Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 577
Filtrer
2.
Nat Commun ; 15(1): 7765, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39237507

RÉSUMÉ

Human parainfluenza virus type 3 (HPIV-3) can cause severe respiratory tract infections. There are no convenient small-animal infection models. Here, we show viral replication in the upper and lower airways of AG129 mice (double IFNα/ß and IFNγ receptor knockout mice) upon intranasal inoculation. By multiplex fluorescence RNAscope and immunohistochemistry followed by confocal microscopy, we demonstrate viral tropism to ciliated cells and club cells of the bronchiolar epithelium. HPIV-3 causes a marked lung pathology. No virus transmission of the virus was observed by cohousing HPIV-3-infected AG129 mice with other mice. Oral treatment with GS-441524, the parent nucleoside of remdesivir, reduced infectious virus titers in the lung, with a relatively normal histology. Intranasal treatment also affords an antiviral effect. Thus, AG129 mice serve as a robust preclinical model for developing therapeutic and prophylactic strategies against HPIV-3. We suggest further investigation of GS-441524 and its prodrug forms to treat HPIV-3 infection in humans.


Sujet(s)
Antiviraux , Modèles animaux de maladie humaine , Poumon , Souris knockout , Virus parainfluenza humain de type 3 , Infections à respirovirus , Animaux , Poumon/virologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Souris , Virus parainfluenza humain de type 3/effets des médicaments et des substances chimiques , Virus parainfluenza humain de type 3/physiologie , Antiviraux/pharmacologie , Infections à respirovirus/traitement médicamenteux , Infections à respirovirus/virologie , Humains , Réplication virale/effets des médicaments et des substances chimiques , Femelle , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/déficit , Adénosine/analogues et dérivés , Adénosine/pharmacologie , Tropisme viral , Benzamides , Phtalimides
3.
Int J Mol Sci ; 25(18)2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39337564

RÉSUMÉ

Excessive production and response to Type I interferons (IFNs) is a hallmark of systemic lupus erythematosus (SLE). Neuropsychiatric lupus (NPSLE) is a common manifestation of human SLE, with major depression as the most common presentation. Clinical studies have demonstrated that IFNα can cause depressive symptoms. We have shown that the kallikrein-kinin system (KKS) [comprised of kallikreins (Klks) and bradykinins] and angiotensin-converting enzyme inhibitors suppressed Type I IFN responses in dendritic cells from lupus-prone mice and human peripheral blood mononuclear cells. Tissue Klk genes are decreased in patients with lupus, and giving exogenous Klk1 ameliorated kidney pathology in mice. We retro-orbitally administered mouse klk1 gene-carrying adenovirus in the Murphy Roths Large lymphoproliferative (MRL/lpr) lupus-prone mice at early disease onset and analyzed immune responses and depressive-like behavior. Klk1 improved depressive-like behavior, suppressed interferon-responsive genes and neuroinflammation, and reduced plasma IFNα levels and proinflammatory cytokines. Klk1 also reduced IFNAR1 and JAK1 protein expression, important upstream molecules in Type I IFN signaling. Klk1 reduced bradykinin B1 receptor expression, which is known to induce proinflammatory response. Together, these findings suggest that Klk1 may be a potential therapeutic candidate to control IFNα production/responses and other inflammatory responses in SLE and NPSLE.


Sujet(s)
Dépression , Modèles animaux de maladie humaine , Interféron de type I , Lupus érythémateux disséminé , Souris de lignée MRL lpr , Kallicréines tissulaires , Animaux , Souris , Kallicréines tissulaires/génétique , Kallicréines tissulaires/métabolisme , Interféron de type I/métabolisme , Dépression/traitement médicamenteux , Dépression/métabolisme , Lupus érythémateux disséminé/métabolisme , Lupus érythémateux disséminé/génétique , Femelle , Humains , Interféron alpha/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Janus kinase 1/métabolisme , Janus kinase 1/génétique , Comportement animal/effets des médicaments et des substances chimiques , Cytokines/métabolisme
4.
Nat Commun ; 15(1): 7165, 2024 Aug 26.
Article de Anglais | MEDLINE | ID: mdl-39187481

