Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.035
Filtrer
1.
Ren Fail ; 46(2): 2379008, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39034431

RÉSUMÉ

Despite efforts to find effective drugs for sepsis-associated acute kidney injury (SA-AKI), mortality rates in patients with SA-AKI have not decreased. Our study evaluated the protective effects of isoflavone osajin (OSJ) on SA-AKI in rats by targeting inflammation, oxidative stress, and apoptosis, which represent the cornerstones in the pathophysiological mechanism of SA-AKI. Polymicrobial sepsis was induced in rats via the cecal ligation and puncture (CLP) technique. Markers of oxidative stress were evaluated in kidney tissues using biochemical methods. The expression of interleukin-33 (IL-33), 8-hydroxydeoxyguanosine (8-OHdG), caspase-3, and kidney injury molecule-1 (KIM-1) was evaluated as indicators of inflammation, DNA damage, apoptosis, and SA-AKI respectively in the kidney tissues using immunohistochemical and immunofluorescent detection methods. The CLP technique significantly (p < 0.001) increased lipid peroxidation (LPO) levels and significantly (p < 0.001) decreased the activities of superoxide dismutase and catalase in kidney tissues. In the renal tissues, strong expression of IL-33, 8-OHdG, caspase-3, and KIM-1 was observed with severe degeneration and necrosis in the tubular epithelium and intense interstitial nephritis. In contrast, the administration of OSJ significantly (p < 0.001) reduced the level of LPO, markedly improved biomarkers of antioxidant status, decreased the levels of serum creatinine and urea, lowered the expression of IL-33, 8-OHdG, caspase-3, and KIM-1 and alleviated changes in renal histopathology. A promising binding score was found via a molecular docking investigation of the OSJ-binding mode with mouse IL-33 (PDB Code: 5VI4). Therefore, OSJ protects against SA-AKI by suppressing the IL-33/LPO/8-OHdG/caspase-3 pathway and improving the antioxidant system.


Sujet(s)
Atteinte rénale aigüe , Antioxydants , Apoptose , Rein , Simulation de docking moléculaire , Stress oxydatif , Sepsie , Animaux , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/étiologie , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/prévention et contrôle , Antioxydants/pharmacologie , Antioxydants/usage thérapeutique , Sepsie/complications , Sepsie/traitement médicamenteux , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Apoptose/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Isoflavones/pharmacologie , Isoflavones/usage thérapeutique , Modèles animaux de maladie humaine , Interleukine-33/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Caspase-3/métabolisme , Rat Sprague-Dawley , Molécules d'adhérence cellulaire
2.
Proc Natl Acad Sci U S A ; 121(28): e2319994121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38959032

RÉSUMÉ

Upon encountering allergens, CD4+ T cells differentiate into IL-4-producing Th2 cells in lymph nodes, which later transform into polyfunctional Th2 cells producing IL-5 and IL-13 in inflamed tissues. However, the precise mechanism underlying their polyfunctionality remains elusive. In this study, we elucidate the pivotal role of NRF2 in polyfunctional Th2 cells in murine models of allergic asthma and in human Th2 cells. We found that an increase in reactive oxygen species (ROS) in immune cells infiltrating the lungs is necessary for the development of eosinophilic asthma and polyfunctional Th2 cells in vivo. Deletion of the ROS sensor NRF2 specifically in T cells, but not in dendritic cells, significantly abolished eosinophilia and polyfunctional Th2 cells in the airway. Mechanistically, NRF2 intrinsic to T cells is essential for inducing optimal oxidative phosphorylation and glycolysis capacity, thereby driving Th2 cell polyfunctionality independently of IL-33, partially by inducing PPARγ. Treatment with an NRF2 inhibitor leads to a substantial decrease in polyfunctional Th2 cells and subsequent eosinophilia in mice and a reduction in the production of Th2 cytokines from peripheral blood mononuclear cells in asthmatic patients. These findings highlight the critical role of Nrf2 as a spatial and temporal metabolic hub that is essential for polyfunctional Th2 cells, suggesting potential therapeutic implications for allergic diseases.


Sujet(s)
Asthme , Facteur-2 apparenté à NF-E2 , Espèces réactives de l'oxygène , Lymphocytes auxiliaires Th2 , Facteur-2 apparenté à NF-E2/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Lymphocytes auxiliaires Th2/métabolisme , Animaux , Souris , Asthme/immunologie , Asthme/métabolisme , Humains , Espèces réactives de l'oxygène/métabolisme , Récepteur PPAR gamma/métabolisme , Phosphorylation oxydative , Glycolyse , Poumon/immunologie , Poumon/métabolisme , Souris knockout , Modèles animaux de maladie humaine , Femelle , Cytokines/métabolisme , Souris de lignée C57BL , Interleukine-33/métabolisme , Éosinophilie/immunologie , Éosinophilie/métabolisme
3.
Am J Reprod Immunol ; 92(1): e13895, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39001587

RÉSUMÉ

PROBLEM: Preeclampsia (PE) is a hypertensive pregnancy disorder that is a leading cause of maternal and fetal morbidity and mortality characterized by maternal vascular dysfunction, oxidative stress, chronic immune activation, and excessive inflammation. No cure exists beyond delivery of the fetal-placental unit and the mechanisms driving pathophysiology are not fully understood. However, aberrant immune responses have been extensively characterized in clinical studies and shown to mediate PE pathophysiology in animal studies. One pathway that may mediate aberrant immune responses in PE is deficiencies in the IL-33 signaling pathway. In this study, we aim to investigate the impact of IL-33 signaling inhibition on cNK, TH17, and TReg populations, vascular function, and maternal blood pressure during pregnancy. METHOD OF STUDY: In this study, IL-33 signaling was inhibited using two different methods: intraperitoneal administration of recombinant ST2 (which acts as a decoy receptor for IL-33) and administration of a specific IL-33 neutralizing antibody. Maternal blood pressure, uterine artery resistance index, renal and placental oxidative stress, cNK, TH17, and TReg populations, various cytokines, and pre-proendothelin-1 levels were measured. RESULTS: IL-33 signaling inhibition increased maternal blood pressure, uterine artery resistance, placental and renal oxidative stress. IL-33 signaling inhibition also increased placental cNK and TH17 and renal TH17 cells while decreasing placental TReg populations. IL-33 neutralization increased circulating cNK and TH17s and decreased circulating TRegs in addition to increasing pre-proendothelin-1 levels. CONCLUSIONS: Data presented in this study demonstrate a role for IL-33 signaling in controlling vascular function and maternal blood pressure during pregnancy possibly by mediating innate and adaptive immune inflammatory responses, identifying the IL-33 signaling pathway as a potential therapeutic target for managing preeclampsia.


Sujet(s)
Interleukine-33 , Pré-éclampsie , Transduction du signal , Femelle , Grossesse , Interleukine-33/métabolisme , Pré-éclampsie/immunologie , Animaux , Rats , Rat Sprague-Dawley , Cellules Th17/immunologie , Modèles animaux de maladie humaine , Lymphocytes T régulateurs/immunologie , Humains , Stress oxydatif , Placenta/immunologie , Placenta/métabolisme , Pression sanguine , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme
4.
J Cell Mol Med ; 28(12): e18468, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923705

RÉSUMÉ

IL33 plays an important role in cancer. However, the role of liver cancer remains unclear. Open-accessed data was obtained from the Cancer Genome Atlas, Xena, and TISCH databases. Different algorithms and R packages are used to perform various analyses. Here, in our comprehensive study on IL33 in HCC, we observed its differential expression across cancers, implicating its role in cancer development. The single-cell analysis highlighted its primary expression in endothelial cells, unveiling correlations within the HCC microenvironment. Also, the expression level of IL33 was correlated with patients survival, emphasizing its potential prognostic value. Biological enrichment analyses revealed associations with stem cell division, angiogenesis, and inflammatory response. IL33's impact on the immune microenvironment showcased correlations with diverse immune cells. Genomic features and drug sensitivity analyses provided insights into IL33's broader implications. In a pan-cancer context, IL33 emerged as a potential tumour-inhibitor, influencing immune-related molecules. This study significantly advances our understanding of IL33 in cancer biology. IL33 exhibited differential expression across cancers, particularly in endothelial cells within the HCC microenvironment. IL33 is correlated with the survival of HCC patients, indicating potential prognostic value and highlighting its broader implications in cancer biology.


Sujet(s)
Carcinome hépatocellulaire , Régulation de l'expression des gènes tumoraux , Interleukine-33 , Tumeurs du foie , Microenvironnement tumoral , Humains , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/mortalité , Tumeurs du foie/immunologie , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/mortalité , Tumeurs du foie/métabolisme , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Pronostic , Interleukine-33/métabolisme , Interleukine-33/génétique , Marqueurs biologiques tumoraux/génétique
5.
Cell Commun Signal ; 22(1): 318, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858740

RÉSUMÉ

OBJECTIVES: Interleukin 33 (IL-33) is a crucial inflammatory factor that functions as an alarm signal in endometriosis (EMs). Epithelial-mesenchymal transition (EMT), a process related to inflammatory signals, intracellular reactive oxygen species (ROS) production, and lipid peroxidation, have been proposed as potential mechanisms that contribute to the development and progression of EMs. IL-33 is highly upregulated in the ectopic milieu. Moreover, ectopic endometrial cells constitutively express interleukin-33 receptor ST2 (IL-33R). However, the role of IL-33/ST2 in the EMT of EMs remains largely unknown. In this study, we aimed to mechanistically determine the role of IL-33/ST2 in EMs-associated fibrosis. MATERIALS AND METHODS: We established a non-lethal oxidative stress model to explore the conditions that trigger IL-33 induction. We performed α-smooth muscle actin (α-SMA) protein detection, cell counting kit-8 (CCK-8) assays, and scratch assays to analyze the impact of IL-33 on primary endometrial stromal cells (ESCs) proliferation and invasion. Clinical samples from patients with or without EMs were subjected to immunohistochemical (IHC) and and immunofluorescence(IF) staining to assess the clinical relevance of IL-33 receptor ST2 and EMT-related proteins. Furthermore, we used the ectopic human endometrial epithelial cell line 12Z and normal human epithelial cell line EEC to evaluate the effects of IL-33 on Wnt/ß-catenin signaling. The effect of IL-33 on EMT-associated fibrosis was validated in vivo by intraperitoneal injections of IL-33 and antiST2. RESULTS: We observed that ectopic milieu, characterized by ROS, TGF-ß1, and high level of estrogen, triggers the secretion of IL-33 from ectopic ESCs. Ectopic endometrial lesions exhibited higher level of fibrotic characteristics and ST2 expression than that in the normal endometrium. Exogenous recombinant human (rhIL-33) enhanced ESC migration and survival. Similarly, 12Z cells displayed a higher degree of EMT characteristics with elevated expression of CCN4 and Fra-1, downstream target genes of the WNT/ß-catenin pathway, than that observed in EECs. Conversely, blocking IL-33 with neutralizing antibodies, knocking down ST2 or ß-catenin with siRNA, and ß-catenin dephosphorylation abolished its effects on EMT promotion. In vivo validation demonstrated that IL-33 significantly promotes EMs-related fibrosis through the activation of Wnt/ß-catenin signaling. CONCLUSION: Our data strongly support the vital role of the IL-33/ST2 pathway in EMs-associated fibrosis and emphasize the importance of the EMT in the pathophysiology of fibrosis. Targeting the IL-33/ST2/Wnt/ß-catenin axis may hold promise as a feasible therapeutic approach for controlling fibrosis in EMs.


