response, and innate antiviral immunity, shedding light on the pathogenic mechanism of COVID-19.


Sujet(s)
Protéases 3C des coronavirus/génétique , Protéines de la nucléocapside des coronavirus/génétique , Protéine-58 à domaine DEAD/génétique , Helicase/génétique , Protéines liant le poly-adp-ribose/génétique , RNA helicases/génétique , Protéines à motif de reconnaissance de l'ARN/génétique , Protéines de liaison à l'ARN/génétique , Récepteurs immunologiques/génétique , SARS-CoV-2/génétique , Granules de stress/génétique , Animaux , Chlorocebus aethiops , Protéases 3C des coronavirus/immunologie , Protéines de la nucléocapside des coronavirus/immunologie , Protéine-58 à domaine DEAD/immunologie , Helicase/immunologie , Régulation de l'expression des gènes , Cellules HEK293 , Cellules HeLa , Humains , Échappement immunitaire , Phosphoprotéines/génétique , Phosphoprotéines/immunologie , Poly I-C/pharmacologie , Protéines liant le poly-adp-ribose/immunologie , Liaison aux protéines , RNA helicases/immunologie , Protéines à motif de reconnaissance de l'ARN/immunologie , ARN double brin/génétique , ARN double brin/immunologie , Protéines de liaison à l'ARN/immunologie , Récepteurs immunologiques/immunologie , SARS-CoV-2/immunologie , SARS-CoV-2/pathogénicité , Virus Sendai/génétique , Virus Sendai/immunologie , Transduction du signal , Granules de stress/effets des médicaments et des substances chimiques , Granules de stress/immunologie , Granules de stress/virologie , Cellules Vero , Vesiculovirus/génétique , Vesiculovirus/immunologie
16.
Viruses ; 14(1)2022 01 05.
Article de Anglais | MEDLINE | ID: mdl-35062293

RÉSUMÉ

The physiologic function of tripartite motif protein 56 (TRIM56), a ubiquitously expressed E3 ligase classified within the large TRIM protein family, remains elusive. Gene knockdown studies have suggested TRIM56 as a positive regulator of the type I interferon (IFN-I) antiviral response elicited via the Toll-like receptor 3 (TLR3) and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathways, which detect and respond to danger signals-extracellular double-stranded (ds) RNA and cytosolic dsDNA, respectively. However, to what extent these pathways depend on TRIM56 in human cells is unclear. In addition, it is debatable whether TRIM56 plays a part in controlling the expression of IFN-stimulated genes (ISGs) resulting from IFN-I based antiviral treatment. In this study, we created HeLa-derived TRIM56 null cell lines by gene editing and used these cell models to comprehensively examine the impact of endogenous TRIM56 on innate antiviral responses. Our results showed that TRIM56 knockout severely undermined the upregulation of ISGs by extracellular dsRNA and that loss of TRIM56 weakened the response to cytosolic dsDNA. ISG induction and ISGylation following IFN-α stimulation, however, were not compromised by TRIM56 deletion. Using a vesicular stomatitis virus-based antiviral bioactivity assay, we demonstrated that IFN-α could efficiently establish an antiviral state in TRIM56 null cells, providing direct evidence that TRIM56 is not required for the general antiviral action of IFN-I. Altogether, these data ascertain the contributions of TRIM56 to TLR3- and cGAS-STING-dependent antiviral pathways in HeLa cells and add to our understanding of the roles this protein plays in innate immunity.


Sujet(s)
ADN/immunologie , Interféron alpha/immunologie , ARN double brin/immunologie , Protéines à motif tripartite/métabolisme , Ubiquitin-protein ligases/métabolisme , Virus/immunologie , Animaux , Chlorocebus aethiops , Cytosol/métabolisme , Cellules HeLa , Humains , Immunité innée , Protéines membranaires/métabolisme , Nucleotidyltransferases/métabolisme , Récepteur de type Toll-3/métabolisme , Protéines à motif tripartite/génétique , Ubiquitin-protein ligases/génétique , Cellules Vero , Vesiculovirus/immunologie
17.
Nucleic Acids Res ; 49(18): 10604-10617, 2021 10 11.
Article de Anglais | MEDLINE | ID: mdl-34520542