RÉSUMÉ

Programmed cell death 1 (PD-1) is a premier cancer drug target for immune checkpoint blockade (ICB). Because PD-1 receptor inhibition activates tumor-specific T-cell immunity, research has predominantly focused on T-cell-PD-1 expression and its immunobiology. In contrast, cancer cell-intrinsic PD-1 functional regulation is not well understood. Here, we demonstrate induction of PD-1 in melanoma cells via type I interferon receptor (IFNAR) signaling and reversal of ICB efficacy through IFNAR pathway inhibition. Treatment of melanoma cells with IFN-α or IFN-ß triggers IFNAR-mediated Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling, increases chromatin accessibility and resultant STAT1/2 and IFN regulatory factor 9 (IRF9) binding within a PD-1 gene enhancer, and leads to PD-1 induction. IFNAR1 or JAK/STAT inhibition suppresses melanoma-PD-1 expression and disrupts ICB efficacy in preclinical models. Our results uncover type I IFN-dependent regulation of cancer cell-PD-1 and provide mechanistic insight into the potential unintended ICB-neutralizing effects of widely used IFNAR1 and JAK inhibitors.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Interféron de type I , Mélanome , Récepteur-1 de mort cellulaire programmée , Récepteur à l'interféron alpha-bêta , Transduction du signal , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Mélanome/traitement médicamenteux , Mélanome/immunologie , Mélanome/génétique , Mélanome/métabolisme , Humains , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire tumorale , Souris , Interféron de type I/métabolisme , Facteur de transcription STAT-1/métabolisme , Sous-unité gamma du complexe ISGF3/métabolisme , Sous-unité gamma du complexe ISGF3/génétique , Interféron bêta/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Janus kinases/métabolisme , Souris de lignée C57BL , Interféron alpha/pharmacologie , Interféron alpha/métabolisme , Femelle
5.
Proc Natl Acad Sci U S A ; 121(33): e2318190121, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39106307

RÉSUMÉ

We developed a highly sensitive assay for detecting protein-protein interaction using chimeric receptors comprising two molecules of interest in the extracellular domain and interferon alpha and beta receptor subunit 1 or 2 (IFNAR1/2) in the intracellular domain. This intracellular IFNAR1/2 reconstitution system (IFNARRS) proved markedly more sensitive than the NanoBiT system, currently considered one of the best detection systems for protein interaction. Employing chimeric receptors with extracellular domains from the IFNγ or IL-2 receptor and the intracellular domains of IFNAR1/2, the IFNARRS system effectively identifies low IFNγ or IL-2 levels. Cells stably expressing these chimeric receptors responded to IFNγ secreted by activated T cells following various stimuli, including a specific peptide-antigen. The activation signals were further enhanced by the expression of relevant genes, such as costimulators, via IFN-stimulated response elements in the promoters. Besides IFNγ or IL-2, the IFNARRS system demonstrated the capability to detect other cytokines by using the corresponding extracellular domains from these target cytokine receptors.


Sujet(s)
Interféron gamma , Interleukine-2 , Récepteur à l'interféron alpha-bêta , Lymphocytes T , Humains , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Lymphocytes T/métabolisme , Lymphocytes T/immunologie , Interleukine-2/métabolisme , Interféron gamma/métabolisme , Récepteurs à l'interleukine-2/métabolisme , Récepteurs à l'interleukine-2/génétique , Liaison aux protéines , Activation des lymphocytes , Cellules HEK293
6.
J Virol ; 98(9): e0089324, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39194249

RÉSUMÉ

Oropouche fever caused by Oropouche virus (OROV) is a significant zoonosis in Central and South America. Despite its public health significance, we lack high-throughput diagnostics, therapeutics, and a comprehensive knowledge of OROV biology. Reporter viruses are valuable tools to rapidly study virus dynamics and develop neutralization and antiviral screening assays. OROV is a tri-segmented bunyavirus, which makes generating a reporter virus challenging, as introducing foreign elements into the viral genome typically affects fitness. We previously demonstrated that the non-structural gene NSm on the OROV medium (M) segment is non-essential for replication in vitro. Taking advantage of this, we have now generated a recombinant OROV expressing fluorescent protein ZsGreen in place of NSm. This reporter OROV is both stable and pathogenic in IFNAR-/- mice and provides a powerful tool for OROV pathogenesis studies and assay development.IMPORTANCEEmerging and reemerging infectious agents such as zoonotic bunyaviruses are of global health concern. Oropouche virus (OROV) causes recurring outbreaks of acute febrile illness in the Central and South American human populations. Biting midges are the primary transmission vectors, whereas sloths and non-human primates are their reservoir hosts. As global temperatures increase, we will likely see an expansion in arthropod-borne pathogens such as OROV. Therefore, developing reagents to study pathogen biology to aid in identifying druggable targets is essential. Here, we demonstrate the feasibility and use of a fluorescent OROV reporter in mice to study viral dynamics and pathogenesis. We show that this reporter OROV maintains characteristics such as growth and pathogenicity similar to the wild-type virus. Using this reporter virus, we can now develop methods to assist OROV studies and establish various high-throughput assays.


Sujet(s)
Infections à Bunyaviridae , Gènes rapporteurs , Orthobunyavirus , Animaux , Orthobunyavirus/génétique , Orthobunyavirus/pathogénicité , Souris , Infections à Bunyaviridae/virologie , Réplication virale , Humains , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Protéines virales non structurales/génétique , Protéines virales non structurales/métabolisme , Souris knockout
7.
J Neuroinflammation ; 21(1): 166, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956653

RÉSUMÉ

BACKGROUND: Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM. METHODS: A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR. RESULTS: Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation. CONCLUSIONS: HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.


Sujet(s)
Anxiété , Diabète de type 2 , Alimentation riche en graisse , Hypoxie , Glycoprotéines membranaires , Souris de lignée C57BL , Récepteur à l'interféron alpha-bêta , Récepteurs immunologiques , Transduction du signal , Animaux , Souris , Alimentation riche en graisse/effets indésirables , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Anxiété/étiologie , Anxiété/métabolisme , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Hypoxie/métabolisme , Hypoxie/complications , Mâle , Diabète de type 2/complications , Diabète de type 2/métabolisme , Diabète de type 2/psychologie , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Diabète expérimental/complications , Diabète expérimental/métabolisme , Microglie/métabolisme , Facteur de transcription STAT-1/métabolisme , Syndrome d'apnées obstructives du sommeil/complications , Syndrome d'apnées obstructives du sommeil/métabolisme , Syndrome d'apnées obstructives du sommeil/psychologie
8.
Cells ; 13(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38995014

RÉSUMÉ

PD-1 (Programmed cell death protein 1) regulates the metabolic reprogramming of myeloid-derived suppressor cells and myeloid cell differentiation, as well as the type I interferon (IFN-I) signaling pathway in myeloid cells in the tumor microenvironment. PD-1, therefore, is a key inhibitory receptor in myeloid cells. However, the regulation of PD-1 expression in myeloid cells is unknown. We report that the expression level of PDCD1, the gene that encodes the PD-1 protein, is positively correlated with the levels of IFNB1 and IFNAR1 in myeloid cells in human colorectal cancer. Treatment of mouse myeloid cell lines with recombinant IFNß protein elevated PD-1 expression in myeloid cells in vitro. Knocking out IFNAR1, the gene that encodes the IFN-I-specific receptor, diminished the inductive effect of IFNß on PD-1 expression in myeloid cells in vitro. Treatment of tumor-bearing mice with a lipid nanoparticle-encapsulated IFNß-encoding plasmid (IFNBCOL01) increased IFNß expression, resulting in elevated PD-1 expression in tumor-infiltrating myeloid cells. At the molecular level, we determined that IFNß activates STAT1 (signal transducer and activator of transcription 1) and IRFs (interferon regulatory factors) in myeloid cells. Analysis of the cd279 promoter identified IRF2-binding consensus sequence elements. ChIP (chromatin immunoprecipitation) analysis determined that the pSTAT1 directly binds to the irf2 promoter and that IRF2 directly binds to the cd279 promoter in myeloid cells in vitro and in vivo. In colon cancer patients, the expression levels of STAT1, IRF2 and PDCD1 are positively correlated in tumor-infiltrating myeloid cells. Our findings determine that IFNß activates PD-1 expression at least in part by an autocrine mechanism via the stimulation of the pSTAT1-IRF2 axis in myeloid cells.


Sujet(s)
Facteur-2 de régulation d'interféron , Cellules myéloïdes , Récepteur-1 de mort cellulaire programmée , Facteur de transcription STAT-1 , Transduction du signal , Cellules myéloïdes/métabolisme , Cellules myéloïdes/effets des médicaments et des substances chimiques , Animaux , Humains , Facteur de transcription STAT-1/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Souris , Facteur-2 de régulation d'interféron/métabolisme , Facteur-2 de régulation d'interféron/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Interféron de type I/métabolisme , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Interféron bêta/métabolisme , Lignée cellulaire tumorale , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris de lignée C57BL
9.
J Exp Med ; 221(9)2024 09 02.
Article de Anglais | MEDLINE | ID: mdl-39042188

RÉSUMÉ

The contribution of placental immune responses to congenital Zika virus (ZIKV) syndrome remains poorly understood. Here, we leveraged a mouse model of ZIKV infection to identify mechanisms of innate immune restriction exclusively in the fetal compartment of the placenta. ZIKV principally infected mononuclear trophoblasts in the junctional zone, which was limited by mitochondrial antiviral-signaling protein (MAVS) and type I interferon (IFN) signaling mechanisms. Single nuclear RNA sequencing revealed MAVS-dependent expression of IFN-stimulated genes (ISGs) in spongiotrophoblasts but not in other placental cells that use alternate pathways to induce ISGs. ZIKV infection of Ifnar1-/- or Mavs-/- placentas was associated with greater infection of the adjacent immunocompetent decidua, and heterozygous Mavs+/- or Ifnar1+/- dams carrying immunodeficient fetuses sustained greater maternal viremia and tissue infection than dams carrying wild-type fetuses. Thus, MAVS-IFN signaling in the fetus restricts ZIKV infection in junctional zone trophoblasts, which modulates dissemination and outcome for both the fetus and the pregnant mother.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Caduques , Foetus , Interféron de type I , Placenta , Récepteur à l'interféron alpha-bêta , Transduction du signal , Trophoblastes , Infection par le virus Zika , Virus Zika , Femelle , Animaux , Grossesse , Interféron de type I/métabolisme , Interféron de type I/immunologie , Transduction du signal/immunologie , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Placenta/immunologie , Placenta/virologie , Placenta/métabolisme , Infection par le virus Zika/immunologie , Infection par le virus Zika/virologie , Virus Zika/immunologie , Virus Zika/physiologie , Souris , Caduques/immunologie , Caduques/virologie , Caduques/métabolisme , Foetus/immunologie , Foetus/virologie , Trophoblastes/immunologie , Trophoblastes/virologie , Trophoblastes/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Souris de lignée C57BL , Souris knockout , Immunité innée , Complications infectieuses de la grossesse/immunologie , Complications infectieuses de la grossesse/virologie , Modèles animaux de maladie humaine
10.
Int J Mol Sci ; 25(14)2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39063175

RÉSUMÉ

Type I interferon (IFN-I) signaling has been shown to be upregulated in systemic sclerosis (SSc). Dysregulated B-cell functions, including antigen presentation, as well as antibody and cytokine production, all of which may be affected by IFN-I signaling, play an important role in the pathogenesis of the disease. We investigated the IFN-I signature in 71 patients with the more severe form of the disease, diffuse cutaneous SSc (dcSSc), and 33 healthy controls (HCs). Activation via Toll-like receptors (TLRs) can influence the IFN-I signaling cascade; thus, we analyzed the effects of the TLR homologue CD180 ligation on the IFN-I signature in B cells. CD180 stimulation augmented the phosphorylation of signal transducer and activator of transcription 1 (STAT1) in dcSSc B cells (p = 0.0123). The expression of IFN-I receptor (IFNAR1) in non-switched memory B cells producing natural autoantibodies was elevated in dcSSc (p = 0.0109), which was enhanced following anti-CD180 antibody treatment (p = 0.0125). Autoantibodies to IFN-Is (IFN-alpha and omega) correlated (dcSSc p = 0.0003, HC p = 0.0192) and were present at similar levels in B cells from dcSSc and HC, suggesting their regulatory role as natural autoantibodies. It can be concluded that factors other than IFN-alpha may contribute to the elevated IFN-I signature of dcSSc B cells, and one possible candidate is B-cell activation via CD180.


Sujet(s)
Antigènes CD , Autoanticorps , Lymphocytes B , Interféron de type I , Humains , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Adulte d'âge moyen , Femelle , Mâle , Autoanticorps/immunologie , Antigènes CD/métabolisme , Adulte , Interféron de type I/métabolisme , Facteur de transcription STAT-1/métabolisme , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Sclérodermie diffuse/immunologie , Sclérodermie diffuse/métabolisme , Sujet âgé , Régulation positive , Transduction du signal
11.
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
12.
Cell Mol Immunol ; 21(8): 892-904, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38906982

RÉSUMÉ

Type I interferon (IFN-I) exhibits broad-spectrum antiviral properties and is commonly employed in clinical for the treatment of viral infections. In this study, we unveil SENP6 as a potent regulator of IFN-I antiviral activity. SENP6 does not impact the production of IFN-I induced by viruses but rather modulates IFN-I-activated signaling. Mechanistically, SENP6 constitutively interacts with USP8 and inhibits the SUMOylation of USP8, consequently restricting the interaction between USP8 and IFNAR2. The dissociation of USP8 from IFNAR2 enhances IFNAR2 ubiquitination and degradation, thus attenuating IFN-I antiviral activity. Correspondingly, the downregulation of SENP6 promotes the interaction between USP8 and IFNAR2, leading to a reduction in IFNAR2 ubiquitination and, consequently, an enhancement in IFN-I-induced signaling. This study deciphers a critical deSUMOylation-deubiquitination crosstalk that finely regulates the IFN-I response to viral infection.


Sujet(s)
Endopeptidases , Interféron de type I , Récepteur à l'interféron alpha-bêta , Transduction du signal , Sumoylation , Ubiquitin thiolesterase , Ubiquitination , Humains , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Interféron de type I/métabolisme , Ubiquitin thiolesterase/métabolisme , Cellules HEK293 , Endopeptidases/métabolisme , Antiviraux/pharmacologie , Animaux , Liaison aux protéines , Maladies virales/immunologie , Complexes de tri endosomique requis pour le transport
13.
Brain Res ; 1840: 149082, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38866307

RÉSUMÉ

Ubiquitin specific protease 18 (USP18) serves as a potent inhibitor of Type I interferon (IFN) signaling. Previous studies have shown that Usp18 deficient (homozygous Usp18 gene knockout) mice exhibit hydrocephalus; however, the precise molecular mechanism underlying hydrocephalus development remains elusive. In this study, we demonstrate that mice lacking both type I IFN receptor subunit 1 (Ifnar1) and Usp18 (Ifnar1/Usp18 double knockout mice) are viable and do not display a hydrocephalus phenotype. Moreover, we observed that suppression of USP18 in ependymal cells treated with IFN significantly increased cell death, including pyroptosis, and decreased proliferation. These findings suggest that heightened sensitivity to type I IFN during brain development contributes to the onset of hydrocephalus. Furthermore, they imply that inhibition of IFN signaling may hold promise as a therapeutic strategy for hydrocephalus.


Sujet(s)
Hydrocéphalie , Interféron de type I , Souris knockout , Récepteur à l'interféron alpha-bêta , Ubiquitin thiolesterase , Animaux , Hydrocéphalie/génétique , Hydrocéphalie/anatomopathologie , Interféron de type I/métabolisme , Souris , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Souris de lignée C57BL , Transduction du signal/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/anatomopathologie , Encéphale/effets des médicaments et des substances chimiques , Épendyme/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Pyroptose/effets des médicaments et des substances chimiques , Pyroptose/physiologie
14.
Cardiovasc Res ; 120(11): 1351-1364, 2024 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-38836630

RÉSUMÉ

AIMS: Abdominal aortic aneurysm (AAA) represents a life-threatening condition characterized by medial layer degeneration of the abdominal aorta. Nevertheless, knowledge regarding changes in regulators associated with aortic status remains incomplete. A thorough understanding of cell types and signalling pathways involved in the development and progression of AAAs is essential for the development of medical therapy. METHODS AND RESULTS: We harvested specimens of the abdominal aorta with different pathological features in Angiotensin II (AngII)-infused ApoE-/- mice, conducted scRNA-seq, and identified a unique population of interferon-inducible monocytes/macrophages (IFNICs), which were amply found in the AAAs. Gene set variation analysis revealed that activation of the cytosolic DNA sensing cGAS-STING and JAK-STAT pathways promoted the secretion of type I interferons in monocytes/macrophages and differentiated them into IFNICs. We generated myeloid cell-specific deletion of Sting1 (Lyz2-Cre+/-; Sting1flox/flox) mice and performed bone marrow transplantation and found that myeloid cell-specific deletion of Sting1 or Ifnar1 significantly reduced the incidence of AAA, aortic rupture rate, and diameter of the abdominal aorta. Mechanistically, the activated pyroptosis- and inflammation-related signalling pathways, regulated by IRF7 in IFNICs, play critical roles in the developing AAAs. CONCLUSION: IFNICs are a unique monocyte/macrophage subset implicated in the development of AAAs and aortic rupture.


Sujet(s)
Angiotensine-II , Aorte abdominale , Anévrysme de l'aorte abdominale , Rupture aortique , Modèles animaux de maladie humaine , Évolution de la maladie , Macrophages , Protéines membranaires , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Monocytes , Récepteur à l'interféron alpha-bêta , Transduction du signal , Analyse sur cellule unique , Anévrysme de l'aorte abdominale/métabolisme , Anévrysme de l'aorte abdominale/anatomopathologie , Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/génétique , Anévrysme de l'aorte abdominale/prévention et contrôle , Animaux , Macrophages/métabolisme , Macrophages/anatomopathologie , Macrophages/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Monocytes/anatomopathologie , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Aorte abdominale/métabolisme , Aorte abdominale/anatomopathologie , Aorte abdominale/immunologie , Rupture aortique/prévention et contrôle , Rupture aortique/anatomopathologie , Rupture aortique/métabolisme , Rupture aortique/génétique , Rupture aortique/induit chimiquement , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Mâle , RNA-Seq , Interféron de type I/métabolisme , Interféron de type I/génétique , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Janus kinases/métabolisme , Cellules cultivées , Apolipoprotéines E
15.
Int J Biol Sci ; 20(8): 3076-3093, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904031

RÉSUMÉ

Epizootic hemorrhagic disease (EHD), caused by Epizootic hemorrhagic disease virus (EHDV), is an emerging and severe livestock disease. Recent incursion and distribution of EHDV in Europe have outlined the emerging character of EHD. Despite its worldwide impact, numerous knowledge gaps exist. A range of inconveniences restricts utilization of natural hosts of EHDV. Here, we show that adult mice deficient in type I IFN receptor (IFNAR(-/-)) are highly susceptible to EHDV-6 and EHDV-8 infection when the virus is administered subcutaneously. Disease was characterized by ruffled hair, reluctance to move, dehydration and conjunctivitis, with viraemia detected from day 5 post-infection. A deeper characterization of EHDV-8 infection showed viral replication in the lung, liver, spleen, kidney, testis and ovaries. Importantly, increased expression levels of pro-inflammatory cytokines IL-1ß, IL-6 and CXCL2 were observed in spleen after EHDV-8 infection. Furthermore, IFNAR(-/-) adult mice immunized with a EHDV-8 inactivated vaccine elicited neutralizing antibodies specific of EHDV-8 and full protection against challenge with a lethal dose of this virus. This study also explores the possibilities of this animal model for study of BTV and EHDV coinfection. In summary, the IFNAR(-/-) mouse model faithfully recapitulates EHD and can be applied for vaccine testing, which can facilitate progress in addressing the animal health challenge posed by this virus.


Sujet(s)
Modèles animaux de maladie humaine , Virus de la maladie hémorragique épizootique , Récepteur à l'interféron alpha-bêta , Vaccins antiviraux , Animaux , Souris , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Virus de la maladie hémorragique épizootique/immunologie , Virus de la maladie hémorragique épizootique/génétique , Vaccins antiviraux/immunologie , Infections à Reoviridae/immunologie , Femelle , Souris knockout , Anticorps neutralisants/immunologie , Mâle
16.
Immunity ; 57(7): 1696-1709.e10, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38878770

RÉSUMÉ

Aicardi-Goutières syndrome (AGS) is an autoinflammatory disease characterized by aberrant interferon (IFN)-α production. The major cause of morbidity in AGS is brain disease, yet the primary source and target of neurotoxic IFN-α remain unclear. Here, we demonstrated that the brain was the primary source of neurotoxic IFN-α in AGS and confirmed the neurotoxicity of intracerebral IFN-α using astrocyte-driven Ifna1 misexpression in mice. Using single-cell RNA sequencing, we demonstrated that intracerebral IFN-α-activated receptor (IFNAR) signaling within cerebral endothelial cells caused a distinctive cerebral small vessel disease similar to that observed in individuals with AGS. Magnetic resonance imaging (MRI) and single-molecule ELISA revealed that central and not peripheral IFN-α was the primary determinant of microvascular disease in humans. Ablation of endothelial Ifnar1 in mice rescued microvascular disease, stopped the development of diffuse brain disease, and prolonged lifespan. These results identify the cerebral microvasculature as a primary mediator of IFN-α neurotoxicity in AGS, representing an accessible target for therapeutic intervention.


Sujet(s)
Encéphale , Interféron alpha , Microvaisseaux , Malformations du système nerveux , Récepteur à l'interféron alpha-bêta , Animaux , Humains , Souris , Interféron alpha/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Microvaisseaux/anatomopathologie , Malformations du système nerveux/génétique , Maladies auto-immunes du système nerveux/immunologie , Cellules endothéliales/métabolisme , Souris knockout , Mâle , Femelle , Transduction du signal , Souris de lignée C57BL , Astrocytes/métabolisme , Modèles animaux de maladie humaine
17.
J Bone Miner Res ; 39(8): 1132-1146, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-38874138

RÉSUMÉ

Type I interferons (IFN-I) are pleiotropic factors endowed with multiple activities that play important roles in innate and adaptive immunity. Although many studies indicate that IFN-I inducers exert favorable effects on broad-spectrum antivirus, immunomodulation, and anti-tumor activities by inducing endogenous IFN-I and IFN-stimulated genes, their function in bone homeostasis still needs further exploration. Here, our study demonstrates 2 distinct IFN-I inducers, diABZI and poly(I:C), as potential therapeutics to alleviate osteolysis and osteoporosis. First, IFN-I inducers suppress the genes that control osteoclast (OC) differentiation and activity in vitro. Moreover, diABZI alleviates bone loss in Ti particle-induced osteolysis and ovariectomized -induced osteoporosis in vivo by inhibiting OC differentiation and function. In addition, the inhibitory effects of IFN-I inducers on OC differentiation are not observed in macrophages derived from Ifnar1-/-mice, which indicate that the suppressive effect of IFN-I inducers on OC is IFNAR-dependent. Mechanistically, RNAi-mediated silencing of IRF7 and IFIT3 in OC precursors impairs the suppressive effect of the IFN-I inducers on OC differentiation. Taken together, these results demonstrate that IFN-I inducers play a protective role in bone turnover by limiting osteoclastogenesis and bone resorption through the induction of OC-specific mediators via the IFN-I signaling pathway.


OCs are responsible for bone resorption, and their excessive differentiation and enhanced activity will lead to bone resorption diseases such as osteoporosis and osteolysis. Here, our study demonstrates 2 distinct IFN-I inducers, diABZI and poly(I:C), as potential therapeutics to alleviate osteolysis and osteoporosis. IFN-I inducers suppress OC differentiation, and particularly diABZI alleviates bone loss in osteolysis and osteoporosis mouse models. Taken together, IFN-I inducers play a protective role in bone turnover by limiting osteoclastogenesis and bone resorption through the induction of OC-specific mediators via the IFN-I signaling pathway. Our in-depth and comprehensive discovery of the IFN-I inducer would provide new insight into OC biology and therapeutic targets for osteoclastic bone resorption diseases.


Sujet(s)
Résorption osseuse , Différenciation cellulaire , Facteur-7 de régulation d'interféron , Ostéoclastes , Poly I-C , Animaux , Ostéoclastes/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/anatomopathologie , Facteur-7 de régulation d'interféron/métabolisme , Résorption osseuse/anatomopathologie , Souris , Poly I-C/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Femelle , Souris de lignée C57BL , Souris knockout , Interféron de type I/métabolisme , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Humains , Ostéolyse/anatomopathologie , Ostéolyse/métabolisme , Ostéolyse/traitement médicamenteux
18.
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
19.
Front Cell Infect Microbiol ; 14: 1356542, 2024.
Article de Anglais | MEDLINE | ID: mdl-38741892

RÉSUMÉ

Background and objectives: This study aimed to investigate the possible prognostic significance of interferon alpha-beta receptor subunit 2 (IFNAR2) and tyrosine kinase 2 (TYK2) expressions. Methods: We conducted a retrospective study including COVID-19 adult patients. All blood samples were collected before any interventions. The expressions of IFNAR2 and TYK2 were assessed using real-time PCR in venous blood samples of 54 cases and 56 controls. The transcript quantities of IFNAR2 and TYK2 genes were assessed using a Delta-Ct method. Results: Our findings show no significant differences in gene expression levels for IFNAR2 and TYK2 between patients who required oxygen (O2) therapy and those who did not (p-value = 0.732 and p-value = 0.629, respectively). Likewise, there were no significant differences in IFNAR2 and TYK2 expressions between patients hospitalized for less than 7 days and those hospitalized for 7 days or more (p-value = 0.455 and p-value = 0.626, respectively). We also observed a weak correlation between IFNAR2 expression and CRP (p-value = 0.045, r = 0.192). There was a negative correlation between the expression levels of IFNAR2 and TYK2 transcripts in COVID-19 patients (p-value = 0.044; partial correlation coefficient = -0.283). Additionally, IFNAR2 and TYK2 were significantly downregulated in the COVID-19 group compared to healthy subjects (p-value = 0.002 and p-value = 0.028, respectively). However, neither IFNAR2 nor TYK2 expression was significantly different between the case subgroups based on COVID-19 severity. The IFNAR2 ΔΔCt (B = -0.184, 95% CI: -0.524-0.157, p-value = 0.275) and the TYK2 ΔΔCt (B = 0.114, 95% CI: -0.268-0.496, p-value = 0.543) were not found to be significant predictors of hospitalization duration. The area under the curve (AUC) for IFNAR2 expression is 0.655 (p-value = 0.005, 95% CI: 0.554-0.757), suggesting its poor discriminative value. Conclusion: We were unable to comment definitively on the prognostic power of IFNAR2 and TYK2 expressions in COVID-19 patients, and larger-scale studies are needed. The principal limitations of this study included the lack of longitudinal analysis and limited sample size.


Sujet(s)
COVID-19 , Récepteur à l'interféron alpha-bêta , TYK2 Kinase , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , COVID-19/génétique , Pronostic , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/métabolisme , Études rétrospectives , SARS-CoV-2 , TYK2 Kinase/génétique , TYK2 Kinase/métabolisme
20.
Cell Death Dis ; 15(5): 369, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38806478

RÉSUMÉ

Signal transducer and activator of transcription 3 (STAT3) is frequently overexpressed in patients with acute myeloid leukemia (AML). STAT3 exists in two distinct alternatively spliced isoforms, the full-length isoform STAT3α and the C-terminally truncated isoform STAT3ß. While STAT3α is predominantly described as an oncogenic driver, STAT3ß has been suggested to act as a tumor suppressor. To elucidate the role of STAT3ß in AML, we established a mouse model of STAT3ß-deficient, MLL-AF9-driven AML. STAT3ß deficiency significantly shortened survival of leukemic mice confirming its role as a tumor suppressor. Furthermore, RNA sequencing revealed enhanced STAT1 expression and interferon (IFN) signaling upon loss of STAT3ß. Accordingly, STAT3ß-deficient leukemia cells displayed enhanced sensitivity to blockade of IFN signaling through both an IFNAR1 blocking antibody and the JAK1/2 inhibitor Ruxolitinib. Analysis of human AML patient samples confirmed that elevated expression of IFN-inducible genes correlated with poor overall survival and low STAT3ß expression. Together, our data corroborate the tumor suppressive role of STAT3ß in a mouse model in vivo. Moreover, they provide evidence that its tumor suppressive function is linked to repression of the STAT1-mediated IFN response. These findings suggest that the STAT3ß/α mRNA ratio is a significant prognostic marker in AML and holds crucial information for targeted treatment approaches. Patients displaying a low STAT3ß/α mRNA ratio and unfavorable prognosis could benefit from therapeutic interventions directed at STAT1/IFN signaling.


Sujet(s)
Leucémie aigüe myéloïde , Facteur de transcription STAT-3 , Animaux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/métabolisme , Humains , Facteur de transcription STAT-3/métabolisme , Souris , Transduction du signal , Interférons/métabolisme , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Souris de lignée C57BL , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Lignée cellulaire tumorale , Nitriles , Pyrazoles , Pyrimidines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE