Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.708
Filtrer
1.
Sci Adv ; 10(27): eado2365, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38959302

RÉSUMÉ

Pityriasis rubra pilaris (PRP) is a rare inflammatory skin disease with a poorly understood pathogenesis. Through a molecularly driven precision medicine approach and an extensive mechanistic pathway analysis in PRP skin samples, compared to psoriasis, atopic dermatitis, healed PRP, and healthy controls, we identified IL-1ß as a key mediator, orchestrating an NF-κB-mediated IL-1ß-CCL20 axis, including activation of CARD14 and NOD2. Treatment of three patients with the IL-1 antagonists anakinra and canakinumab resulted in rapid clinical improvement and reversal of the PRP-associated molecular signature with a 50% improvement in skin lesions after 2 to 3 weeks. This transcriptional signature was consistent with in vitro stimulation of keratinocytes with IL-1ß. With the central role of IL-1ß underscoring its potential as a therapeutic target, our findings propose a redefinition of PRP as an autoinflammatory keratinization disorder. Further clinical trials are needed to validate the efficacy of IL-1ß antagonists in PRP.


Sujet(s)
Anticorps monoclonaux humanisés , Antagoniste du récepteur à l'interleukine-1 , Interleukine-1 bêta , Kératinocytes , Pityriasis rubra pilaire , Humains , Pityriasis rubra pilaire/traitement médicamenteux , Pityriasis rubra pilaire/anatomopathologie , Pityriasis rubra pilaire/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/antagonistes et inhibiteurs , Antagoniste du récepteur à l'interleukine-1/usage thérapeutique , Kératinocytes/métabolisme , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/anatomopathologie , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/pharmacologie , Mâle , Facteur de transcription NF-kappa B/métabolisme , Protéine adaptatrice de signalisation NOD2/métabolisme , Protéine adaptatrice de signalisation NOD2/génétique , Protéine adaptatrice de signalisation NOD2/antagonistes et inhibiteurs , Femelle , Protéines adaptatrices de signalisation CARD/métabolisme , Protéines adaptatrices de signalisation CARD/génétique , Peau/anatomopathologie , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Interleukine-1/antagonistes et inhibiteurs , Interleukine-1/métabolisme , Interleukine-1/génétique , Adulte d'âge moyen , Guanylate cyclase/métabolisme , Guanylate cyclase/antagonistes et inhibiteurs , Guanylate cyclase/génétique , Adulte , Transduction du signal/effets des médicaments et des substances chimiques , Protéines membranaires
2.
Biosci Rep ; 44(6)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38836325

RÉSUMÉ

Natural and synthetic polymeric materials, particularly soft and hard tissue replacements, are paramount in medicine. We prepared calcium-incorporated sulfonated polyether-ether ketone (SPEEK) polymer membranes for bone applications. The bioactivity was higher after 21 days of immersion in simulated body fluid (SBF) due to calcium concentration in the membrane. We present a new biomaterial healing system composed of calcium and sulfonated polyether ether ketone (Ca-SPEEK) that can function as a successful biomaterial without causing inflammation when tested on bone marrow cells. The Ca-SPEEK exhibited 13 ± 0.5% clot with low fibrin mesh formation compared to 21 ± 0.5% in SPEEK. In addition, the Ca-SPEEK showed higher protein adsorption than SPEEK membranes. As an inflammatory response, IL-1 and TNF-α in the case of Ca-SPEEK were lower than those for SPEEK. We found an early regulation of IL-10 in the case of Ca-SPEEK at 6 h, which may be attributed to the down-regulation of the inflammatory markers IL-1 and TNF-α. These results evidence the innovative bioactivity of Ca-SPEEK with low inflammatory response, opening venues for bone applications.


Sujet(s)
Matériaux biocompatibles , Cellules de la moelle osseuse , Calcium , Polymères , Facteur de nécrose tumorale alpha , Animaux , Souris , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Polymères/composition chimique , Polymères/pharmacologie , Calcium/métabolisme , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Benzophénones/composition chimique , Benzophénones/pharmacologie , Inflammation/traitement médicamenteux , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Cétones/composition chimique , Cétones/pharmacologie , Test de matériaux , Interleukine-1/métabolisme , Interleukine-10/métabolisme
3.
Biomolecules ; 14(6)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38927050

RÉSUMÉ

Schnitzler syndrome is a rare disorder characterized by a chronic urticarial rash associated with immunoglobulin M (IgM) monoclonal gammopathy. Schnitzler syndrome shares strong clinicopathologic similarities with monogenic IL-1-mediated autoinflammatory disorders and is now considered an acquired adult-onset autoinflammatory disease. The spectacular effect of interleukin-1 inhibitors demonstrates the key role of this cytokine in the pathogenesis of the disease. However, the physiopathology of Schnitzler syndrome remains elusive, and the main question regarding the relationship between autoinflammatory features and monoclonal gammopathy is still unanswered. The purpose of this narrative review is to describe what is currently known about the pathogenesis of this peculiar disease, as well as to address its diagnosis and management.


Sujet(s)
Syndrome de Schnitzler , Syndrome de Schnitzler/traitement médicamenteux , Syndrome de Schnitzler/diagnostic , Humains , Immunoglobuline M/immunologie , Interleukine-1/antagonistes et inhibiteurs , Interleukine-1/métabolisme
5.
Front Immunol ; 15: 1416162, 2024.
Article de Anglais | MEDLINE | ID: mdl-38895127

RÉSUMÉ

Introduction: IL6 signaling plays an important role in triggering labor and IL6 is an established biomarker of intrauterine infection/inflammation (IUI) driven preterm labor (PTL). The biology of IL6 during IUI at the maternal-fetal interface was investigated in samples from human subjects and non-human primates (NHP). Methods: Pregnant women with histologic chorioamnionitis diagnosed by placenta histology were recruited (n=28 term, n=43 for preterm pregnancies from 26-36 completed weeks of gestation). IUI was induced in Rhesus macaque by intraamniotic injection of lipopolysachharide (LPS, n=23). IL1 signaling was blocked using Anakinra (human IL-1 receptor antagonist, n=13), and Tumor necrosis factor (TNF) signaling was blocked by anti TNF-antibody (Adalimumab n=14). The blockers were given before LPS. All animals including controls (intraamniotic injection of saline n=27), were delivered 16h after LPS/saline exposure at about 80% gestation. Results: IUI induced a robust expression of IL6 mRNAs in the fetal membranes (chorion-amnion-decidua tissue) both in humans (term and preterm) and NHP. The major sources of IL6 mRNA expression were the amnion mesenchymal cells (AMC) and decidua stroma cells. Additionally, during IUI in the NHP, ADAM17 (a protease that cleaves membrane bound IL6 receptor (IL6R) to release a soluble form) and IL6R mRNA increased in the fetal membranes, and the ratio of IL6 and soluble forms of IL6R, gp130 increased in the amniotic fluid signifying upregulation of IL6 trans-signaling. Both IL1 and TNF blockade suppressed LPS-induced IL6 mRNAs in the AMC and variably decreased elements of IL6 trans-signaling. Discussion: These data suggest that IL1 and TNF blockers may be useful anti-inflammatory agents via suppression of IL6 signaling at the maternal-fetal interface.


Sujet(s)
Interleukine-6 , Macaca mulatta , Transduction du signal , Facteur de nécrose tumorale alpha , Femelle , Grossesse , Humains , Animaux , Interleukine-6/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Chorioamnionite/immunologie , Chorioamnionite/métabolisme , Chorioamnionite/médecine vétérinaire , Lipopolysaccharides/immunologie , Interleukine-1/métabolisme , Adulte , Travail obstétrical prématuré/immunologie , Travail obstétrical prématuré/métabolisme , Inflammation/immunologie , Inflammation/métabolisme , Antagoniste du récepteur à l'interleukine-1/métabolisme , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Placenta/métabolisme , Placenta/immunologie
6.
FASEB J ; 38(11): e23731, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38855909

RÉSUMÉ

Ca2+ permeation through TRPV4 in fibroblasts is associated with pathological matrix degradation. In human gingival fibroblasts, IL-1ß binding to its signaling receptor (IL-1R1) induces activation of extracellular regulated kinase (ERK) and MMP1 expression, processes that require Ca2+ flux across the plasma membrane. It is not known how IL-1R1, which does not conduct Ca2+, generates Ca2+ signals in response to IL-1. We examined whether TRPV4 mediates the Ca2+ fluxes required for ERK signaling in IL-1 stimulated gingival fibroblasts. TRPV4 was immunostained in fibroblasts of human gingival connective tissue and in focal adhesions of cultured mouse gingival fibroblasts. Human gingival fibroblasts treated with IL-1ß showed no change of TRPV4 expression but there was increased MMP1 expression. In mouse, gingival fibroblasts expressing TRPV4, IL-1 strongly increased [Ca2+]i. Pre-incubation of cells with IL-1 Receptor Antagonist blocked Ca2+ entry induced by IL-1 or the TRPV4 agonist GSK101. Knockout of TRPV4 or expression of a non-Ca2+-conducting TRPV4 pore-mutant or pre-incubation with the TRPV4 inhibitor RN1734, blocked IL-1-induced Ca2+ transients and expression of the mouse interstitial collagenase, MMP13. Treatment of mouse gingival fibroblasts with GSK101 phenocopied Ca2+ and ERK responses induced by IL-1; these responses were absent in TRPV4-null cells or cells expressing a non-conducting TRPV4 pore-mutant. Immunostained IL-1R1 localized with TRPV4 in adhesions within cell extensions. While TRPV4 immunoprecipitates analyzed by mass spectrometry showed no association with IL-1R1, TRPV4 associated with Src-related proteins and Src co-immunoprecipitated with TRPV4. Src inhibition reduced IL-1-induced Ca2+ responses. The functional linkage of TRPV4 with IL-1R1 expands its repertoire of innate immune signaling processes by mediating IL-1-driven Ca2+ responses that drive matrix remodeling in fibroblasts. Thus, inhibiting TRPV4 activity may provide a new pharmacological approach for blunting matrix degradation in inflammatory diseases.


Sujet(s)
Signalisation calcique , Fibroblastes , Gencive , Canaux cationiques TRPV , Canaux cationiques TRPV/métabolisme , Canaux cationiques TRPV/génétique , Animaux , Humains , Souris , Fibroblastes/métabolisme , Gencive/métabolisme , Gencive/cytologie , Calcium/métabolisme , Système de signalisation des MAP kinases , Cellules cultivées , Extracellular Signal-Regulated MAP Kinases/métabolisme , Interleukine-1/métabolisme , Interleukine-1/pharmacologie , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie
7.
Front Immunol ; 15: 1393096, 2024.
Article de Anglais | MEDLINE | ID: mdl-38855101

RÉSUMÉ

Introduction: Antibody production and the generation of memory B cells are regulated by T follicular helper (Tfh) and T follicular regulatory (Tfr) cells in germinal centers. However, the precise role of Tfr cells in controlling antibody production is still unclear. We have previously shown that both Tfh and Tfr cells express the IL-1R1 agonist receptor, whereas only Tfr cells express the IL-1R2 decoy and IL-1Ra antagonist receptors. We aimed to investigate the role of IL-1 receptors in the regulation of B cell responses by Tfh and Tfr. Methods: We generated mice with IL-1 receptors inactivated in Tfh or Tfr and measured antibody production and cell activation after immunisation. Results: While IL-1ß levels are increased in the draining lymph node after immunisation, antigen-specific antibody levels and cell phenotypes indicated that IL-1ß can activate both Tfh and Tfr cells through IL-1R1 stimulation. Surprisingly, expression of IL-1R2 and IL-1Ra on Tfr cells does not block IL-1 activation of Tfh cells, but rather prevents IL-1/IL-1R1-mediated early activation of Tfr cells. IL-1Rs also regulate the antibody response to autoantigens and its associated pathophysiology in an experimental lupus model. Discussion: Collectively, our results show that IL-1 inhibitory receptors expressed by Tfr cells prevent their own activation and suppressive function, thus licensing IL-1-mediated activation of Tfh cells after immunisation. Further mechanistic studies should unravel these complex interactions between IL-1ß and follicular helper and regulatory T cells and provide new avenues for therapeutic intervention.


Sujet(s)
Centre germinatif , Lymphocytes T auxiliaires folliculaires , Lymphocytes T régulateurs , Animaux , Centre germinatif/immunologie , Souris , Lymphocytes T auxiliaires folliculaires/immunologie , Lymphocytes T régulateurs/immunologie , Activation des lymphocytes/immunologie , Récepteur à l'interleukine-1 de type I/génétique , Récepteur à l'interleukine-1 de type I/immunologie , Souris de lignée C57BL , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/immunologie , Interleukine-1/métabolisme , Interleukine-1/immunologie , Récepteurs à l'interleukine-1/métabolisme , Récepteurs à l'interleukine-1/immunologie , Production d'anticorps/immunologie
8.
Int J Biol Sci ; 20(8): 3094-3112, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904012

RÉSUMÉ

Atopic dermatitis (AD) is a common inflammation skin disease that involves dysregulated interplay between immune cells and keratinocytes. Interleukin-38 (IL-38), a poorly characterized IL-1 family cytokine, its role and mechanism in the pathogenesis of AD is elusive. Here, we show that IL-38 is mainly secreted by epidermal keratinocytes and highly expressed in the skin and downregulated in AD lesions. We generated IL-38 keratinocyte-specific knockout mice (K14Cre/+-IL-38f/f ) and induced AD models by 2,4-dinitrofluorobenzene (DNFB). Unexpectedly, after treatment with DNFB, K14Cre/+-IL-38f/f mice were less susceptible to cutaneous inflammation of AD. Moreover, keratinocyte-specific deletion of IL-38 suppressed the migration of Langerhans cells (LCs) into lymph nodes which results in disturbed differentiation of CD4+T cells and decreased the infiltration of immune cells into AD lesions. LCs are a type of dendritic cell that reside specifically in the epidermis and regulate immune responses. We developed LC-like cells in vitro from mouse bone marrow (BM) and treated with recombined IL-38. The results show that IL-38 depended on IL-36R, activated the phosphorylated expression of IRAK4 and NF-κB P65 and upregulated the expression of CCR7 to promoting the migration of LCs, nevertheless, the upregulation disappeared with the addition of IL-36 receptor antagonist (IL-36RA), IRAK4 or NF-κB P65 inhibitor. Furthermore, after treatment with IRAK4 inhibitors, the experimental AD phenotypes were alleviated and so IRAK4 is considered a promising target for the treatment of inflammatory diseases. Overall, our findings indicated a potential pathway that IL-38 depends on IL-36R, leading to LCs migration to promote AD by upregulating CCR7 via IRAK4/NF-κB and implied the prevention and treatment of AD, supporting potential clinical utilization of IRAK4 inhibitors in AD treatment.


Sujet(s)
Mouvement cellulaire , Eczéma atopique , Cellules de Langerhans , Animaux , Eczéma atopique/métabolisme , Cellules de Langerhans/métabolisme , Souris , Souris knockout , Interleukine-1/métabolisme , Kératinocytes/métabolisme , 1-Fluoro-2,4-dinitro-benzène , Facteur de transcription NF-kappa B/métabolisme , Interleukines/métabolisme
9.
Phytomedicine ; 131: 155783, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38838402

RÉSUMÉ

BACKGROUND: Psoriasis, a chronic immune-mediated skin disease with pathological features such as aberrant differentiation of keratinocytes, dermal-epidermal inflammation, and angiogenesis. 2,3,5,4'-Tetrahydroxy stilbene 2-Ο-ß-d-glucoside (2354Glu) is a natural small molecule polyhydrostilbenes isolated from Polygonum multiglorum Thunb. The regulation of IL-36 subfamily has led to new pharmacologic strategies to reverse psoriasiform dermatitis. PURPOSE: Here we investigated the therapeutic potential of 2354Glu and elucidated the underlying mechanism in psoriasis. METHODS: The effects of 2354Glu on IL-36 signaling were assessed by psoriasiform in vivo, in vitro and ex vivo model. The in vivo mice model of psoriasis-like skin inflammation was established by applying imiquimod (IMQ), and the in vitro and ex vitro models were established by stimulating mouse primary keratinocyte, human keratinocytes cells (HaCaT) and ex vivo skin tissue isolated from the mice back with Polyinosine-polycytidylic acid (Poly(I:C)), IMQ, IL-36γ and Lipopolysaccharide (LPS) respectively. Moreover, NETs formation was inhibited by Cl-amidine to evaluate the effect of NETs in psoriatic mouse model. The effects of 2354Glu on skin inflammation were assessed by western blot, H&E, immunohistochemistry, immunofluorescence, enzyme-linked immunosorbent assay and real-time quantitative PCR. RESULTS: In Poly(I:C)-stimulated keratinocytes, the secretion of IL-36 was inhibited after treatment with 2354Glu, similar to the effects of TLR3, P2X7R and caspase-1 inhibitors. In aldara (imiquimod)-induced mice, 2354Glu (100 and 25 mg/kg) improved immune cell infiltration and hyperkeratosis in psoriasis by directly targeting IL-36 in keratinocytes through P2X7R-caspase-1. When treatment with 2354Glu (25 mg/kg) was insufficient to inhibit IL-36γ, NETs reduced pathological features and IL-36 signaling by interacting with keratinocytes to combat psoriasis like inflammation. CONCLUSION: These results indicated that NETs had a beneficial effect on psoriasiform dermatitis. 2354Glu alleviates psoriasis by directly targeting IL-36/P2X7R axis and NET formation, providing a potential candidate for the treatment of psoriasis.


Sujet(s)
Modèles animaux de maladie humaine , Glucosides , Imiquimod , Interleukine-1 , Psoriasis , Stilbènes , Animaux , Psoriasis/traitement médicamenteux , Glucosides/pharmacologie , Humains , Interleukine-1/métabolisme , Stilbènes/pharmacologie , Souris , Kératinocytes/effets des médicaments et des substances chimiques , Polygonum/composition chimique , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Souris de lignée BALB C , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Caspase-1/métabolisme
10.
Commun Biol ; 7(1): 756, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38907105

RÉSUMÉ

Tuberous sclerosis complex 2 (TSC2) crucially suppresses Rheb activity to prevent mTORC1 activation. However, mutations in TSC genes lead to mTORC1 overactivation, thereby causing various developmental disorders and cancer. Therefore, the discovery of novel Rheb inhibitors is vital to prevent mTOR overactivation. Here, we reveals that the anti-inflammatory cytokine IL-37d can bind to lysosomal Rheb and suppress its activity independent of TSC2, thereby preventing mTORC1 activation. The binding of IL-37d to Rheb switch-II subregion destabilizes the Rheb-mTOR and mTOR-S6K interactions, further halting mTORC1 signaling. Unlike TSC2, IL-37d is reduced under ethanol stimulation, which results in mitigating the suppression of lysosomal Rheb-mTORC1 activity. Consequently, the recombinant human IL-37d protein (rh-IL-37d) with a TAT peptide greatly improves alcohol-induced liver disorders by hindering Rheb-mTORC1 axis overactivation in a TSC2- independent manner. Together, IL-37d emerges as a novel Rheb suppressor independent of TSC2 to terminate mTORC1 activation and improve abnormal lipid metabolism in the liver.


Sujet(s)
Maladies alcooliques du foie , Complexe-1 cible mécanistique de la rapamycine , Protéine homologue de Ras enrichie dans le cerveau , Transduction du signal , Protéine-2 du complexe de la sclérose tubéreuse , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Protéine homologue de Ras enrichie dans le cerveau/métabolisme , Protéine homologue de Ras enrichie dans le cerveau/génétique , Humains , Animaux , Souris , Protéine-2 du complexe de la sclérose tubéreuse/métabolisme , Protéine-2 du complexe de la sclérose tubéreuse/génétique , Maladies alcooliques du foie/métabolisme , Maladies alcooliques du foie/génétique , Interleukine-1/métabolisme , Interleukine-1/génétique , Souris de lignée C57BL , Mâle , Cellules HEK293
11.
J Cutan Pathol ; 51(8): 618-623, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38689501

RÉSUMÉ

BACKGROUND: Psoriasis is an inflammatory skin disease driven by upregulation of cytokines in the Th17 pathway, including interleukin-36 (IL-36). Previous studies have highlighted the utility of IL-36 immunostaining for psoriasis compared to spongiotic dermatitis and other psoriasiform dermatoses; however, no study has examined the role of IL-36 staining in distinguishing psoriasis from pityriasis rosea (PR) and pityriasis lichenoides (PL), known histologic mimickers of psoriasis. METHODS: We compared the immunostaining pattern of IL-36 for 21 PR cases, 22 PL cases, and 10 psoriasis cases. We graded the immunostaining as 0, negative; 1, focal weak; 2, diffuse weak; 3, focal, strong; or 4, diffuse strong. We further categorized stains as negative (0-2 score) or positive (3-4 score) and utilized Fisher's exact test to compare the immunostaining pattern of these entities. RESULTS: All psoriasis specimens were positive for IL-36, whereas all PR specimens were negative (p = 0.00000002). Twenty PL specimens were negative (p = 0.000001). Nine of 10 pityriasis lichenoides et varioliformis acuta cases were negative (p = 0.00012), and 11 of 12 cases of pityriasis lichenoides chronica were negative (p = 0.00003). CONCLUSIONS: Our findings highlight the potential role of IL-36 immunostaining in distinguishing psoriasis from other psoriasiform dermatoses, including PR and PL.


Sujet(s)
Immunohistochimie , Interleukine-1 , Pityriasis lichénoïde , Pityriasis rosé , Psoriasis , Humains , Pityriasis lichénoïde/diagnostic , Pityriasis lichénoïde/anatomopathologie , Pityriasis lichénoïde/métabolisme , Psoriasis/diagnostic , Psoriasis/métabolisme , Psoriasis/anatomopathologie , Pityriasis rosé/diagnostic , Pityriasis rosé/anatomopathologie , Pityriasis rosé/métabolisme , Diagnostic différentiel , Interleukine-1/métabolisme , Immunohistochimie/méthodes , Mâle , Femelle , Adulte , Adulte d'âge moyen
12.
J Transl Med ; 22(1): 494, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38790051

RÉSUMÉ

BACKGROUND: Diabetic cardiomyopathy (DCM), a serious complication of diabetes, leads to structural and functional abnormalities of the heart and ultimately evolves to heart failure. IL-37 exerts a substantial influence on the regulation of inflammation and metabolism. Whether IL-37 is involved in DCM is unknown. METHODS: The plasma samples were collected from healthy controls, diabetic patients and DCM patients, and the level of IL-37 and its relationship with heart function were observed. The changes in cardiac function, myocardial fibrosis and mitochondrial injury in DCM mice with or without IL-37 intervention were investigated in vivo. By an in vitro co-culture approach involving HG challenge of cardiomyocytes and fibroblasts, the interaction carried out by cardiomyocytes on fibroblast profibrotic activation was studied. Finally, the possible interactive mediator between cardiomyocytes and fibroblasts was explored, and the intervention role of IL-37 and its relevant molecular mechanisms. RESULTS: We showed that the level of plasma IL-37 in DCM patients was upregulated compared to that in healthy controls and diabetic patients. Both recombinant IL-37 administration or inducing IL-37 expression alleviated cardiac dysfunction and myocardial fibrosis in DCM mice. Mechanically, hyperglycemia impaired mitochondria through SIRT1/AMPK/PGC1α signaling, resulting in significant cardiomyocyte apoptosis and the release of extracellular vesicles containing mtDNA. Fibroblasts then engulfed these mtDNA-enriched vesicles, thereby activating TLR9 signaling and the cGAS-STING pathway to initiate pro-fibrotic process and adverse remodeling. However, the presence of IL-37 ameliorated mitochondrial injury by preserving the activity of SIRT1-AMPK-PGC1α axis, resulting in a reduction in release of mtDNA-enriched vesicle and ultimately attenuating the progression of DCM. CONCLUSIONS: Collectively, our study demonstrates a protective role of IL-37 in DCM, offering a promising therapeutic agent for this disease.


Sujet(s)
ADN mitochondrial , Cardiomyopathies diabétiques , Fibrose , Interleukine-1 , Myocytes cardiaques , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Apoptose/effets des médicaments et des substances chimiques , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/traitement médicamenteux , ADN mitochondrial/métabolisme , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Interleukine-1/métabolisme , Souris de lignée C57BL , Myocarde/anatomopathologie , Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Sirtuine-1/métabolisme
13.
Biochem Biophys Res Commun ; 722: 150158, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-38795455

RÉSUMÉ

The cytokine interleukin-38 (IL-38), a recently discovered member of the IL-1 family, has been shown to regulate inflammation and improve hepatic endoplasmic reticulum stress and lipid metabolism in individuals with obesity. However, its impact on insulin signaling in skeletal muscle cells and the underlying mechanisms remain unclear. In vitro obesity models were established using palmitate treatment, and Western blot analysis was performed to assess target proteins. Commercial kits were used to measure glucose uptake in cultured myocytes. Our study showed that IL-38 treatment alleviated the impairment of insulin signaling, including IRS-1 and Akt phosphorylation, and increased glucose uptake in palmitate-treated C2C12 myocytes. Increased levels of STAT3-mediated signaling and oxidative stress were observed in these cells following palmitate treatment, and these effects were reversed by IL-38 treatment. In addition, IL-38 treatment upregulated the expression of PPARδ, SIRT1 and antioxidants. Knockdown of PPARδ or SIRT1 using appropriate siRNAs abrogated the effects of IL-38 on insulin signaling, oxidative stress, and the STAT3-dependent pathway. These results suggest that IL-38 alleviates insulin resistance by inhibiting STAT3-mediated signaling and oxidative stress in skeletal muscle cells through PPARδ/SIRT1. This study provides fundamental evidence to support the potential use of IL-38 as a safe therapeutic agent for the treatment of insulin resistance and type 2 diabetes.


Sujet(s)
Hyperlipidémies , Insulinorésistance , Stress oxydatif , Facteur de transcription STAT-3 , Transduction du signal , Sirtuine-1 , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Facteur de transcription STAT-3/métabolisme , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire , Hyperlipidémies/métabolisme , Hyperlipidémies/traitement médicamenteux , Récepteur PPAR delta/métabolisme , Récepteur PPAR delta/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/anatomopathologie , Interleukines/métabolisme , Interleukines/génétique , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Interleukine-1/métabolisme , Interleukine-1/génétique
14.
Mol Cell Endocrinol ; 591: 112274, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38777211

RÉSUMÉ

It has been reported that immune factors are associated with the occurrence of polycystic ovary syndrome (PCOS). Interleukin-1 (IL-1) is a member of the interleukin family that widely participates in the regulation of the inflammatory response in the immune system. In addition, it has been reported that aberrant IL-1 accumulation in serum is associated with the occurrence of PCOS. However, little is known about how IL-1 participates in the pathogenesis of PCOS. In the present study, we demonstrated that the immune microenvironment was altered in follicular fluid from PCOS patients and that the expression levels of two IL-1 cytokines, IL-1α and IL-1ß were increased. Transcriptome analysis revealed that IL-1α and IL-1ß treatment induced primary human granulosa-lutein (hGL) cell inflammatory response and increased the expression of serpin family E member 1 (SERPINE1). Mechanistically, we demonstrated that IL-1α and IL-1ß upregulated SERPINE1 expression through IL-1R1-mediated activation of downstream P50 and P52 signaling pathways in human granulosa cells. Our study highlighted the role of immune state changes in the occurrence of PCOS and provided new insight into the treatment of patients with IL-1-induced ovarian function disorders.


Sujet(s)
Cellules de la granulosa , Interleukine-1 , Cellules lutéales , Inhibiteur-1 d'activateur du plasminogène , Syndrome des ovaires polykystiques , Transduction du signal , Humains , Femelle , Inhibiteur-1 d'activateur du plasminogène/génétique , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Cellules lutéales/métabolisme , Cellules lutéales/effets des médicaments et des substances chimiques , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/génétique , Interleukine-1/métabolisme , Interleukine-1/génétique , Cellules de la granulosa/métabolisme , Cellules de la granulosa/effets des médicaments et des substances chimiques , Interleukine-1 bêta/métabolisme , Adulte , Liquide folliculaire/métabolisme , Interleukine-1 alpha/métabolisme , Interleukine-1 alpha/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Récepteur à l'interleukine-1 de type I/génétique , Récepteur à l'interleukine-1 de type I/métabolisme , Cellules cultivées
15.
Cytokine ; 179: 156598, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38583255

RÉSUMÉ

BACKGROUND: Allograft rejection remains a major obstacle to long-term graft survival. Although previous studies have demonstrated that IL-37 exhibited significant immunomodulatory effects in various diseases, research on its role in solid organ transplantation has not been fully elucidated. In this study, the therapeutic effect of recombinant human IL-37 (rhIL-37) was evaluated in a mouse cardiac allotransplantation model. METHODS: The C57BL/6 recipients mouse receiving BALB/c donor hearts were treated with rhIL-37. Graft pathological and immunohistology changes, immune cell populations, and cytokine profiles were analyzed on postoperative day (POD) 7. The proliferative capacities of Th1, Th17, and Treg subpopulations were assessed in vitro. Furthermore, the role of the p-mTOR pathway in rhIL-37-induced CD4+ cell inhibition was also elucidated. RESULTS: Compared to untreated groups, treatment of rhIL-37 achieved long-term cardiac allograft survival and effectively alleviated allograft rejection indicated by markedly reduced infiltration of CD4+ and CD11c+ cells and ameliorated graft pathological changes. rhIL-37 displayed significantly less splenic populations of Th1 and Th17 cells, as well as matured dendritic cells. The percentages of Tregs in splenocytes were significantly increased in the therapy group. Furthermore, rhIL-37 markedly decreased the levels of TNF-α and IFN-γ, but increased the level of IL-10 in the recipients. In addition, rhIL-37 inhibited the expression of p-mTOR in CD4+ cells of splenocytes. In vitro, similar to the in vivo experiments, rhIL-37 caused a decrease in the proportion of Th1 and Th17, as well as an increase in the proportion of Treg and a reduction in p-mTOR expression in CD4+ cells. CONCLUSIONS: We demonstrated that rhIL-37 effectively suppress acute rejection and induce long-term allograft acceptance. The results highlight that IL-37 could be novel and promising candidate for prevention of allograft rejection.


Sujet(s)
Allogreffes , Rejet du greffon , Transplantation cardiaque , Interleukine-1 , Souris de lignée BALB C , Souris de lignée C57BL , Protéines recombinantes , Animaux , Rejet du greffon/immunologie , Rejet du greffon/prévention et contrôle , Humains , Souris , Protéines recombinantes/pharmacologie , Interleukine-1/métabolisme , Survie du greffon/effets des médicaments et des substances chimiques , Survie du greffon/immunologie , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Mâle , Sérine-thréonine kinases TOR/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
17.
Ocul Surf ; 32: 182-191, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38490477

RÉSUMÉ

PURPOSE: To explore novel role and molecular mechanism of a natural osmoprotectant ectoine in protecting corneal epithelial cell survival and barrier from hyperosmotic stress. METHODS: Primary human corneal epithelial cells (HCECs) were established from donor limbus. The confluent cultures in isosmolar medium were switched to hyperosmotic media (400-500 mOsM), with or without ectoine or rhIL-37 for different time periods. Cell viability and proliferation were evaluated by MTT or WST assay. The integrity of barrier proteins and the expression of cytokines and cathepsin S were evaluated by RT-qPCR, ELISA, and immunostaining with confocal microscopy. RESULTS: HCECs survived well in 450mOsM but partially damaged in 500mOsM medium. Ectoine well protected HCEC survival and proliferation at 500mOsM. The integrity of epithelial barrier was significantly disrupted in HCECs exposed to 450mOsM, as shown by 2D and 3D confocal immunofluorescent images of tight junction proteins ZO-1 and occludin. Ectoine at 5-20 mM well protected these barrier proteins under hyperosmotic stress. The expression of TNF-α, IL-1ß, IL-6 and IL-8 were dramatically stimulated by hyperosmolarity but significantly suppressed by Ectoine at 5-40 mM. Cathepsin S, which was stimulated by hyperosmolarity, directly disrupted epithelial barrier. Interestingly, anti-inflammatory cytokine IL-37 was suppressed by hyperosmolarity, but restored by ectoine at mRNA and protein levels. Furthermore, rhIL-37 suppressed cathepsin S and rescued cell survival and barrier in HCECs exposed to hyperosmolarity. CONCLUSION: Our findings demonstrate that ectoine protects HCEC survival and barrier from hyperosmotic stress by promoting IL-37. This provides new insight into pathogenesis and therapeutic potential for dry eye disease.


Sujet(s)
Acides aminés diaminés , Survie cellulaire , Épithélium antérieur de la cornée , Pression osmotique , Humains , Survie cellulaire/effets des médicaments et des substances chimiques , Épithélium antérieur de la cornée/métabolisme , Épithélium antérieur de la cornée/effets des médicaments et des substances chimiques , Épithélium antérieur de la cornée/anatomopathologie , Cellules cultivées , Acides aminés diaminés/pharmacologie , Interleukine-1/métabolisme , Interleukine-1/pharmacologie , Test ELISA , Microscopie confocale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytokines/métabolisme
18.
Br J Pharmacol ; 181(15): 2429-2442, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38532634

RÉSUMÉ

BACKGROUND AND PURPOSE: The interleukin (IL)-36 pathway is a critical player in the pathogenesis of pustular psoriasis. However, therapies targeting this pathway are limited or unaffordable (e.g. the anti-IL-36 receptor antibody). AMP-activated protein kinase (AMPK), a regulator of cellular energy and metabolism, is known to participate in inflammatory diseases. However, its role in IL-36-induced skin inflammation remains unclear. Therefore, we sought to investigate the role of AMPK signals in regulating IL-36-induced responses in the skin. EXPERIMENTAL APPROACH: IL-36-stimulated primary normal human epidermal keratinocytes (NHEKs) and IL-36-injected (intradermally) BALB/c mice served as the cell and animal models, respectively. Additionally, 5-aminoimidazole-4-carboxamide riboside (AICAR) and A769662 served as AMPK activators. KEY RESULTS: AICAR and A769662 significantly suppressed the IL-36-induced IL-8 (CXCL8) and CCL20 production from NHEKs. IL-36-induced IκBζ protein expression was prominently reduced and IKK/IκBα phosphorylation was attenuated by AICAR and A769662. Conversely, AMPKα knockdown increased IκBζ protein expression and IKK/IκBα phosphorylation in IL-36-treated NHEKs. Furthermore, AICAR and A769662 enhanced IL-36-induced-IκBζ protein degradation via the proteasome-dependent but not the lysosome-dependent pathway. Pretreatment of NHEKs with IL-36 slightly suppressed the AICAR- and A769662-triggered phosphorylation of AMPK and acetyl-CoA carboxylase. In the mouse model, topical application of AICAR significantly reduced ear swelling, redness, epidermal thickening, neutrophil infiltration and inflammatory and antimicrobial peptide gene expression. CONCLUSION AND IMPLICATIONS: AMPK activation suppresses IL-36-induced IL-8 and CCL20 release by regulating IκBζ expression in keratinocytes and reduces IL-36-induced skin inflammation in mice, suggesting that AMPK activation is a potential strategy for treating patients with IL-36-mediated inflammatory skin disorders.