RÉSUMÉ

RNA hydrolysis presents problems in manufacturing, long-term storage, world-wide delivery and in vivo stability of messenger RNA (mRNA)-based vaccines and therapeutics. A largely unexplored strategy to reduce mRNA hydrolysis is to redesign RNAs to form double-stranded regions, which are protected from in-line cleavage and enzymatic degradation, while coding for the same proteins. The amount of stabilization that this strategy can deliver and the most effective algorithmic approach to achieve stabilization remain poorly understood. Here, we present simple calculations for estimating RNA stability against hydrolysis, and a model that links the average unpaired probability of an mRNA, or AUP, to its overall hydrolysis rate. To characterize the stabilization achievable through structure design, we compare AUP optimization by conventional mRNA design methods to results from more computationally sophisticated algorithms and crowdsourcing through the OpenVaccine challenge on the Eterna platform. We find that rational design on Eterna and the more sophisticated algorithms lead to constructs with low AUP, which we term 'superfolder' mRNAs. These designs exhibit a wide diversity of sequence and structure features that may be desirable for translation, biophysical size, and immunogenicity. Furthermore, their folding is robust to temperature, computer modeling method, choice of flanking untranslated regions, and changes in target protein sequence, as illustrated by rapid redesign of superfolder mRNAs for B.1.351, P.1 and B.1.1.7 variants of the prefusion-stabilized SARS-CoV-2 spike protein. Increases in in vitro mRNA half-life by at least two-fold appear immediately achievable.


Sujet(s)
Algorithmes , ARN double brin/composition chimique , ARN messager/composition chimique , ARN viral/composition chimique , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique , Appariement de bases , Séquence nucléotidique , COVID-19/prévention et contrôle , Humains , Hydrolyse , Stabilité de l'ARN , ARN double brin/génétique , ARN double brin/immunologie , ARN messager/génétique , ARN messager/immunologie , ARN viral/génétique , ARN viral/immunologie , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Thermodynamique
18.
J Gen Virol ; 102(7)2021 07.
Article de Anglais | MEDLINE | ID: mdl-34236957

RÉSUMÉ

Mosquito-borne flaviviruses are significant contributors to the arboviral disease burdens both in Australia and globally. While routine arbovirus surveillance remains a vital exercise to identify known flaviviruses in mosquito populations, novel or divergent and emerging species can be missed by these traditional methods. The MAVRIC (monoclonal antibodies to viral RNA intermediates in cells) system is an ELISA-based method for broad-spectrum isolation of positive-sense and double-stranded RNA (dsRNA) viruses based on detection of dsRNA in infected cells. While the MAVRIC ELISA has successfully been used to detect known and novel flaviviruses in Australian mosquitoes, we previously reported that dsRNA could not be detected in dengue virus-infected cells using this method. In this study we identified additional flaviviruses which evade detection of dsRNA by the MAVRIC ELISA. Utilising chimeric flaviviruses we demonstrated that this outcome may be dictated by the non-structural proteins and/or untranslated regions of the flaviviral genome. In addition, we report a modified fixation method that enables improved detection of flavivirus dsRNA and inactivation of non-enveloped viruses from mosquito populations using the MAVRIC system. This study demonstrates the utility of anti-dsRNA monoclonal antibodies for identifying viral replication in insect and vertebrate cell systems and highlights a unique characteristic of flavivirus replication.


Sujet(s)
Culicidae/virologie , Flavivirus/isolement et purification , Flavivirus/physiologie , ARN double brin/analyse , ARN viral/analyse , Aedes/virologie , Animaux , Anticorps monoclonaux , Australie , Lignée cellulaire , Virus de la dengue/génétique , Virus de la dengue/isolement et purification , Virus de la dengue/physiologie , Test ELISA , Flavivirus/génétique , ARN double brin/immunologie , ARN viral/immunologie , Protéines de l'enveloppe virale/analyse , Protéines de l'enveloppe virale/métabolisme , Protéines virales non structurales/analyse , Protéines virales non structurales/métabolisme , Réplication virale
19.
Nature ; 597(7874): 109-113, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34261127

RÉSUMÉ

Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that produces the second messenger cG[2'-5']pA[3'-5']p (2'3'-cGAMP) and controls activation of innate immunity in mammalian cells1-5. Animal genomes typically encode multiple proteins with predicted homology to cGAS6-10, but the function of these uncharacterized enzymes is unknown. Here we show that cGAS-like receptors (cGLRs) are innate immune sensors that are capable of recognizing divergent molecular patterns and catalysing synthesis of distinct nucleotide second messenger signals. Crystal structures of human and insect cGLRs reveal a nucleotidyltransferase signalling core shared with cGAS and a diversified primary ligand-binding surface modified with notable insertions and deletions. We demonstrate that surface remodelling of cGLRs enables altered ligand specificity and used a forward biochemical screen to identify cGLR1 as a double-stranded RNA sensor in the model organism Drosophila melanogaster. We show that RNA recognition activates Drosophila cGLR1 to synthesize the novel product cG[3'-5']pA[2'-5']p (3'2'-cGAMP). A crystal structure of Drosophila stimulator of interferon genes (dSTING) in complex with 3'2'-cGAMP explains selective isomer recognition, and 3'2'-cGAMP induces an enhanced antiviral state in vivo that protects from viral infection. Similar to radiation of Toll-like receptors in pathogen immunity, our results establish cGLRs as a diverse family of metazoan pattern recognition receptors.


Sujet(s)
Drosophila melanogaster/métabolisme , Nucléotides cycliques/métabolisme , Nucleotidyltransferases/métabolisme , ARN double brin/métabolisme , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , Systèmes de seconds messagers , Séquence d'acides aminés , Animaux , Cristallographie aux rayons X , Protéines de Drosophila/composition chimique , Protéines de Drosophila/métabolisme , Drosophila melanogaster/immunologie , Drosophila melanogaster/virologie , Femelle , Humains , Immunité innée , Mâle , Protéines membranaires/composition chimique , Protéines membranaires/métabolisme , Modèles moléculaires , Nucleotidyltransferases/composition chimique , Nucleotidyltransferases/immunologie , ARN double brin/analyse , ARN double brin/immunologie , Récepteurs de reconnaissance de motifs moléculaires/composition chimique , Récepteurs de reconnaissance de motifs moléculaires/immunologie , Virus/immunologie
20.
Nature ; 597(7874): 114-118, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34261128

RÉSUMÉ

In mammals, cyclic GMP-AMP (cGAMP) synthase (cGAS) produces the cyclic dinucleotide 2'3'-cGAMP in response to cytosolic DNA and this triggers an antiviral immune response. cGAS belongs to a large family of cGAS/DncV-like nucleotidyltransferases that is present in both prokaryotes1 and eukaryotes2-5. In bacteria, these enzymes synthesize a range of cyclic oligonucleotides and have recently emerged as important regulators of phage infections6-8. Here we identify two cGAS-like receptors (cGLRs) in the insect Drosophila melanogaster. We show that cGLR1 and cGLR2 activate Sting- and NF-κB-dependent antiviral immunity in response to infection with RNA or DNA viruses. cGLR1 is activated by double-stranded RNA to produce the cyclic dinucleotide 3'2'-cGAMP, whereas cGLR2 produces a combination of 2'3'-cGAMP and 3'2'-cGAMP in response to an as-yet-unidentified stimulus. Our data establish cGAS as the founding member of a family of receptors that sense different types of nucleic acids and trigger immunity through the production of cyclic dinucleotides beyond 2'3'-cGAMP.


Sujet(s)
Drosophila melanogaster/immunologie , Nucleotidyltransferases/immunologie , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , Virus/immunologie , Séquence d'acides aminés , Animaux , Lignée cellulaire , Protéines de Drosophila/métabolisme , Drosophila melanogaster/métabolisme , Drosophila melanogaster/virologie , Femelle , Humains , Immunité innée/génétique , Immunité innée/immunologie , Ligands , Mâle , Protéines membranaires/métabolisme , Modèles moléculaires , Facteur de transcription NF-kappa B/métabolisme , Nucléotides cycliques/métabolisme , Nucleotidyltransferases/classification , Nucleotidyltransferases/déficit , Nucleotidyltransferases/métabolisme , ARN double brin/analyse , ARN double brin/immunologie , ARN double brin/métabolisme , Récepteurs de reconnaissance de motifs moléculaires/classification , Récepteurs de reconnaissance de motifs moléculaires/déficit , Récepteurs de reconnaissance de motifs moléculaires/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE