Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.502
Filtrer
1.
Cancer Lett ; 595: 216999, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-38823762

RÉSUMÉ

Tumor protein p63 isoform ΔNp63 plays roles in the squamous epithelium and squamous cell carcinomas (SCCs), including esophageal SCC (ESCC). By integrating data from cell lines and our latest patient-derived organoid cultures, derived xenograft models, and clinical sample transcriptomic analyses, we identified a novel and robust oncogenic role of ΔNp63 in ESCC. We showed that ΔNp63 maintains the repression of cancer cell endogenous retrotransposon expression and cellular double-stranded RNA sensing. These subsequently lead to a restricted cancer cell viral mimicry response and suppressed induction of tumor-suppressive type I interferon (IFN-I) signaling through the regulations of Signal transducer and activator of transcription 1, Interferon regulatory factor 1, and cGAS-STING pathway. The cancer cell ΔNp63/IFN-I signaling axis affects both the cancer cell and tumor-infiltrating immune cell (TIIC) compartments. In cancer cells, depletion of ΔNp63 resulted in reduced cell viability. ΔNp63 expression is negatively associated with the anticancer responses to viral mimicry booster treatments targeting cancer cells. In the tumor microenvironment, cancer cell TP63 expression negatively correlates with multiple TIIC signatures in ESCC clinical samples. ΔNp63 depletion leads to increased cancer cell antigen presentation molecule expression and enhanced recruitment and reprogramming of tumor-infiltrating myeloid cells. Similar IFN-I signaling and TIIC signature association with ΔNp63 were also observed in lung SCC. These results support the potential application of ΔNp63 as a therapeutic target and a biomarker to guide candidate anticancer treatments exploring viral mimicry responses.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Facteurs de transcription , Microenvironnement tumoral , Protéines suppresseurs de tumeurs , Humains , Microenvironnement tumoral/immunologie , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/immunologie , Carcinome épidermoïde de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/virologie , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/immunologie , Tumeurs de l'oesophage/virologie , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Lignée cellulaire tumorale , Animaux , Survie cellulaire , Régulation de l'expression des gènes tumoraux , Souris , Transduction du signal , Interféron de type I/métabolisme
2.
Nat Immunol ; 25(6): 969-980, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38831104

RÉSUMÉ

Rare genetic variants in toll-like receptor 7 (TLR7) are known to cause lupus in humans and mice. UNC93B1 is a transmembrane protein that regulates TLR7 localization into endosomes. In the present study, we identify two new variants in UNC93B1 (T314A, located proximally to the TLR7 transmembrane domain, and V117L) in a cohort of east Asian patients with childhood-onset systemic lupus erythematosus. The V117L variant was associated with increased expression of type I interferons and NF-κB-dependent cytokines in patient plasma and immortalized B cells. THP-1 cells expressing the variant UNC93B1 alleles exhibited exaggerated responses to stimulation of TLR7/-8, but not TLR3 or TLR9, which could be inhibited by targeting the downstream signaling molecules, IRAK1/-4. Heterozygous mice expressing the orthologous Unc93b1V117L variant developed a spontaneous lupus-like disease that was more severe in homozygotes and again hyperresponsive to TLR7 stimulation. Together, this work formally identifies genetic variants in UNC93B1 that can predispose to childhood-onset systemic lupus erythematosus.


Sujet(s)
Prédisposition génétique à une maladie , Lupus érythémateux disséminé , Récepteur de type Toll-7 , Lupus érythémateux disséminé/génétique , Humains , Animaux , Récepteur de type Toll-7/génétique , Récepteur de type Toll-7/métabolisme , Souris , Enfant , Femelle , Protéines de transport membranaire/génétique , Protéines de transport membranaire/métabolisme , Mâle , Âge de début , Variation génétique , Facteur de transcription NF-kappa B/métabolisme , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Adolescent , Cellules THP-1 , Interféron de type I/métabolisme
3.
Phytomedicine ; 130: 155373, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38850630

RÉSUMÉ

BACKGROUND: Acute respiratory distress syndrome (ARDS) is an acute respiratory disease characterized by bilateral chest radiolucency and severe hypoxemia. Quzhou Fructus Aurantii ethyl acetate extract (QFAEE), which is prepared from the traditional Chinese respiratory anti-inflammatory natural herb Quzhou Fructus Arantii, has the potential to alleviate ARDS. In this work, we aimed to investigate the potential and mechanism underlying the action of QFAEE on ARDS and how QFAEE modulates the STING pathway to reduce type I interferon release to alleviate the inflammatory response. METHODS: Lipopolysaccharide (LPS), a potential proinflammatory stimulant capable of causing pulmonary inflammation with edema after nasal drops, was employed to model ARDS in vitro and in vivo. Under QFAEE intervention, the mechanism of action of QFAEE to alleviate ARDS was explored in this study. TREX1-/- mice were sued as a research model for the activation of the congenital STING signaling pathway. The effect of QFAEE on TREX1-/- mice could explain the STING-targeted effect of QFAEE on alleviating the inflammatory response. Our explorations covered several techniques, Western blot, histological assays, immunofluorescence staining, transcriptomic assays and qRT-PCR to determine the potential mechanism of action of QFAEE in antagonizing the inflammatory response in the lungs, as well as the mechanism of action of QFAEE in targeting the STING signaling pathway to regulate the release of type I interferon. RESULTS: QFAEE effectively alleviates ARDS symptoms in LPS-induced ARDS. We revealed that the mechanism underlying LPS-induced ARDS is the STING-TBK1 signaling pathway and further elucidated the molecular mechanism of QFAEE in the prevention and treatment of ARDS. QFAEE reduced the release of type I interferons by inhibiting the STING-TBK1-IRF3 axis, thus alleviating LPS-induced pneumonia and lung cell death in mice. Another key finding is that activation of the STING pathway by activators or targeted knockdown of the TREX1 gene can also induce ARDS. As expected, QFAEE was found to be an effective protective agent in alleviating ARDS and the antagonistic effect of QFAEE on ARDS was achieved by inhibiting the STING signaling pathway. CONCLUSIONS: The main anti-inflammatory effect of QFAEE was achieved by inhibiting the STING signaling pathway and reducing the release of type I interferons. According to this mechanism of effect, QFAEE can effectively alleviate ARDS and can be considered a potential therapeutic agent. In addition, the STING pathway plays an essential role in the development and progression of ARDS, and it is a potential target for ARDS therapy.


Sujet(s)
Anti-inflammatoires , Interféron de type I , Lipopolysaccharides , Protéines membranaires , , Animaux , Interféron de type I/métabolisme , Souris , Anti-inflammatoires/pharmacologie , Protéines membranaires/métabolisme , /traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Mâle , Humains , Souris de lignée C57BL , Médicaments issus de plantes chinoises/pharmacologie , Extraits de plantes/pharmacologie , Pneumopathie infectieuse/traitement médicamenteux , Pneumopathie infectieuse/induit chimiquement
4.
Cell Death Dis ; 15(6): 403, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858387

RÉSUMÉ

Necroptosis is an inflammatory form of cell suicide that critically depends on the kinase activity of Receptor Interacting Protein Kinase 3 (RIPK3). Previous studies showed that immunization with necroptotic cells conferred protection against subsequent tumor challenge. Since RIPK3 can also promote apoptosis and NF-κB-dependent inflammation, it remains difficult to determine the contribution of necroptosis-associated release of damage-associated molecular patterns (DAMPs) in anti-tumor immunity. Here, we describe a system that allows us to selectively induce RIPK3-dependent necroptosis or apoptosis with minimal NF-κB-dependent inflammatory cytokine expression. In a syngeneic tumor challenge model, immunization with necroptotic cells conferred superior protection against subsequent tumor challenge. Surprisingly, this protective effect required CD4+ T cells rather than CD8+ T cells and is dependent on host type I interferon signaling. Our results provide evidence that death-dependent type I interferon production following necroptosis is sufficient to elicit protective anti-tumor immunity.


Sujet(s)
Nécroptose , Receptor-Interacting Protein Serine-Threonine Kinases , Nécroptose/immunologie , Animaux , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Souris , Souris de lignée C57BL , Interféron de type I/métabolisme , Lymphocytes T CD8+/immunologie , Transduction du signal , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Tumeurs/immunologie , Tumeurs/anatomopathologie , Humains , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques
5.
J Exp Med ; 221(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38869480

RÉSUMÉ

While conventional wisdom initially postulated that PD-L1 serves as the inert ligand for PD-1, an emerging body of literature suggests that PD-L1 has cell-intrinsic functions in immune and cancer cells. In line with these studies, here we show that engagement of PD-L1 via cellular ligands or agonistic antibodies, including those used in the clinic, potently inhibits the type I interferon pathway in cancer cells. Hampered type I interferon responses in PD-L1-expressing cancer cells resulted in enhanced efficacy of oncolytic viruses in vitro and in vivo. Consistently, PD-L1 expression marked tumor explants from cancer patients that were best infected by oncolytic viruses. Mechanistically, PD-L1 promoted a metabolic shift characterized by enhanced glycolysis rate that resulted in increased lactate production. In turn, lactate inhibited type I IFN responses. In addition to adding mechanistic insight into PD-L1 intrinsic function, our results will also help guide the numerous ongoing efforts to combine PD-L1 antibodies with oncolytic virotherapy in clinical trials.


Sujet(s)
Antigène CD274 , Interféron de type I , Thérapie virale de cancers , Virus oncolytiques , Antigène CD274/métabolisme , Antigène CD274/immunologie , Antigène CD274/génétique , Humains , Interféron de type I/métabolisme , Interféron de type I/immunologie , Virus oncolytiques/physiologie , Animaux , Thérapie virale de cancers/méthodes , Lignée cellulaire tumorale , Souris , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/métabolisme , Glycolyse , Transduction du signal , Acide lactique/métabolisme , Femelle
6.
PLoS Pathog ; 20(6): e1012287, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38843304

RÉSUMÉ

The kinetics of type I interferon (IFN) induction versus the virus replication compete, and the result of the competition determines the outcome of the infection. Chaperone proteins that involved in promoting the activation kinetics of PRRs rapidly trigger antiviral innate immunity. We have previously shown that prior to the interaction with MAVS to induce type I IFN, 14-3-3η facilitates the oligomerization and intracellular redistribution of activated MDA5. Here we report that the cleavage of 14-3-3η upon MDA5 activation, and we identified Caspase-3 activated by MDA5-dependent signaling was essential to produce sub-14-3-3η lacking the C-terminal helix (αI) and tail. The cleaved form of 14-3-3η (sub-14-3-3η) could strongly interact with MDA5 but could not support MDA5-dependent type I IFN induction, indicating the opposite functions between the full-length 14-3-3η and sub-14-3-3η. During human coronavirus or enterovirus infections, the accumulation of sub-14-3-3η was observed along with the activation of Caspase-3, suggesting that RNA viruses may antagonize 14-3-3η by promoting the formation of sub-14-3-3η to impair antiviral innate immunity. In conclusion, sub-14-3-3η, which could not promote MDA5 activation, may serve as a negative feedback to return to homeostasis to prevent excessive type I IFN production and unnecessary inflammation.


Sujet(s)
Protéines 14-3-3 , Caspase-3 , Hélicase IFIH1 inductrice de l'interféron , Protéines 14-3-3/métabolisme , Humains , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Hélicase IFIH1 inductrice de l'interféron/génétique , Caspase-3/métabolisme , Immunité innée , Cellules HEK293 , Animaux , Transduction du signal , Interféron de type I/métabolisme
7.
Nat Commun ; 15(1): 4772, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858384

RÉSUMÉ

The underlying mechanisms of atherosclerosis, the second leading cause of death among Werner syndrome (WS) patients, are not fully understood. Here, we establish an in vitro co-culture system using macrophages (iMφs), vascular endothelial cells (iVECs), and vascular smooth muscle cells (iVSMCs) derived from induced pluripotent stem cells. In co-culture, WS-iMφs induces endothelial dysfunction in WS-iVECs and characteristics of the synthetic phenotype in WS-iVSMCs. Transcriptomics and open chromatin analysis reveal accelerated activation of type I interferon signaling and reduced chromatin accessibility of several transcriptional binding sites required for cellular homeostasis in WS-iMφs. Furthermore, the H3K9me3 levels show an inverse correlation with retrotransposable elements, and retrotransposable element-derived double-stranded RNA activates the DExH-box helicase 58 (DHX58)-dependent cytoplasmic RNA sensing pathway in WS-iMφs. Conversely, silencing type I interferon signaling in WS-iMφs rescues cell proliferation and suppresses cellular senescence and inflammation. These findings suggest that Mφ-specific inhibition of type I interferon signaling could be targeted to treat atherosclerosis in WS patients.


Sujet(s)
Athérosclérose , Inflammation , Interféron de type I , Macrophages , Rétroéléments , Syndrome de Werner , Interféron de type I/métabolisme , Syndrome de Werner/génétique , Syndrome de Werner/métabolisme , Humains , Athérosclérose/métabolisme , Athérosclérose/immunologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Macrophages/métabolisme , Macrophages/immunologie , Rétroéléments/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Cellules souches pluripotentes induites/métabolisme , Transduction du signal , Techniques de coculture , Myocytes du muscle lisse/métabolisme , Cellules endothéliales/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Vieillissement de la cellule , Prolifération cellulaire
8.
Mol Neurodegener ; 19(1): 48, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38886816

RÉSUMÉ

BACKGROUND: Aging significantly elevates the risk of developing neurodegenerative diseases. Neuroinflammation is a universal hallmark of neurodegeneration as well as normal brain aging. Which branches of age-related neuroinflammation, and how they precondition the brain toward pathological progression, remain ill-understood. The presence of elevated type I interferon (IFN-I) has been documented in the aged brain, but its role in promoting degenerative processes, such as the loss of neurons in vulnerable regions, has not been studied in depth. METHODS: To comprehend the scope of IFN-I activity in the aging brain, we surveyed IFN-I-responsive reporter mice at multiple ages. We also examined 5- and 24-month-old mice harboring selective ablation of Ifnar1 in microglia to observe the effects of manipulating this pathway during the aging process using bulk RNA sequencing and histological parameters. RESULTS: We detected age-dependent IFN-I signal escalation in multiple brain cell types from various regions, especially in microglia. Selective ablation of Ifnar1 from microglia in aged mice significantly reduced overall brain IFN-I signature, dampened microglial reactivity, lessened neuronal loss, restored expression of key neuronal genes and pathways, and diminished the accumulation of lipofuscin, a core hallmark of cellular aging in the brain. CONCLUSIONS: Overall, our study demonstrates pervasive IFN-I activity during normal mouse brain aging and reveals a pathogenic, pro-degenerative role played by microglial IFN-I signaling in perpetuating neuroinflammation, neuronal dysfunction, and molecular aggregation. These findings extend the understanding of a principal axis of age-related inflammation in the brain, one likely shared with multiple neurological disorders, and provide a rationale to modulate aberrant immune activation to mitigate neurodegenerative process at all stages.


Sujet(s)
Vieillissement , Encéphale , Interféron de type I , Microglie , Transduction du signal , Animaux , Vieillissement/métabolisme , Interféron de type I/métabolisme , Souris , Encéphale/métabolisme , Encéphale/anatomopathologie , Transduction du signal/physiologie , Microglie/métabolisme , Récepteur à l'interféron alpha-bêta/métabolisme , Neurones/métabolisme
9.
Vet Res ; 55(1): 79, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886840

RÉSUMÉ

Porcine deltacoronavirus (PDCoV) is an enteropathogenic coronavirus that has been reported to use various strategies to counter the host antiviral innate immune response. The cGAS-STING signalling pathway plays an important role in antiviral innate immunity. However, it remains unclear whether PDCoV achieves immune evasion by regulating the cGAS-STING pathway. Here, we demonstrated that the nonstructural protein 2 (nsp2) encoded by PDCoV inhibits cGAS-STING-mediated type I and III interferon (IFN) responses via the regulation of porcine STING (pSTING) stability. Mechanistically, ectopically expressed PDCoV nsp2 was found to interact with the N-terminal region of pSTING. Consequently, pSTING was degraded through K48-linked ubiquitination and the proteasomal pathway, leading to the disruption of cGAS-STING signalling. Furthermore, K150 and K236 of pSTING were identified as crucial residues for nsp2-mediated ubiquitination and degradation. In summary, our findings provide a basis for elucidating the immune evasion mechanism of PDCoV and will contribute to the development of targets for anti-coronavirus drugs.


Sujet(s)
Deltacoronavirus (genre) , Protéines virales non structurales , Animaux , Suidae , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Deltacoronavirus (genre)/génétique , Deltacoronavirus (genre)/physiologie , Maladies des porcs/virologie , Maladies des porcs/immunologie , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/virologie , Infections à coronavirus/immunologie , Infections à coronavirus/métabolisme , Interféron de type I/métabolisme , Interféron de type I/génétique , Immunité innée , Cellules HEK293 , Échappement immunitaire , Ubiquitination
10.
Curr Opin Immunol ; 87: 102430, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38824869

RÉSUMÉ

Lambda interferons (IFNλs), also termed type III interferons (IFNs) or interleukins-28/29, have been in the shadow of type I IFNs for a long time. Their common induction mechanisms and signalling cascades with type I IFNs have made difficult the unwinding of their unique nonredundant functions. However, this is now changing with mounting evidence supporting a major role of IFNλs as a specialized antiviral defense system in the body, mediating protection at mucosal barrier surfaces while limiting immunopathology. Here, we review the latest progress on the complex activities of IFNλs in the respiratory tract, focusing on their multiple effects in IFNλ receptor-expressing cells, the modulation of innate and adaptive immune responses in the context of infections and respiratory diseases, and their similarities and differences with type I IFNs. We also discuss their potential in therapeutic applications and the most recent developments in that direction.


Sujet(s)
Immunité acquise , Immunité innée , Interféron lambda , Interférons , Appareil respiratoire , Humains , Animaux , Interférons/métabolisme , Interférons/immunologie , Appareil respiratoire/immunologie , Appareil respiratoire/métabolisme , Transduction du signal/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie
11.
Front Immunol ; 15: 1380220, 2024.
Article de Anglais | MEDLINE | ID: mdl-38799458

RÉSUMÉ

African swine fever (ASF) is an acute hemorrhagic and devastating infectious disease affecting domestic pigs and wild boars. It is caused by the African swine fever virus (ASFV), which is characterized by genetic diversity and sophisticated immune evasion strategies. To facilitate infection, ASFV encodes multiple proteins to antagonize host innate immune responses, thereby contributing to viral virulence and pathogenicity. The molecular mechanisms employed by ASFV-encoded proteins to modulate host antiviral responses have not been comprehensively elucidated. In this study, it was observed that the ASFV MGF505-6R protein, a member of the multigene family 505 (MGF505), effectively suppressed the activation of the interferon-beta (IFN-ß) promoter, leading to reduced mRNA levels of antiviral genes. Additional evidence has revealed that MGF505-6R antagonizes the cGAS-STING signaling pathway by interacting with the stimulator of interferon genes (STING) for degradation in the autophagy-lysosomal pathway. The domain mapping revealed that the N-terminal region (1-260aa) of MGF505-6R is the primary domain responsible for interacting with STING, while the CTT domain of STING is crucial for its interaction with MGF505-6R. Furthermore, MGF505-6R also inhibits the activation of STING by reducing the K63-linked polyubiquitination of STING, leading to the disruption of STING oligomerization and TANK binding kinase 1 (TBK1) recruitment, thereby impairing the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3). Collectively, our study elucidates a novel strategy developed by ASFV MGF505-6R to counteract host innate immune responses. This discovery may offer valuable insights for further exploration of ASFV immune evasion mechanisms and antiviral strategies.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Protéines membranaires , Protéines virales , Animaux , Virus de la peste porcine africaine/immunologie , Virus de la peste porcine africaine/génétique , Suidae , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines membranaires/immunologie , Peste porcine africaine/immunologie , Peste porcine africaine/virologie , Peste porcine africaine/métabolisme , Protéines virales/immunologie , Protéines virales/métabolisme , Protéines virales/génétique , Humains , Immunité innée , Interféron de type I/métabolisme , Interféron de type I/immunologie , Facteur-3 de régulation d'interféron/métabolisme , Facteur-3 de régulation d'interféron/immunologie , Transduction du signal , Protéolyse , Cellules HEK293 , Interactions hôte-pathogène/immunologie , Échappement immunitaire , Interféron bêta/métabolisme , Interféron bêta/immunologie , Interféron bêta/génétique
12.
Nat Commun ; 15(1): 4484, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38802340

RÉSUMÉ

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


Sujet(s)
Interféron de type I , Janus kinase 1 , Récepteur à l'interféron alpha-bêta , Facteur de transcription STAT-2 , Transduction du signal , TYK2 Kinase , Ubiquitination , Humains , Cellules HEK293 , Interféron de type I/métabolisme , Sous-unité gamma du complexe ISGF3/métabolisme , Sous-unité gamma du complexe ISGF3/génétique , Janus kinase 1/métabolisme , Phosphorylation , Protéome/métabolisme , Récepteur à l'interféron alpha-bêta/métabolisme , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-2/métabolisme , TYK2 Kinase/métabolisme , Ubiquitin-protein ligases/métabolisme
13.
Cell Rep Med ; 5(5): 101569, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38744279

RÉSUMÉ

Systemic lupus erythematosus (SLE) displays a hallmark interferon (IFN) signature. Yet, clinical trials targeting type I IFN (IFN-I) have shown variable efficacy, and blocking IFN-II failed to treat SLE. Here, we show that IFN type levels in SLE vary significantly across clinical and transcriptional endotypes. Whereas skin involvement correlated with IFN-I alone, systemic features like nephritis associated with co-elevation of IFN-I, IFN-II, and IFN-III, indicating additive IFN effects in severe SLE. Notably, while high IFN-II/-III levels without IFN-I had a limited effect on disease activity, IFN-II was linked to IFN-I-independent transcriptional profiles (e.g., OXPHOS and CD8+GZMH+ cells), and IFN-III enhanced IFN-induced gene expression when co-elevated with IFN-I. Moreover, dysregulated IFNs do not explain the IFN signature in 64% of patients or clinical manifestations including cytopenia, serositis, and anti-phospholipid syndrome, implying IFN-independent endotypes in SLE. This study sheds light on mechanisms underlying SLE heterogeneity and the variable response to IFN-targeted therapies in clinical trials.


Sujet(s)
Interférons , Lupus érythémateux disséminé , Humains , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/anatomopathologie , Interférons/métabolisme , Interférons/génétique , Femelle , Adulte , Mâle , Transcriptome/génétique , Interféron de type I/métabolisme , Interféron de type I/génétique , Adulte d'âge moyen , Transcription génétique , Régulation de l'expression des gènes
14.
Int J Mol Sci ; 25(10)2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38791547

RÉSUMÉ

The COVID-19 pandemic has made assessing vaccine efficacy more challenging. Besides neutralizing antibody assays, systems vaccinology studies use omics technology to reveal immune response mechanisms and identify gene signatures in human peripheral blood mononuclear cells (PBMCs). However, due to their low proportion in PBMCs, profiling the immune response signatures of dendritic cells (DCs) is difficult. Here, we develop a predictive model for evaluating early immune responses in dendritic cells. We establish a THP-1-derived dendritic cell (TDDC) model and stimulate their maturation in vitro with an optimal dose of attenuated yellow fever 17D (YF-17D). Transcriptomic analysis reveals that type I interferon (IFN-I)-induced immunity plays a key role in dendritic cells. IFN-I regulatory biomarkers (IRF7, SIGLEC1) and IFN-I-inducible biomarkers (IFI27, IFI44, IFIT1, IFIT3, ISG15, MX1, OAS2, OAS3) are identified and validated in vitro and in vivo. Furthermore, we apply this TDDC approach to various types of vaccines, providing novel insights into their early immune response signatures and their heterogeneity in vaccine recipients. Our findings suggest that a standardizable TDDC model is a promising predictive approach to assessing early immunity in DCs. Further research into vaccine efficacy assessment approaches on various types of immune cells could lead to a systemic regimen for vaccine development in the future.


Sujet(s)
Cellules dendritiques , Vaccination , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Humains , Vaccination/méthodes , Interféron de type I/métabolisme , Interféron de type I/immunologie , Cellules THP-1 , COVID-19/immunologie , COVID-19/prévention et contrôle , Animaux , SARS-CoV-2/immunologie , Marqueurs biologiques , Vaccins contre la COVID-19/immunologie , Analyse de profil d'expression de gènes , Souris , Transcriptome , Vaccin antiamaril/immunologie
15.
J Clin Invest ; 134(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38690736

RÉSUMÉ

Pain and inflammation are biologically intertwined responses that warn the body of potential danger. In this issue of the JCI, Defaye, Bradaia, and colleagues identified a functional link between inflammation and pain, demonstrating that inflammation-induced activation of stimulator of IFN genes (STING) in dorsal root ganglia nociceptors reduced pain-like behaviors in a rodent model of inflammatory pain. Utilizing mice with a gain-of-function STING mutation, Defaye, Bradaia, and colleagues identified type I IFN regulation of voltage-gated potassium channels as the mechanism of this pain relief. Further investigation into mechanisms by which proinflammatory pathways can reduce pain may reveal druggable targets and insights into new approaches for treating persistent pain.


Sujet(s)
Ganglions sensitifs des nerfs spinaux , Protéines membranaires , Douleur , Animaux , Souris , Ganglions sensitifs des nerfs spinaux/métabolisme , Douleur/génétique , Douleur/métabolisme , Douleur/immunologie , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Humains , Nocicepteurs/métabolisme , Inflammation/génétique , Inflammation/immunologie , Inflammation/métabolisme , Canaux potassiques voltage-dépendants/génétique , Canaux potassiques voltage-dépendants/métabolisme , Canaux potassiques voltage-dépendants/immunologie , Interféron de type I/métabolisme , Interféron de type I/génétique , Interféron de type I/immunologie
16.
J Clin Immunol ; 44(6): 129, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38773012

RÉSUMÉ

Mutations in genes of the DNA polymerase complex have been linked to impaired immunological function next to distinct syndromic features. Biallelic mutations in PRIM1 are associated with a primordial dwarfism syndrome with variable hypogammaglobulinemia. The disease is mostly lethal in infancy due to pulmonary infections as well as hepatic cirrhosis. We studied 3 novel patients with PRIM1-deficiency with a focus on immunological consequences. All three shared dysmorphic features including a prominent forehead, triangular face and bilateral cryptorchidism. P1 carried the novel homozygous PRIM1 splice variant c.103+2T>G, allowing residual protein expression and associated with a mild clinical phenotype. P2 and P3 carried the known homozygous variant c.638+36C>G and died in infancy. Paradoxically, B cell lymphopenia was most pronounced in P1. No other significant lymphocyte abnormalities were detected. Interestingly, all 3 patients showed variable, but intermittently excessive Type I interferon signatures. In summary, the B-cell deficiency in PRIM1-deficiency is markedly variable and the severity of syndromic manifestations is not predictive of the immunological phenotype. We highlight a potential contribution of pathological type I interferon activation to disease pathogenesis which warrants further investigations.


Sujet(s)
Allèles , Lymphocytes B , Mutation , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Mâle , Lymphocytes B/immunologie , Déficits immunitaires/génétique , Déficits immunitaires/diagnostic , Interféron de type I/métabolisme , Mutation/génétique , Phénotype
17.
PLoS Pathog ; 20(5): e1012230, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38776321

RÉSUMÉ

While macrophage is one of the major type I interferon (IFN-I) producers in multiple tissues during viral infections, it also serves as an important target cell for many RNA viruses. However, the regulatory mechanism for the IFN-I response of macrophages to respond to a viral challenge is not fully understood. Here we report ADAP, an immune adaptor protein, is indispensable for the induction of the IFN-I response of macrophages to RNA virus infections via an inhibition of the conjugation of ubiquitin-like ISG15 (ISGylation) to RIG-I. Loss of ADAP increases RNA virus replication in macrophages, accompanied with a decrease in LPS-induced IFN-ß and ISG15 mRNA expression and an impairment in the RNA virus-induced phosphorylation of IRF3 and TBK1. Moreover, using Adap-/- mice, we show ADAP deficiency strongly increases the susceptibility of macrophages to RNA-virus infection in vivo. Mechanically, ADAP selectively interacts and functionally cooperates with RIG-I but not MDA5 in the activation of IFN-ß transcription. Loss of ADAP results in an enhancement of ISGylation of RIG-I, whereas overexpression of ADAP exhibits the opposite effect in vitro, indicating ADAP is detrimental to the RNA virus-induced ISGylation of RIG-I. Together, our data demonstrate a novel antagonistic activity of ADAP in the cell-intrinsic control of RIG-I ISGylation, which is indispensable for initiating and sustaining the IFN-I response of macrophages to RNA virus infections and replication.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéine-58 à domaine DEAD , Interféron de type I , Macrophages , Souris knockout , Infections à virus à ARN , Ubiquitines , Animaux , Macrophages/virologie , Macrophages/métabolisme , Macrophages/immunologie , Souris , Infections à virus à ARN/immunologie , Infections à virus à ARN/métabolisme , Ubiquitines/métabolisme , Ubiquitines/génétique , Protéine-58 à domaine DEAD/métabolisme , Interféron de type I/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Cytokines/métabolisme , Souris de lignée C57BL , Humains , Récepteurs immunologiques/métabolisme , Interféron bêta/métabolisme , Virus à ARN/immunologie , Facteur-3 de régulation d'interféron/métabolisme
18.
Nat Commun ; 15(1): 4153, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755212

RÉSUMÉ

Viral myocarditis, an inflammatory disease of the heart, causes significant morbidity and mortality. Type I interferon (IFN)-mediated antiviral responses protect against myocarditis, but the mechanisms are poorly understood. We previously identified A Disintegrin And Metalloproteinase domain 9 (ADAM9) as an important factor in viral pathogenesis. ADAM9 is implicated in a range of human diseases, including inflammatory diseases; however, its role in viral infection is unknown. Here, we demonstrate that mice lacking ADAM9 are more susceptible to encephalomyocarditis virus (EMCV)-induced death and fail to mount a characteristic type I IFN response. This defect in type I IFN induction is specific to positive-sense, single-stranded RNA (+ ssRNA) viruses and involves melanoma differentiation-associated protein 5 (MDA5)-a key receptor for +ssRNA viruses. Mechanistically, ADAM9 binds to MDA5 and promotes its oligomerization and thereby downstream mitochondrial antiviral-signaling protein (MAVS) activation in response to EMCV RNA stimulation. Our findings identify a role for ADAM9 in the innate antiviral response, specifically MDA5-mediated IFN production, which protects against virus-induced cardiac damage, and provide a potential therapeutic target for treatment of viral myocarditis.


Sujet(s)
Protéines ADAM , Infections à cardiovirus , Virus de l'encéphalomyocardite , Immunité innée , Interféron de type I , Hélicase IFIH1 inductrice de l'interféron , Protéines membranaires , Souris knockout , Myocardite , Animaux , Virus de l'encéphalomyocardite/immunologie , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Hélicase IFIH1 inductrice de l'interféron/génétique , Hélicase IFIH1 inductrice de l'interféron/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie , Infections à cardiovirus/immunologie , Infections à cardiovirus/virologie , Protéines ADAM/métabolisme , Protéines ADAM/génétique , Protéines ADAM/immunologie , Souris , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines membranaires/immunologie , Myocardite/immunologie , Myocardite/virologie , Humains , Souris de lignée C57BL , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/immunologie , Transduction du signal/immunologie , Mâle , Cellules HEK293
19.
Sci Adv ; 10(22): eadk5011, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38809975

RÉSUMÉ

Healthy behavioral patterns could modulate organ functions to enhance the body's immunity. However, how exercise regulates antiviral innate immunity remains elusive. Here, we found that exercise promotes type I interferon (IFN-I) production in the liver and enhances IFN-I immune activity of the body. Despite the possibility that many exercise-induced factors could affect IFN-I production, we identified Gpld1 as a crucial molecule, and the liver as the major organ to promote IFN-I production after exercise. Exercise largely loses the efficiency to induce IFN-I in Gpld1-/- mice. Further studies demonstrated that exercise-produced 3-hydroxybutanoic acid (3-HB) critically induces Gpld1 expression in the liver. Gpld1 blocks the PP2A-IRF3 interaction, thus enhancing IRF3 activation and IFN-I production, and eventually improving the body's antiviral ability. This study reveals that exercise improves antiviral innate immunity by linking the liver metabolism to systemic IFN-I activity and uncovers an unknown function of liver cells in innate immunity.


Sujet(s)
Immunité innée , Facteur-3 de régulation d'interféron , Interféron de type I , Foie , Conditionnement physique d'animal , Animaux , Mâle , Souris , Antiviraux , Cytokines , Facteur-3 de régulation d'interféron/métabolisme , Interféron de type I/métabolisme , Foie/métabolisme , Foie/immunologie , Souris de lignée C57BL , Souris knockout , Transduction du signal , Ubiquitines , Glycosylphosphatidylinositol Diacylglycerol-Lyase/métabolisme
20.
Anticancer Res ; 44(6): 2577-2585, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38821598

RÉSUMÉ

BACKGROUND/AIM: Nuclear factor erythroid-derived 2-related factor-2 (NRF2) is a transcription factor that regulates stress response genes. It negatively regulates the immune system by acting as a transcriptional repressor of inflammatory genes or suppressing type I interferon (IFN) production pathways. NRF2 is often over-expressed in some tumors, including non-small cell lung cancer, and modulates these tumors via an immune-cold microenvironment. Thus, strategies to convert cold tumors into hot tumors are effective for cancer treatment. MATERIALS AND METHODS: NRF2 was knocked-down or over-expressed in human cancer cells (A549, HeLa, H1299, H1650) and mouse mammary adenocarcinoma TS/A cells. Cells were irradiated or transfected with poly(I:C), and changes in type I IFN levels were examined using quantitative real-time polymerase chain reaction and western blotting. Cytosolic DNA was assayed via PicoGreen staining and immune and cancer cells were co-cultured. RESULTS: Regulation of NRF2 expression altered type I IFN levels in the human lung cancer cell line A549 and several solid tumors. Down-regulation of NRF2 resulted in increased levels of cytosolic DNA and activated the cGAS-STING pathway. We confirmed that type I IFN was induced in NRF2-down-regulated tumor cells using ionizing radiation (IR). Furthermore, when dendritic cells and macrophages were co-cultured with IR-exposed NRF2 knockdown tumor cells, the immune cells produced more IFNB1 and CXCL10. CONCLUSION: The immunosuppressive tumor cell environment is improved by NRF2 down-regulation, and IR treatment may promote immune cell signaling activation.


Sujet(s)
Interféron de type I , Facteur-2 apparenté à NF-E2 , Rayonnement ionisant , Transduction du signal , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Humains , Interféron de type I/métabolisme , Animaux , Souris , Lignée cellulaire tumorale , Cellules A549 , Tumeurs du poumon/radiothérapie , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Microenvironnement tumoral/immunologie , Nucleotidyltransferases/génétique , Nucleotidyltransferases/métabolisme , Macrophages/immunologie , Macrophages/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...