Sujet(s)
AMP-Activated Protein Kinases , 5-Amino-imidazole-4-carboxamide , Souris de lignée BALB C , Peau , Animaux , AMP-Activated Protein Kinases/métabolisme , Humains , 5-Amino-imidazole-4-carboxamide/analogues et dérivés , 5-Amino-imidazole-4-carboxamide/pharmacologie , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/métabolisme , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Ribonucléotides/pharmacologie , Interleukine-1/métabolisme , Souris , Interleukine-8/métabolisme , Chimiokine CCL20/métabolisme , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Cellules cultivées , Activation enzymatique/effets des médicaments et des substances chimiques , Protéines adaptatrices de la transduction du signal
19.
Int J Mol Sci ; 25(6)2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38542222

RÉSUMÉ

Mast cells (MCs) are derived from hematopoietic progenitors, mature in vascularized tissues, and participate in innate and acquired immunity. Neuroinflammation is a highly debated topic in the biomedical literature; however, the impact of tumor necrosis factor (TNF) and IL-33 on MCs in the brain has not been widely addressed. MCs can be activated by IgE binding to FcεRI, as well as by different antigens. After activation, MCs mediate various immunological and inflammatory responses through TNF and IL-33. TNF has two receptors: TNFR1, a p55 molecule, and TNFR2, a p75 molecule. This cytokine is the only one of its kind to be stored in the granules of MCs and can also be generated by de novo synthesis via mRNA. In the central nervous system (CNS), TNF is produced almost exclusively by microglial cells, neurons, astrocytes, and, minimally, by endothelial cells. After its release into brain tissue, TNF rapidly induces the adhesion molecules endothelial leukocyte adhesion molecule 1 (ELAM-1), intercellular adhesion molecule 1 (ICAM-1), and vascular cell adhesion molecule 1 (VCAM-1) in endothelial cells. TNF causes the chemoattraction of neutrophils by inducing several molecules, including CXC chemokines (IL-8). Both MCs and microglial cells act as a primary barrier against foreign molecules in the CNS, producing pro-inflammatory cytokines such as IL-33. IL-33 belongs to the IL-1 family, is activated through the ST2L/IL1-RAcP receptor complex, and mediates both the innate and adaptive immune response. IL-33 is a nuclear transcription factor expressed in the brain, where it induces pro-inflammatory cytokines (TNF and IL-1) and chemokines (CCL2, CCL3, CCL5, and CXCL10). Therefore, MCs and microglia in the CNS are a source of pro-inflammatory cytokines, including TNF and IL-33, that mediate many brain diseases. The inhibition of TNF and IL-33 may represent a new therapeutic approach that could complement existing neuroinflammatory therapies.


Sujet(s)
Cytokines , Maladies neuro-inflammatoires , Humains , Cytokines/métabolisme , Mastocytes/métabolisme , Interleukine-33/métabolisme , Cellules endothéliales/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Molécule-1 d'adhérence intercellulaire/métabolisme , Interleukine-1/métabolisme
20.
Adv Healthc Mater ; 13(13): e2303957, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38339835

RÉSUMÉ

Interleukin-37 (IL-37) is a potent anti-inflammatory cytokine belonging to the IL-1 family. This study investigates the regulatory mechanism and reparative effects of IL-37 on HF-related human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) and engineered human heart tissue subjected to hypoxia and H2O2 treatment. The contractile force and Ca2+ conduction capacity of the tissue are assessed using a stretching platform and high-resolution fluorescence imaging system. This investigation reveals that IL-37 treatment significantly enhances cell viability, calcium transient levels, contractile force, and Ca2+ conduction capacity in HF-related hiPSC-CMs and engineered human heart tissue. Notably, IL-37 facilitates the upregulation of sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) through enhancing nuclear p-STAT3 levels. This effect is mediated by the binding of p-STAT3 to the SERCA2a promoter, providing a novel insight on the reparative potential of IL-37 in HF. IL-37 demonstrates its ability to enhance systolic function by modulating myocardial calcium handling via the p-STAT3/SERCA2a axis in HF-related engineered human heart tissue (as shown in schematic diagram).


Sujet(s)
Calcium , Interleukine-1 , Myocytes cardiaques , Facteur de transcription STAT-3 , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Ingénierie tissulaire , Humains , Facteur de transcription STAT-3/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Interleukine-1/métabolisme , Interleukine-1/pharmacologie , Ingénierie tissulaire/méthodes , Myocytes cardiaques/métabolisme , Calcium/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Défaillance cardiaque/métabolisme , Myocarde/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...