Sujet(s)
Endométriose , Transition épithélio-mésenchymateuse , Protéine-1 analogue au récepteur de l'interleukin-1 , Interleukine-33 , bêta-Caténine , Femelle , Endométriose/métabolisme , Endométriose/anatomopathologie , Endométriose/génétique , Interleukine-33/métabolisme , Interleukine-33/génétique , Transition épithélio-mésenchymateuse/génétique , Humains , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Protéine-1 analogue au récepteur de l'interleukin-1/génétique , bêta-Caténine/métabolisme , Animaux , Phosphorylation , Souris , Endomètre/anatomopathologie , Endomètre/métabolisme , Adulte , Prolifération cellulaire , Mouvement cellulaire , Transduction du signal
6.
Nat Commun ; 15(1): 5226, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38890291

RÉSUMÉ

IL-33 plays a significant role in inflammation, allergy, and host defence against parasitic helminths. The model gastrointestinal nematode Heligmosomoides polygyrus bakeri secretes the Alarmin Release Inhibitor HpARI2, an effector protein that suppresses protective immune responses and asthma in its host by inhibiting IL-33 signalling. Here we reveal the structure of HpARI2 bound to mouse IL-33. HpARI2 contains three CCP-like domains, and we show that it contacts IL-33 primarily through the second and third of these. A large loop which emerges from CCP3 directly contacts IL-33 and structural comparison shows that this overlaps with the binding site on IL-33 for its receptor, ST2, preventing formation of a signalling complex. Truncations of HpARI2 which lack the large loop from CCP3 are not able to block IL-33-mediated signalling in a cell-based assay and in an in vivo female mouse model of asthma. This shows that direct competition between HpARI2 and ST2 is responsible for suppression of IL-33-dependent responses.


Sujet(s)
Asthme , Protéines d'helminthes , Protéine-1 analogue au récepteur de l'interleukin-1 , Interleukine-33 , Nematospiroides dubius , Animaux , Interleukine-33/métabolisme , Interleukine-33/composition chimique , Nematospiroides dubius/immunologie , Protéines d'helminthes/métabolisme , Protéines d'helminthes/composition chimique , Protéines d'helminthes/immunologie , Souris , Femelle , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Asthme/immunologie , Asthme/métabolisme , Humains , Transduction du signal , Infections à Strongylida/immunologie , Infections à Strongylida/parasitologie , Infections à Strongylida/métabolisme , Liaison aux protéines , Modèles animaux de maladie humaine , Sites de fixation , Souris de lignée BALB C , Souris de lignée C57BL
7.
Science ; 384(6703): eadl0370, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38935708

RÉSUMÉ

Innate lymphoid cells (ILCs) and adaptive T lymphocytes promote tissue homeostasis and protective immune responses. Their production depends on the transcription factor GATA3, which is further elevated specifically in ILC2s and T helper 2 cells to drive type-2 immunity during tissue repair, allergic disorders, and anti-helminth immunity. The control of this crucial up-regulation is poorly understood. Using CRISPR screens in ILCs we identified previously unappreciated myocyte-specific enhancer factor 2d (Mef2d)-mediated regulation of GATA3-dependent type-2 lymphocyte differentiation. Mef2d-deletion from ILC2s and/or T cells specifically protected against an allergen lung challenge. Mef2d repressed Regnase-1 endonuclease expression to enhance IL-33 receptor production and IL-33 signaling and acted downstream of calcium-mediated signaling to translocate NFAT1 to the nucleus to promote type-2 cytokine-mediated immunity.


Sujet(s)
Facteur de transcription GATA-3 , Immunité innée , Interleukine-33 , Facteurs de transcription MEF2 , Facteurs de transcription NFATC , Pneumopathie infectieuse , Lymphocytes auxiliaires Th2 , Animaux , Souris , Facteurs de transcription MEF2/métabolisme , Facteurs de transcription MEF2/génétique , Lymphocytes auxiliaires Th2/immunologie , Interleukine-33/métabolisme , Facteurs de transcription NFATC/métabolisme , Pneumopathie infectieuse/immunologie , Facteur de transcription GATA-3/métabolisme , Facteur de transcription GATA-3/génétique , Souris de lignée C57BL , Différenciation cellulaire , Signalisation calcique , Hypersensibilité/immunologie , Poumon/immunologie , Allergènes/immunologie , Lymphocytes/immunologie , Protéine-1 analogue au récepteur de l'interleukin-1
8.
Cytokine ; 180: 156672, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38852492

RÉSUMÉ

BACKGROUND: Despite recent advances in therapeutic regimens, the prognosis of acute myeloid leukemia (AML) remains poor. Following our previous finding that interleukin-33 (IL-33) promotes cell survival along with activated NF-κB in AML, we further investigated the role of NF-κB during leukemia development. METHODS: Flow cytometry was performed to value the apoptosis and proliferation. qRT-PCR and western blot were performed to detect the expression of IL-6, active caspase 3, BIRC2, Bcl-2, and Bax, as well as activated NF-κB p65 and AKT. Finally, xenograft mouse models and AML patient samples were used to verify the findings observed in AML cell lines. RESULTS: IL-33-mediated NF-κB activation in AML cell lines contributes to a reduction in apoptosis, an increase in proliferation rate as well as a decrease in drug sensitivity, which were reversed by NF-κB inhibitor, Bay-117085. Moreover, IL-33 decreased the expression of active caspase-3 while increasing the levels of BIRC2, Bcl-2, and Bax, and these effects were blocked by Bay-117085. Additionally, NF-κB activation induced by IL-33 increases the production of IL-6 and autocrine activation of AKT. Co-culture of bone marrow stroma with AML cells resulted in increased IL-33 expression by leukemia cells, along with decreased apoptosis level and reduced drug sensitivity. Finally, we confirmed the in vivo pro-tumor effect mediated by IL-33/ NF-κB axis using a xenograft model of AML. CONCLUSION: Our data indicate that IL-33/IL1RL1-dependent signaling contributes to AML cell activation of NF-κB, which in turn causes autocrine IL-6-induced activation of pAKT, supporting IL-33/NF-κB/pAKT as a potential target for AML therapy.


Sujet(s)
Apoptose , Résistance aux médicaments antinéoplasiques , Interleukine-33 , Leucémie aigüe myéloïde , Facteur de transcription NF-kappa B , Humains , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/traitement médicamenteux , Apoptose/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Animaux , Interleukine-33/métabolisme , Souris , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines proto-oncogènes c-akt/métabolisme
10.
Nat Commun ; 15(1): 5056, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38871792

RÉSUMÉ

Type 2 innate lymphoid cells (ILC2) initiate early allergic inflammation in the lung, but the factors that promote subsequent resolution of type 2 inflammation and prevent prolonged ILC2 activation are not fully known. Here we show that SLAM-family receptors (SFR) play essential roles in this process. We demonstrate dynamic expression of several SFRs on ILC2s during papain-induced type 2 immunity in mice. SFR deficiency exacerbates ILC2-driven eosinophil infiltration in the lung, and results in a significant increase in IL-13 production by ILC2s exclusively in mediastinal lymph nodes (MLN), leading to increased dendritic cell (DC) and TH2 cell numbers. In MLNs, we observe more frequent interaction between ILC2s and bystander T cells, with T cell-expressed SFRs (especially SLAMF3 and SLAMF5) acting as self-ligands to suppress IL-13 production by ILC2s. Mechanistically, homotypic engagement of SFRs at the interface between ILC2s and T cells delivers inhibitory signaling primarily mediated by SHIP-1. This prevents activation of NF-κB, driven by IL-7 and IL-33, two major drivers of ILC2-mediated type 2 immunity. Thus, our study shows that an ILC2-DC-TH2 regulatory axis may promote the resolution of pulmonary type 2 immune responses, and highlights SLAMF3/SLAMF5 as potential therapeutic targets for ameliorating type 2 immunity.


Sujet(s)
Immunité innée , Inflammation , Poumon , Lymphocytes , Souris de lignée C57BL , Famille des molécules de signalisation de l'activation des lymphocytes , Animaux , Souris , Inflammation/immunologie , Inflammation/métabolisme , Lymphocytes/immunologie , Lymphocytes/métabolisme , Poumon/immunologie , Poumon/anatomopathologie , Famille des molécules de signalisation de l'activation des lymphocytes/métabolisme , Famille des molécules de signalisation de l'activation des lymphocytes/génétique , Papaïne , Lymphocytes auxiliaires Th2/immunologie , Interleukine-13/métabolisme , Interleukine-13/immunologie , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/métabolisme , Interleukine-33/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris knockout , Transduction du signal , Facteur de transcription NF-kappa B/métabolisme
11.
Proc Natl Acad Sci U S A ; 121(22): e2310864121, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38781213

RÉSUMÉ

IL-22 plays a critical role in defending against mucosal infections, but how IL-22 production is regulated is incompletely understood. Here, we show that mice lacking IL-33 or its receptor ST2 (IL-1RL1) were more resistant to Streptococcus pneumoniae lung infection than wild-type animals and that single-nucleotide polymorphisms in IL33 and IL1RL1 were associated with pneumococcal pneumonia in humans. The effect of IL-33 on S. pneumoniae infection was mediated by negative regulation of IL-22 production in innate lymphoid cells (ILCs) but independent of ILC2s as well as IL-4 and IL-13 signaling. Moreover, IL-33's influence on IL-22-dependent antibacterial defense was dependent on housing conditions of the mice and mediated by IL-33's modulatory effect on the gut microbiota. Collectively, we provide insight into the bidirectional crosstalk between the innate immune system and the microbiota. We conclude that both genetic and environmental factors influence the gut microbiota, thereby impacting the efficacy of antibacterial immune defense and susceptibility to pneumonia.


Sujet(s)
Immunité innée , Protéine-1 analogue au récepteur de l'interleukin-1 , , Interleukine-33 , Interleukines , Streptococcus pneumoniae , Animaux , Interleukine-33/immunologie , Interleukine-33/génétique , Interleukine-33/métabolisme , Interleukines/métabolisme , Interleukines/immunologie , Interleukines/génétique , Souris , Streptococcus pneumoniae/immunologie , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Protéine-1 analogue au récepteur de l'interleukin-1/génétique , Protéine-1 analogue au récepteur de l'interleukin-1/immunologie , Humains , Souris knockout , Microbiote/immunologie , Souris de lignée C57BL , Pneumonie à pneumocoques/immunologie , Pneumonie à pneumocoques/microbiologie , Microbiome gastro-intestinal/immunologie , Lymphocytes/immunologie , Lymphocytes/métabolisme , Polymorphisme de nucléotide simple
12.
Nat Commun ; 15(1): 4099, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38816352

RÉSUMÉ

Chronic inflammation is a major cause of cancer worldwide. Interleukin 33 (IL-33) is a critical initiator of cancer-prone chronic inflammation; however, its induction mechanism by environmental causes of chronic inflammation is unknown. Herein, we demonstrate that Toll-like receptor (TLR)3/4-TBK1-IRF3 pathway activation links environmental insults to IL-33 induction in the skin and pancreas inflammation. An FDA-approved drug library screen identifies pitavastatin to effectively suppress IL-33 expression by blocking TBK1 membrane recruitment/activation through the mevalonate pathway inhibition. Accordingly, pitavastatin prevents chronic pancreatitis and its cancer sequela in an IL-33-dependent manner. The IRF3-IL-33 axis is highly active in chronic pancreatitis and its associated pancreatic cancer in humans. Interestingly, pitavastatin use correlates with a significantly reduced risk of chronic pancreatitis and pancreatic cancer in patients. Our findings demonstrate that blocking the TBK1-IRF3-IL-33 signaling axis suppresses cancer-prone chronic inflammation. Statins present a safe and effective prophylactic strategy to prevent chronic inflammation and its cancer sequela.


Sujet(s)
Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Facteur-3 de régulation d'interféron , Interleukine-33 , Tumeurs du pancréas , Protein-Serine-Threonine Kinases , Quinoléines , Transduction du signal , Interleukine-33/métabolisme , Animaux , Facteur-3 de régulation d'interféron/métabolisme , Humains , Tumeurs du pancréas/prévention et contrôle , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Souris , Protein-Serine-Threonine Kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Inflammation/prévention et contrôle , Inflammation/métabolisme , Pancréatite chronique/prévention et contrôle , Pancréatite chronique/métabolisme , Récepteur de type Toll-3/métabolisme , Souris de lignée C57BL , Récepteur de type Toll-4/métabolisme , Acide mévalonique/métabolisme , Mâle , Femelle , Souris knockout
13.
Life Sci ; 348: 122701, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38724005

RÉSUMÉ

Hyperinflammatory responses are pivotal in the cardiomyocyte senescence pathophysiology, with IL33 serving as a crucial pro-inflammatory mediator. Our previous findings highlighted RND3's suppressive effect on IL33 expression. This study aims to explore the role of RND3 in IL33/ST2 signaling activation and in cardiomyocyte senescence. Intramyocardial injection of exogenous IL33 reduces the ejection fraction and fractional shortening of rats, inducing the appearance of senescence-associated secretory phenotype (SASP) in myocardial tissues. Recombinant IL33 treatment of AC16 cardiomyocytes significantly upregulated expression of SASP factors like IL1α, IL6, and MCP1, and increased the p-p65/p65 ratio and proportions of SA-ß-gal and γH2AX-positive cells. NF-κB inhibitor pyrrolidinedithiocarbamate ammonium (PDTC) and ST2 antibody astegolimab treatments mitigated above effects. RND3 gene knockout H9C2 cardiomyocytes using CRISPR/Cas9 technology upregulated IL33, ST2L, IL1α, IL6, and MCP1 levels, decreased sST2 levels, and increased SA-ß-gal and γH2AX-positive cells. A highly possibility of binding between RND3 and IL33 proteins was showed by molecular docking and co-immunoprecipitation, and loss of RND3 attenuated ubiquitination mediated degradation of IL33; what's more, a panel of ubiquitination regulatory genes closely related to RND3 were screened using qPCR array. In contrast, RND3 overexpression in rats by injection of AAV9-CMV-RND3 particles inhibited IL33, ST2L, IL1α, IL6, and MCP1 expression in cardiac tissues, decreased serum IL33 levels, and increased sST2 levels. These results suggest that RND3 expression in cardiomyocytes modulates cell senescence by inhibiting the IL33/ST2/NF-κB signaling pathway, underscoring its potential as a therapeutic target in cardiovascular senescence.


Sujet(s)
Vieillissement de la cellule , Interleukine-33 , Myocytes cardiaques , Transduction du signal , Animaux , Mâle , Rats , Lignée cellulaire , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Interleukine-33/métabolisme , Myocytes cardiaques/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Rat Sprague-Dawley , Récepteurs à l'interleukine-1 , Protéines G rho/métabolisme , Protéines G rho/génétique
14.
Immunity ; 57(6): 1274-1288.e6, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38821053

RÉSUMÉ

Severe asthma and sinus disease are consequences of type 2 inflammation (T2I), mediated by interleukin (IL)-33 signaling through its membrane-bound receptor, ST2. Soluble (s)ST2 reduces available IL-33 and limits T2I, but little is known about its regulation. We demonstrate that prostaglandin E2 (PGE2) drives production of sST2 to limit features of lung T2I. PGE2-deficient mice display diminished sST2. In humans with severe respiratory T2I, urinary PGE2 metabolites correlate with serum sST2. In mice, PGE2 enhanced sST2 secretion by mast cells (MCs). Mice lacking MCs, ST2 expression by MCs, or E prostanoid (EP)2 receptors by MCs showed reduced sST2 lung concentrations and strong T2I. Recombinant sST2 reduced T2I in mice lacking PGE2 or ST2 expression by MCs back to control levels. PGE2 deficiency also reversed the hyperinflammatory phenotype in mice lacking ST2 expression by MCs. PGE2 thus suppresses T2I through MC-derived sST2, explaining the severe T2I observed in low PGE2 states.


Sujet(s)
Dinoprostone , Protéine-1 analogue au récepteur de l'interleukin-1 , Interleukine-33 , Poumon , Mastocytes , Souris knockout , Animaux , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Protéine-1 analogue au récepteur de l'interleukin-1/génétique , Mastocytes/immunologie , Mastocytes/métabolisme , Dinoprostone/métabolisme , Souris , Interleukine-33/métabolisme , Humains , Poumon/immunologie , Poumon/métabolisme , Poumon/anatomopathologie , Asthme/immunologie , Asthme/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Souris de lignée C57BL , Inflammation/immunologie , Femelle , Mâle , Transduction du signal , Pneumopathie infectieuse/immunologie , Pneumopathie infectieuse/métabolisme
15.
Cell Death Dis ; 15(5): 356, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38778059

RÉSUMÉ

IL-33 is a danger signal that binds to its receptor ST2L to promote tumor progression. This study identifies the IL-33/ST2L positive-feedback loop and the trafficking of ST2L membrane presentation in macrophages that contribute to lung tumor progression. Mechanistically, IL-33 induces ST2L upregulation by activating NF-κB, which binds to the promoter region of the ST2L gene. Moreover, Rab37, a small GTPase involved in membrane trafficking, mediates ST2L trafficking to the plasma membrane of M2 macrophages. This IL-33/NF-κB/ST2L/Rab37 axis promotes positive-feedback loops that enhance ST2L expression and membrane trafficking in M2 macrophages. Notably, neutralizing antibodies against IL-33 or ST2L block NF-κB activity, suppress M2 macrophage polarization, and synergistically inhibit tumor growth when combined with cisplatin treatment in vitro/vivo. Clinically, Rab37+/ST2L+/CD206+ tumor-infiltrating M2 macrophages correlate with advanced-stage lung cancer patients with poor response to chemotherapy. These findings unveil a positive-feedback mechanism and provide a basis for IL-33/ST2L-targeting therapy for cancer.


Sujet(s)
Interleukine-33 , Tumeurs du poumon , Macrophages , Facteur de transcription NF-kappa B , Protéines G rab , Interleukine-33/métabolisme , Interleukine-33/génétique , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Facteur de transcription NF-kappa B/métabolisme , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Animaux , Protéines G rab/métabolisme , Protéines G rab/génétique , Souris , Rétrocontrôle physiologique , Lignée cellulaire tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Femelle
16.
Immunology ; 172(4): 653-668, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38786548

RÉSUMÉ

The mechanisms underlying the development of steroid resistance in asthma remain unclear. To establish whether as well as the mechanisms by which the activation of Janus kinases (JAKs) is involved in the development of steroid resistance in asthma, murine steroid-resistant models of the proliferation of group 2 innate lymphoid cells (ILC2s) in vitro and asthmatic airway inflammation in vivo were analysed. ILC2s in the lungs of BALB/c mice were sorted and then incubated with IL-33, thymic stromal lymphopoietin (TSLP), and/or IL-7 with or without dexamethasone (10 nM), the pan-JAK inhibitor, delgocitinib (1-10 000 nM), and/or the Bcl-xL inhibitor, navitoclax (1-100 nM), followed by the detection of viable and apoptotic cells. The anti-apoptotic factor, Bcl-xL was detected in ILC2s by flow cytometry. As a steroid-resistant asthma model, ovalbumin (OVA)-sensitized BALB/c mice were intratracheally challenged with OVA at a high dose of 500 µg four times. Dexamethasone (1 mg/kg, i.p.), delgocitinib (3-30 mg/kg, p.o.), or navitoclax (30 mg/kg, p.o.) was administered during the challenges. Cellular infiltration into the lungs was analysed by flow cytometry. Airway remodelling was histologically evaluated. The following results were obtained. (1) Cell proliferation concomitant with a decrease in apoptotic cells was induced when ILC2s were cultured with TSLP and/or IL-7, and was potently inhibited by dexamethasone. In contrast, when the culture with TSLP and IL-7 was performed in the presence of IL-33, the proliferative response exhibited steroid resistance. Steroid-resistant ILC2 proliferation was suppressed by delgocitinib in a concentration-dependent manner. (2) The culture with IL-33, TSLP, and IL-7 induced the overexpression of Bcl-xL, which was clearly inhibited by delgocitinib, but not by dexamethasone. When ILC2s were treated with navitoclax, insensitivity to dexamethasone was significantly cancelled. (3) The development of airway remodelling and the infiltration of ILC2s into the lungs in the asthma model were not suppressed by dexamethasone, but were dose-dependently inhibited by delgocitinib. Combination treatment with dexamethasone and either delgocitinib or navitoclax synergistically suppressed these responses. Therefore, JAKs appear to play significant roles in the induction of steroid resistance by up-regulating Bcl-xL in ILC2s. The inhibition of JAKs and Bcl-xL has potential as pharmacotherapy for steroid-resistant asthma, particularly that mediated by ILC2s.


Sujet(s)
Asthme , Dexaméthasone , Résistance aux substances , Immunité innée , Janus kinases , Lymphocytes , Souris de lignée BALB C , Protéine bcl-X , Animaux , Asthme/traitement médicamenteux , Asthme/immunologie , Asthme/métabolisme , Protéine bcl-X/métabolisme , Lymphocytes/immunologie , Lymphocytes/métabolisme , Lymphocytes/effets des médicaments et des substances chimiques , Souris , Dexaméthasone/pharmacologie , Dexaméthasone/usage thérapeutique , Immunité innée/effets des médicaments et des substances chimiques , Janus kinases/métabolisme , Poumon/immunologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Femelle , Cytokines/métabolisme , Modèles animaux de maladie humaine , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Interleukine-33/métabolisme , Lymphopoïétine stromale thymique , Sulfonamides/pharmacologie
17.
Cell Immunol ; 401-402: 104828, 2024.
Article de Anglais | MEDLINE | ID: mdl-38759328

RÉSUMÉ

Renal fibrosis is a common pathway of chronic kidney disease (CKD) progression involving primary kidney injury and kidney diseases. Group 2 innate lymphoid cells (ILC2s) mediate type 2 immune responses irrespective of antigen presentation and play a reno-protective role in kidney injury and disease. In the present study, we observed a decrease in kidney-resident ILC2s in CKD and found that enrichment of ILC2s in the kidney ameliorates renal fibrosis. In CKD kidney, ILC2s preferentially produced IL-13 over IL-5 in response to IL-33 stimulation, regardless of ST2L expression. Moreover, GATA3 expression was decreased in ILC2s, and T-bet+ ILC1s and RORγt+ ILC3s were increased in CKD kidney. Adoptive transfer of kidney ILC2s into adenine-induced CKD model mouse improved renal function and fibrosis. Renal fibroblasts cultured with IL33-activated kidney ILC2s suppressed myofibroblast trans-differentiation through Acta2 and Fn-1 regulation. These results suggest that kidney ILC2s prevent CKD progression via improvement of renal fibrosis. Our findings also suggest that ILC2s may contribute to the development of new therapeutic agents and strategies for tissue fibroses.


Sujet(s)
Adénine , Fibrose , Immunité innée , Rein , Lymphocytes , Souris de lignée C57BL , Insuffisance rénale chronique , Animaux , Insuffisance rénale chronique/immunologie , Insuffisance rénale chronique/induit chimiquement , Souris , Lymphocytes/immunologie , Lymphocytes/métabolisme , Adénine/pharmacologie , Adénine/analogues et dérivés , Rein/anatomopathologie , Rein/immunologie , Mâle , Modèles animaux de maladie humaine , Interleukine-33/métabolisme , Interleukine-13/métabolisme
18.
J Biol Chem ; 300(6): 107366, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38750790

RÉSUMÉ

Host anti-inflammatory responses are critical for the progression of visceral leishmaniasis, and the pleiotropic cytokine interleukin (IL)-33 was found to be upregulated in infection. Here, we documented that IL-33 induction is a consequence of elevated cAMP-mediated exchange protein activated by cAMP (EPAC)/calcineurin-dependent signaling and essential for the sustenance of infection. Leishmania donovani-infected macrophages showed upregulation of IL-33 and its neutralization resulted in decreased parasite survival and increased inflammatory responses. Infection-induced cAMP was involved in IL-33 production and of its downstream effectors PKA and EPAC, only the latter was responsible for elevated IL-33 level. EPAC initiated Rap-dependent phospholipase C activation, which triggered the release of intracellular calcium followed by calcium/calmodulin complex formation. Screening of calmodulin-dependent enzymes affirmed involvement of the phosphatase calcineurin in cAMP/EPAC/calcium/calmodulin signaling-induced IL-33 production and parasite survival. Activated calcineurin ensured nuclear localization of the transcription factors, nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha required for IL-33 transcription, and we further confirmed this by chromatin immunoprecipitation assay. Administering specific inhibitors of nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha in BALB/c mouse model of visceral leishmaniasis decreased liver and spleen parasite burden along with reduction in IL-33 level. Splenocyte supernatants of inhibitor-treated infected mice further documented an increase in tumor necrosis factor alpha and IL-12 level with simultaneous decrease of IL-10, thereby indicating an overall disease-escalating effect of IL-33. Thus, this study demonstrates that cAMP/EPAC/calcineurin signaling is crucial for the activation of IL-33 and in effect creates anti-inflammatory responses, essential for infection.


Sujet(s)
Calcineurine , AMP cyclique , Interleukine-33 , Leishmania donovani , Leishmaniose viscérale , Souris de lignée BALB C , Transduction du signal , Animaux , Calcineurine/métabolisme , AMP cyclique/métabolisme , Souris , Leishmaniose viscérale/immunologie , Leishmaniose viscérale/métabolisme , Leishmaniose viscérale/parasitologie , Interleukine-33/métabolisme , Macrophages/métabolisme , Macrophages/parasitologie , Facteurs d'échange de nucléotides guanyliques/métabolisme , Facteurs d'échange de nucléotides guanyliques/génétique
19.
Neuropharmacology ; 253: 109982, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38701943

RÉSUMÉ

Perioperative neurocognitive disorders (PND) are cognitive dysfunctions that usually occur in elderly patients after anesthesia and surgery. Microglial overactivation is a key underlying mechanism. Interleukin-33 (IL-33) is a member of the IL-1 family that orchestrates microglial function. In the present study, we explored how IL-33, which regulates microglia, contributes to cognitive improvement in a male mouse model of PND. An exploratory laparotomy was performed to establish a PND model. The expression levels of IL-33 and its receptor ST2 were evaluated using Western blot. IL-33/ST2 secretion, microglial density, morphology, phagocytosis of synapse, and proliferation, and dystrophic microglia were assessed using immunofluorescence. Synaptic plasticity was measured using Golgi staining and long-term potentiation. The Morris water maze and open field test were used to evaluate cognitive function and anxiety. Hippocampal expression of IL-33 and ST2 were elevated on postoperative day 3. We confirmed that IL-33 was secreted by astrocytes and neurons, whereas ST2 mainly colocalized with microglia. IL-33 treatment induced microgliosis after anesthesia and surgery. These microglia had larger soma sizes and shorter and fragmented branches. Compared to the Surgery group, IL-33 treatment reduced the synaptic phagocytosis of microglia and increased microglial proliferation and dystrophic microglia. IL-33 treatment also reversed the impaired synaptic plasticity and cognitive function caused by anesthesia and surgery. In conclusion, these results indicate that IL-33 plays a key role in regulating microglial state and synaptic phagocytosis in a PND mouse model. IL-33 treatment has a therapeutic potential for improving cognitive dysfunction in PND.


Sujet(s)
Interleukine-33 , Souris de lignée C57BL , Microglie , Animaux , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Interleukine-33/métabolisme , Mâle , Souris , Plasticité neuronale/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/anatomopathologie , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/physiologie , Complications post-opératoires cognitives/métabolisme , Phagocytose/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Troubles neurocognitifs/métabolisme , Troubles neurocognitifs/traitement médicamenteux , Modèles animaux de maladie humaine , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme
20.
Clin. transl. oncol. (Print) ; 26(4): 924-935, Abr. 2024. graf, ilus
Article de Anglais | IBECS | ID: ibc-VR-55

RÉSUMÉ

Purpose: Non-small cell lung cancer (NSCLC) is a complex disease that remains a major public health concern worldwide. One promising avenue for NSCLC treatment is the targeting of transcription factors that regulate key pathways involved in cancer progression. In this study, we investigated the role of the transcription factor ZNF263 in NSCLC and its impact on the regulation of IL33, apoptosis, and autophagy. Methods: Levels of ZNF263 in tissues and cell lines were identified, after which the effects of its knockdown on cellular malignant behaviors, apoptosis and autophagy were assessed. Based on bioinformatics analysis, ZNF263 was found to bind to IL33 promoter, their mutual relationship was confirmed, as well as the role of IL33 in the regulation of ZNF263. The involvement of ZNF263 in the growth of xenograft tumors was assessed using tumor-bearing nude mouse models. Results: Experimental results revealed that ZNF263 was upregulated in NSCLC tissue samples and cell lines. Its expression level is positively correlated with cellular malignant behaviors. We further demonstrated that ZNF263 upregulated IL33 expression, which, in turn, promoted the proliferation and migration, inhibited apoptosis and autophagy in NSCLC cells. Furthermore, ZNF263 knockdown reduced the growth of xenograft tumors in nude mice. Conclusion: This finding suggests that the inhibition of ZNF263 or IL33 may represent a novel therapeutic strategy for NSCLC. Importantly, our results highlight the crucial role of transcription factors in NSCLC and their potential as therapeutic targets.(AU)


Sujet(s)
Humains , Mâle , Femelle , Carcinome pulmonaire non à petites cellules , Autophagie , Protéines de liaison à l'ADN , Interleukine-33/métabolisme , Interleukine-33/usage thérapeutique , Tumeurs du poumon/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE