Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.847
Filtrer
1.
J Biochem Mol Toxicol ; 38(9): e23841, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39235091

RÉSUMÉ

Considering detrimental impacts of combustible cigarettes (CCs) on the exacerbation of diabetes mellitus (DM), a significant number of DM patients have substituted CCs with electronic nicotine delivery systems (ENDS). Herewith, we compared CCs and ENDS-dependent modulation of immune cell-driven inflammation in DM patients who used ENDS (DMENDS), CCs (DMCC) or were non-smokers (DMAIR), paving the way for the better understanding of ENDS-induced biological effects. Multiple low dose streptozotocin (MLD-STZ)-induced mice model of DM was used to support clinical findings. Both CCs and ENDS aggravated MLD-STZ-induced DM. Pancreatic injury and inflammation were more severe in CC-exposed than in ENDS-exposed diabetic mice. CCs promoted activation of NLRP3 inflammasome, enhanced production of inflammatory cytokines in neutrophils, macrophages and remarkably improved antigen presenting capacity of dendritic cells which resulted in the expansion of TNF-α, IFN-γ and IL-17-producing Th1 and Th17 lymphocytes, NK and NKT cells. Compared to CCs, ENDS more intensively promoted expansion of FoxP3-expressing, IL-10-producing NK and NKT cells and triggered less intense systemic inflammatory response in diabetic animals. Similar findings were observed in DM patients. The highest numbers of inflammatory, TNF-α and IL-1ß-producing neutrophils and monocytes, TNF-α and IFN-γ-producing T lymphocytes, NK and NKT cells were determined in the blood of DMCC patients, while total number of immunosuppressive, TGF-ß-producing CD3 + CD4 + T cells was the highest in the blood of DMENDS patients. In conclusion, although both CC and ENDS aggravate on-going inflammation in DM, ENDS have weaker capacity to induce production of inflammatory cytokines in immune cells than CCs.


Sujet(s)
Diabète expérimental , Dispositifs électroniques d'administration de nicotine , Inflammation , Animaux , Diabète expérimental/immunologie , Souris , Humains , Inflammation/immunologie , Mâle , Femelle , Adulte d'âge moyen , Souris de lignée C57BL , Cytokines/métabolisme , Cytokines/sang , Streptozocine , Adulte
2.
Front Immunol ; 15: 1429205, 2024.
Article de Anglais | MEDLINE | ID: mdl-39100662

RÉSUMÉ

Islet transplantation is a promising therapy for diabetes treatment. However, the molecular underpinnings governing the immune response, particularly T-cell dynamics in syngeneic and allogeneic transplant settings, remain poorly understood. Understanding these T cell dynamics is crucial for enhancing graft acceptance and managing diabetes treatment more effectively. This study aimed to elucidate the molecular mechanisms, gene expression differences, biological pathway alterations, and intercellular communication patterns among T-cell subpopulations after syngeneic and allogeneic islet transplantation. Using single-cell RNA sequencing, we analyzed cellular heterogeneity and gene expression profiles using the Seurat package for quality control and dimensionality reduction through t-SNE. Differentially expressed genes (DEGs) were analyzed among different T cell subtypes. GSEA was conducted utilizing the HALLMARK gene sets from MSigDB, while CellChat was used to infer and visualize cell-cell communication networks. Our findings revealed genetic variations within T-cell subpopulations between syngeneic and allogeneic islet transplants. We identified significant DEGs across these conditions, highlighting molecular discrepancies that may underpin rejection or other immune responses. GSEA indicated activation of the interferon-alpha response in memory T cells and suppression in CD4+ helper and γδ T cells, whereas TNFα signaling via NFκB was particularly active in regulatory T cells, γδ T cells, proliferating T cells, and activated CD8+ T cells. CellChat analysis revealed complex communication patterns within T-cell subsets, notably between proliferating T cells and activated CD8+ T cells. In conclusion, our study provides a comprehensive molecular landscape of T-cell diversity in islet transplantation. The insights into specific gene upregulation in xenotransplants suggest potential targets for improving graft tolerance. The differential pathway activation across T-cell subsets underscores their distinct roles in immune responses posttransplantation.


Sujet(s)
Transplantation d'ilots de Langerhans , Analyse sur cellule unique , Transplantation homologue , Animaux , Souris , Analyse sur cellule unique/méthodes , Souris de lignée C57BL , Analyse de séquence d'ARN , Transcriptome , Transplantation isogénique , Analyse de profil d'expression de gènes , Diabète expérimental/immunologie , Diabète expérimental/génétique , Rejet du greffon/immunologie , Rejet du greffon/génétique , Mâle , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Souris de lignée BALB C , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Survie du greffon/immunologie , Survie du greffon/génétique
3.
JCI Insight ; 9(16)2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39171525

RÉSUMÉ

Type 2 diabetes (T2D) is on the rise worldwide and is associated with various complications in the oral cavity. Using an adult-onset diabetes preclinical model, we demonstrated profound periodontal alterations in T2D mice, including inflamed gingiva, disintegrated periodontal ligaments (PDLs), marked alveolar bone loss, and unbalanced bone remodeling due to decreased formation and increased resorption. Notably, we observed elevated levels of the Wnt signaling inhibitor sclerostin in the alveolar bone of T2D mice. Motivated by these findings, we investigated whether a sclerostin-neutralizing antibody (Scl-Ab) could rescue the compromised periodontium in T2D mice. Administering Scl-Ab subcutaneously once a week for 4 weeks, starting 4 weeks after T2D induction, led to substantial increases in bone mass. This effect was attributed to the inhibition of osteoclasts and promotion of osteoblasts in both control and T2D mice, effectively reversing the bone loss caused by T2D. Furthermore, Scl-Ab stimulated PDL cell proliferation, partially restored the PDL fibers, and mitigated inflammation in the periodontium. Our study thus established a T2D-induced periodontitis mouse model characterized by inflammation and tissue degeneration. Scl-Ab emerged as a promising intervention to counteract the detrimental effects of T2D on the periodontium, exhibiting limited side effects on other craniofacial hard tissues.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Résorption alvéolaire , Diabète de type 2 , Animaux , Souris , Diabète de type 2/immunologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/complications , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/métabolisme , Résorption alvéolaire/prévention et contrôle , Résorption alvéolaire/étiologie , Résorption alvéolaire/anatomopathologie , Mâle , Maladies parodontales/immunologie , Anticorps neutralisants/pharmacologie , Desmodonte/anatomopathologie , Desmodonte/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Diabète expérimental/immunologie , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Souris de lignée C57BL , Parodontite/immunologie , Parodontite/anatomopathologie , Parodontite/traitement médicamenteux , Remodelage osseux/effets des médicaments et des substances chimiques
4.
Biochem Pharmacol ; 226: 116413, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38971333

RÉSUMÉ

Chronic nonhealing diabetic wounds are a critical clinical challenge. Regulatory T cells (Tregs) are immunosuppressive modulators affecting wound healing progression by controlling the inflammatory response. The current study attempted to investigate whether the exosomes derived from cord blood (CB) Tregs can accelerate the healing process. Exosomes were isolated from CB-Treg cultures using ultracentrifugation and validated with different specific markers of exosomes. The purified CB-Treg-derived exosomes were co-cultured with peripheral blood mononuclear cells (PBMCs) and CD14+ monocytes. The migration-promoting effect of CB-Treg-derived exosomes on fibroblasts and endothelial cells was investigated. We used thermosensitive Pluronic F-127 hydrogel (PF-127) loaded with CB-Treg-derived exosomes in a diabetic wound healing mouse model. CB-Treg-derived exosomes with 30-120 nm diameters revealed exosome-specific markers, such as TSG101, Alix, and CD63. CB-Treg-derived exosomes were mainly bound to the monocytes when co-cultured with PBMCs, and promoted monocyte polarization to the anti-inflammatory phenotype (M2) in vitro. CB-Treg-derived exosomes enhanced the migration of endothelial cells and fibroblasts. Furthermore, CB-Treg-derived exosomes treatment accelerated wound healing by downregulating inflammatory factor levels and upregulating the M2 macrophage ratio in vivo. Our findings indicated that CB-Treg-derived exosomes could be a promising cell-free therapeutic strategy for diabetic wound healing, partly by targeting monocytes.


Sujet(s)
Diabète expérimental , Exosomes , Sang foetal , Monocytes , Lymphocytes T régulateurs , Cicatrisation de plaie , Exosomes/métabolisme , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Animaux , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Cicatrisation de plaie/physiologie , Monocytes/métabolisme , Monocytes/effets des médicaments et des substances chimiques , Monocytes/immunologie , Souris , Sang foetal/cytologie , Humains , Diabète expérimental/métabolisme , Diabète expérimental/immunologie , Mâle , Souris de lignée C57BL , Techniques de coculture , Cellules cultivées , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques
5.
J Diabetes Investig ; 15(9): 1191-1201, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38943657

RÉSUMÉ

AIMS/INTRODUCTION: Regulatory T cells (Tregs) have protected against many cardiovascular diseases. This study was intended to explore the effect of Tregs on diabetic cardiomyopathy (DCM) using a db/db mouse model. MATERIALS AND METHODS: Eight-week-old male db/db mice were randomly divided into four groups: the control group, administered 200 µL phosphate-buffered saline; the small-dose Treg group, administered 105 Tregs; the large-dose Treg group, administered 106 Tregs; and the PC group, administered 100 µg anti-CD25 specific antibody (PC61) and 106 Tregs. After 12 weeks, mice were euthanized. Transthoracic echocardiography was carried out at the beginning and end of the experiment. Relevant basic experiments to evaluate the effects of Tregs on DCM were carried out. RESULTS: Echocardiography showed that the impaired diastolic and systolic functions were significantly improved in mice administered large-dose Tregs. Large-dose Tregs significantly ameliorated myocardial hypertrophy and fibrosis, and decreased hypertrophic gene expression and collagen deposition. The protective effects of Tregs on diabetic hearts were associated with decreased oxidative stress, inflammatory response and apoptosis. In addition, Tregs promoted the activation of the phosphatidylinositol 3-kinase-protein kinase B signaling pathway, whereas they inhibited extracellular signal-regulated kinase 1/2 and Jun N-terminal kinase phosphorylation, which might be responsible for the cardioprotective role of Tregs against DCM. CONCLUSIONS: Tregs ameliorated myocardial hypertrophy and fibrosis, improved cardiac dysfunction, and protected against DCM progression in db/db mice. The mechanisms involved a decrease of inflammatory response, oxidative stress and apoptosis, which might be mediated by phosphatidylinositol 3-kinase-protein kinase B and mitogen-activated protein kinase pathways. Hence, Tregs might serve as a promising therapeutic approach for DCM treatment.


Sujet(s)
Cardiomyopathies diabétiques , Lymphocytes T régulateurs , Animaux , Lymphocytes T régulateurs/immunologie , Cardiomyopathies diabétiques/prévention et contrôle , Cardiomyopathies diabétiques/immunologie , Mâle , Souris , Stress oxydatif , Apoptose , Diabète expérimental/complications , Diabète expérimental/immunologie , Modèles animaux de maladie humaine , Transduction du signal
6.
Front Immunol ; 15: 1347018, 2024.
Article de Anglais | MEDLINE | ID: mdl-38887289

RÉSUMÉ

Purpose: Inflammation is involved in the pathogenesis of diabetes, however the impact of diabetes on organ-specific autoimmune diseases remains unexplored. Experimental autoimmune uveoretinitis (EAU) is a widely accepted animal model of human endogenous uveitis. In this study, we investigated the effects of diabetic conditions on the development of EAU using a mouse diabetes model. Methods: EAU was induced in wild-type C57BL/6 (WT) mice and Ins2Akita (Akita) mice with spontaneous diabetes by immunization with IRBP peptide. Clinical and histopathological examinations, and analysis of T cell activation state were conducted. In addition, alternations in the composition of immune cell types and gene expression profiles of relevant immune functions were identified using single-cell RNA sequencing. Results: The development of EAU was significantly attenuated in immunized Akita (Akita-EAU) mice compared with immunized WT (WT-EAU) mice, although T cells were fully activated in Akita-EAU mice, and the differentiation into Th17 cells and regulatory T (Treg) cells was promoted. However, Th1 cell differentiation was inhibited in Akita-EAU mice, and single-cell analysis indicated that gene expression associated AP-1 signaling pathway (JUN, FOS, and FOSB) was downregulated not only in Th1 cells but also in Th17, and Treg cells in Akita-EAU mice at the onset of EAU. Conclusions: In diabetic mice, EAU was significantly attenuated. This was related to selective inhibition of Th1 cell differentiation and downregulated AP-1 signaling pathway in both Th1 and Th17 cells.


Sujet(s)
Maladies auto-immunes , Différenciation cellulaire , Souris de lignée C57BL , Transduction du signal , Lymphocytes auxiliaires Th1 , Cellules Th17 , Facteur de transcription AP-1 , Uvéite , Animaux , Uvéite/immunologie , Cellules Th17/immunologie , Lymphocytes auxiliaires Th1/immunologie , Souris , Facteur de transcription AP-1/métabolisme , Différenciation cellulaire/immunologie , Maladies auto-immunes/immunologie , Diabète expérimental/immunologie , Modèles animaux de maladie humaine , Femelle
7.
Front Immunol ; 15: 1415004, 2024.
Article de Anglais | MEDLINE | ID: mdl-38895119

RÉSUMÉ

Introduction: This study examined the impact of 5'-(N- ethylcarboxamido)adenosine (NECA) in the peripheral blood of healthy individuals, those with diabetes mellitus, diabetic retinopathy (DR), and C57BL/6 mice, both in vivo and in vitro. Methods: Enzyme-linked immunosorbent assay (ELISA) and flow cytometry (FCM) were used to evaluate the effects of NECA on dendritic cells (DCs) and mouse bone marrow-derived dendritic cells (BMDCs) and the effects of NECA-treated DCs on Treg and Th17 cells. The effect of NECA on the Toll-like receptor (TLR) pathway in DCs was evaluated using polymerase chain reaction (PCR) and western blotting (WB). Results: FCM and ELISA showed that NECA inhibited the expression of surface markers of DCs and BMDCs, increased anti-inflammatory cytokines and decreased proinflammatory cytokines. PCR and WB showed that NCEA decreased mRNA transcription and protein expression in the TLR-4-MyD88-NF-kß pathway in DCs and BMDCs. The DR severity in streptozocin (STZ) induced diabetic mice was alleviated. NECA-treated DCs and BMDCs were co-cultivated with CD4+T cells, resulting in modulation of Treg and Th17 differentiation, along with cytokine secretion alterations. Conclusion: NECA could impair DCs' ability to present antigens and mitigate the inflammatory response, thereby alleviating the severity of DR.


Sujet(s)
Cellules dendritiques , Rétinopathie diabétique , Souris de lignée C57BL , Transduction du signal , Récepteurs de type Toll , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Rétinopathie diabétique/immunologie , Rétinopathie diabétique/métabolisme , Souris , Humains , Mâle , Récepteurs de type Toll/métabolisme , Diabète expérimental/immunologie , Femelle , Cellules Th17/immunologie , Cellules Th17/métabolisme , Cytokines/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Adulte d'âge moyen , Inflammation/immunologie
8.
JCI Insight ; 9(15)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38889387

RÉSUMÉ

Diabetes mellitus (DM) is an independent risk factor for atrial fibrillation (AF). The mechanisms underlying DM-associated AF are unclear. AF and DM are both related to inflammation. We investigated whether DM-associated inflammation contributed to AF risk. Mice were fed with high-fat diet to induce type II DM and were subjected to IL-1ß antibodies, macrophage depletion by clodronate liposomes, a mitochondrial antioxidant (mitoTEMPO), or a cardiac ryanodine receptor 2 (RyR2) stabilizer (S107). All tests were performed at 36-38 weeks of age. DM mice presented with increased AF inducibility, enhanced mitochondrial reactive oxygen species (mitoROS) generation, and activated innate immunity in the atria, as evidenced by enhanced monocyte chemoattractant protein-1 (MCP-1) expression, macrophage infiltration, and IL-1ß levels. Signs of aberrant RyR2 Ca2+ leak were observed in the atria of DM mice. IL-1ß neutralization, macrophage depletion, and exposure to mitoTEMPO and S107 significantly ameliorated the AF vulnerability in DM mice. Atrial overexpression of MCP-1 increased AF occurrence in normal mice through the same mechanistic signaling cascade as observed in DM mice. In conclusion, macrophage-mediated IL-1ß contributed to DM-associated AF risk through mitoROS modulation of RyR2 Ca2+ leak.


Sujet(s)
Fibrillation auriculaire , Diabète expérimental , Interleukine-1 bêta , Macrophages , Animaux , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/étiologie , Fibrillation auriculaire/immunologie , Souris , Interleukine-1 bêta/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Diabète expérimental/complications , Diabète expérimental/métabolisme , Diabète expérimental/immunologie , Mâle , Diabète de type 2/métabolisme , Diabète de type 2/complications , Diabète de type 2/immunologie , Chimiokine CCL2/métabolisme , Atrium du coeur/métabolisme , Atrium du coeur/anatomopathologie , Canal de libération du calcium du récepteur à la ryanodine/métabolisme , Espèces réactives de l'oxygène/métabolisme , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables , Inflammation/métabolisme
9.
Immunol Lett ; 267: 106862, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38702033

RÉSUMÉ

BACKGROUND: Diabetic retinopathy (DR) stands as a prominent complication of diabetes. Berberine (BBR) has reported to be effective to ameliorate the retinal damage of DR. Studying the potential immunological mechanisms of BBR on the streptozotocin (STZ) induced DR mouse model will explain the therapeutic mechanisms of BBR and provide theoretical basis for the clinical application of this drug. METHODS: C57BL/6 J mice were induced into a diabetic state using a 50 mg/(kg·d) dose of STZ over a 5-day period. Subsequently, they were subjected to a high-fat diet (HFD) for one month. Following a 5-week treatment with 100 mg/(kg·d) BBR, the concentrations of inflammatory factors in the mice's peripheral blood were determined using an enzyme-linked immunosorbent assay (ELISA). Hematoxylin-eosin staining was employed to scrutinize pathological changes in the mice's retinas, while flow cytometry assessed the proportions of T-lymphocyte subsets and the activation status of dendritic cells (DCs) in the spleen and lymph nodes. CD4+T cells and DC2.4 cell lines were utilized to investigate the direct and indirect effects of BBR on T cells under high glucose conditions in vitro. RESULTS: Following 5 weeks of BBR treatment in the streptozotocin (STZ) mouse model of DR, we observed alleviation of retinal lesions and a down-regulation in the secretion of inflammatory cytokines, namely TNF-α, IL-1ß, and IL-6, in the serum of these mice. And in the spleen and lymph nodes of these mice, BBR inhibited the proportion of Th17 cells and promoted the proportion of Treg cells, thereby down-regulating the Th17/Treg ratio. Additionally, in vitro experiments, BBR directly inhibited the expression of the transcription factor RORγt and promoted the expression of the transcription factor Foxp3 in T cells, resulting in a down-regulation of the Th17/Treg ratio. Furthermore, BBR indirectly modulated the Th17/Treg ratio by suppressing the secretion of TNF-α, IL-1ß, and IL-6 by DCs and enhancing the secretion of indoleamine 2,3-dioxygenase (IDO) and transforming growth factor-beta (TGF-ß) by DCs. This dual action inhibited Th17 cell differentiation while promoting Treg cells. CONCLUSION: Our findings indicate that BBR regulate T cell subpopulation differentiation, reducing the Th17/Treg ratio by directly or indirectly pathway. This represents a potential therapeutic avenue of BBR for improving diabetic retinopathy.


Sujet(s)
Berbérine , Diabète expérimental , Rétinopathie diabétique , Lymphocytes T régulateurs , Cellules Th17 , Animaux , Berbérine/pharmacologie , Berbérine/usage thérapeutique , Rétinopathie diabétique/traitement médicamenteux , Rétinopathie diabétique/immunologie , Rétinopathie diabétique/étiologie , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Souris , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/immunologie , Mâle , Cytokines/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/métabolisme , Rétine/anatomopathologie , Rétine/immunologie , Rétine/effets des médicaments et des substances chimiques , Rétine/métabolisme
10.
Sci Rep ; 14(1): 10340, 2024 05 06.
Article de Anglais | MEDLINE | ID: mdl-38710764

RÉSUMÉ

This study aims to evaluate the role of trefoil factor 3 (TFF3) peptides in type 2 diabetes mellitus (T2DM) from an inflammatory perspective. The focus was on exploring how TFF3 affects the function of T cells. TFF3 overexpression model was constructed using lentivirus in Jurkat cell lines. We evaluated the impact of TFF3 on the proliferation, apoptosis, and IL-17A levels of Jurkat cells cultured in high glucose. The T2DM model was induced in TFF3 knockout (KO) mice through streptozotocin combined with high-fat diet. The measurements included glucose tolerance, insulin tolerance, inflammation markers, Th17 cell proportion, and pancreatic pathological changes. The T2DM modeling led to splenomegaly in mice, and increased expression of TFF3 in their spleens. Overexpression of TFF3 increased the proportion of IL-17+ T cells and the levels of Th17-related cytokines in Jurkat cells. There was no difference in body weight and blood glucose levels between wild-type and TFF3 KO mice. However, T2DM mice lacking the TFF3 gene showed improved glucose utilization, ameliorated pancreatic pathology, decreased inflammation levels, and reduced Th17 cell ratio. TFF3 may be involved in the chronic inflammatory immune response in T2DM. Its mechanism may be related to the regulation of the RORγt/IL-17 signaling pathway and its impact on T cell proliferation and apoptosis.


Sujet(s)
Diabète de type 2 , Souris knockout , Cellules Th17 , Facteur en trèfle-3 , Cellules Th17/immunologie , Cellules Th17/métabolisme , Animaux , Humains , Diabète de type 2/métabolisme , Diabète de type 2/immunologie , Souris , Facteur en trèfle-3/métabolisme , Facteur en trèfle-3/génétique , Cellules Jurkat , Interleukine-17/métabolisme , Diabète expérimental/immunologie , Diabète expérimental/métabolisme , Mâle , Prolifération cellulaire , Apoptose , Alimentation riche en graisse/effets indésirables
11.
Int Immunopharmacol ; 134: 112254, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38749333

RÉSUMÉ

BACKGROUND: Patients with diabetes are particularly susceptible to Legionella pneumophila (LP) infection, but the exact pathogenesis of LP infection in diabetic patients is still not fully understood. Herein, we investigated the effect of diabetes on immune function during LP infection in vitro and in vivo. METHODS: The time course of LP infection in macrophages under normal and high-glucose (HG) conditions was examined in vitro. Western blot was used to determine nucleotide-binding oligomerization domain 1 (NOD1), kinase 1/2 (ERK1/2), mitogen-activated protein kinase p38 (MAPK p38), and c-Jun N-terminal kinases (JNK). Enzyme-linked immunosorbent assay (ELISA) was used to assess the secretion of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Cell Counting Kit-8 (CCK8) assay assessed U937 cell viability after treating cells with different concentrations of high sugar medium and ML130 (NOD1 inhibitor). For the in vivo study, normal and streptozocin-induced diabetic guinea pigs were infected with LP for 6, 24, and 72 h, after which NOD1, MAPK-related signals, TNF-α, and IL-6 expression in lung tissues were assessed using immunohistochemistry, western blot, and RT-PCR. RESULTS: HG attenuated the upregulation of NOD1 expression and reduced TNF-α and IL-6 secretion caused by LP compared with LP-infected cells exposed to normal glucose levels (all p < 0.05). In diabetic guinea pigs, HG inhibited the upregulation of NOD1 expression in lung tissues and the activation of p38, ERK1/2, and cJNK caused by LP infection compared to control pigs (all p < 0.05). CONCLUSION: HG attenuates the response of macrophages to LP infection by inhibiting NOD1 upregulation and the activation of MAPK signaling.


Sujet(s)
Glucose , Legionella pneumophila , Macrophages , Protéine adaptatrice de signalisation NOD1 , Protéine adaptatrice de signalisation NOD1/métabolisme , Protéine adaptatrice de signalisation NOD1/génétique , Animaux , Humains , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Legionella pneumophila/immunologie , Glucose/métabolisme , Cochons d'Inde , Mâle , Interleukine-6/métabolisme , Maladie des légionnaires/immunologie , Diabète expérimental/immunologie , Diabète expérimental/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Cellules U937 , Facteur de nécrose tumorale alpha/métabolisme , Souris
12.
Int Immunopharmacol ; 133: 112056, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38626546

RÉSUMÉ

OBJECTIVES: The aim of this study was to investigate the effect of 4µ8c, an inhibitor targeting the endoplasmic reticulum stress-associated factor IRE1α, on macrophage polarization in an experimental model of diabetic periodontitis through ex vivo experiments. MATERIALS AND METHODS: Local alveolar bone parameters were evaluated using Micro-CT following intraperitoneal administration of 4µ8c in mice with experimental diabetic periodontitis. Surface markers indicating macrophage polarization were identified using immunofluorescence. In vitro experiments were performed employing bone marrow-derived macrophages and gingival fibroblasts. Macrophage polarization was determined using flow cytometry. Principal impacted signaling pathways were identified through Western blot analysis. RESULTS: Results from both in vitro and in vivo experiments demonstrated that 4µ8c mitigated alveolar bone resorption and inflammation in mice with diabetic periodontitis. Furthermore, it modulated macrophage polarization towards the M2 phenotype and augmented M2 macrophage polarization through the MAPK signaling pathway. CONCLUSIONS: These findings suggest that inhibiting IRE1α can modulate macrophage polarization and alleviate ligature-induced diabetic periodontitis via the MAPK signaling pathway. This unveils a novel mechanism, offering a scientific foundation for the treatment of experimental diabetic periodontitis.


Sujet(s)
Diabète de type 2 , Stress du réticulum endoplasmique , Endoribonucleases , Macrophages , Parodontite , Protein-Serine-Threonine Kinases , Animaux , Humains , Mâle , Souris , Résorption alvéolaire/immunologie , Cellules cultivées , Diabète expérimental/immunologie , Diabète expérimental/métabolisme , Diabète de type 2/immunologie , Diabète de type 2/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/immunologie , Endoribonucleases/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Parodontite/immunologie , Parodontite/métabolisme , Protein-Serine-Threonine Kinases/métabolisme
13.
Int Immunopharmacol ; 132: 112019, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38599099

RÉSUMÉ

OBJECTIVE: The current study aimed to assess the modulating effect of IL-2 encapsulated chitosan-nanoparticles (CSNPs) on the function of Treg cells through induction of type 1 diabetes (T1D). Treg cell function was monitored by the forkhead box P3 (FoxP3) and transforming growth factor beta (TGFß) levels, correlating them with blood glucose and serum insulin levels. MATERIALS AND METHODS: In this case-control study, a low dose of IL-2 (free and chitosan-loaded) was injected into a diabetic mice group. The levels of FoxP3 and TGF-ß 1 were assessed using Enzyme-Linked Immunosorbent Assay. In addition, blood glucose and serum insulin levels were determined. RESULTS: The mean glucose level decreased significantly after free rIL-2 or rIL-2 / CSNPs treatment. Meanwhile, the mean serum insulin level was significantly increased after treatment with free rIL-2 or rIL-2/CSNPs. The mean levels of FoxP3 and TGFß 1 were significantly increased with either free rIL-2 or rIL-2/CSNPs compared to the T1D untreated group (P < 0.001). In the treated mice group receiving free CSNPs, there was a significant negative correlation between glucose and insulin levels. Moreover, FoxP3 & TGFß 1 levels had a significant positive correlation. In treated mice groups with free rIL-2 and IL-2 CSNPs, there was a significant positive correlation between FoxP3 and glucose levels. A significant negative correlation was found after conducting a correlation between insulin level and FoxP3 in the T1D/ rIL-2 / CSNPs group. CONCLUSIONS: Low-dose IL-2 selectively modulates FoxP3 + Tregs, and TGFß 1 increases their levels. These results demonstrated that IL-2-free and chitosan-loaded nanoparticles can be therapeutic agents in T1D.


Sujet(s)
Glycémie , Chitosane , Diabète expérimental , Diabète de type 1 , Facteurs de transcription Forkhead , Insuline , Interleukine-2 , Nanoparticules , Lymphocytes T régulateurs , Animaux , Chitosane/composition chimique , Chitosane/administration et posologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Interleukine-2/métabolisme , Interleukine-2/sang , Diabète expérimental/traitement médicamenteux , Diabète expérimental/immunologie , Glycémie/effets des médicaments et des substances chimiques , Souris , Diabète de type 1/traitement médicamenteux , Diabète de type 1/immunologie , Diabète de type 1/sang , Facteurs de transcription Forkhead/métabolisme , Insuline/sang , Mâle , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/sang , Streptozocine , Humains
14.
Acta Pharmacol Sin ; 45(7): 1466-1476, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38514862

RÉSUMÉ

Disturbances in intestinal immune homeostasis predispose susceptible individuals to type 1 diabetes (T1D). G-protein-coupled receptor 41 (GPR41) is a receptor for short-chain fatty acids (SCFAs) mainly produced by gut microbiota, which plays key roles in maintaining intestinal homeostasis. In this study, we investigated the role of GPR41 in the progression of T1D. In non-obese diabetic (NOD) mice, we found that aberrant reduction of GPR41 expression in the pancreas and colons was associated with the development of T1D. GPR41-deficient (Gpr41-/-) mice displayed significantly exacerbated streptozotocin (STZ)-induced T1D compared to wild-type mice. Furthermore, Gpr41-/- mice showed enhanced gut immune dysregulation and increased migration of gut-primed IFN-γ+ T cells to the pancreas. In bone marrow-derived dendritic cells from Gpr41-/- mice, the expression of suppressor of cytokine signaling 3 (SOCS) was significantly inhibited, while the phosphorylation of STAT3 was significantly increased, thus promoting dendritic cell (DC) maturation. Furthermore, adoptive transfer of bone marrow-derived dendritic cells (BMDC) from Gpr41-/- mice accelerated T1D in irradiated NOD mice. We conclude that GPR41 is essential for maintaining intestinal and pancreatic immune homeostasis and acts as a negative regulator of DC maturation in T1D. GPR41 may be a potential therapeutic target for T1D.


Sujet(s)
Cellules dendritiques , Diabète expérimental , Diabète de type 1 , Souris de lignée NOD , Souris knockout , Récepteurs couplés aux protéines G , Streptozocine , Animaux , Diabète de type 1/immunologie , Diabète de type 1/métabolisme , Récepteurs couplés aux protéines G/déficit , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris , Diabète expérimental/métabolisme , Diabète expérimental/immunologie , Souris de lignée C57BL , Facteur de transcription STAT-3/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Interféron gamma/métabolisme , Pancréas/métabolisme , Pancréas/anatomopathologie , Pancréas/immunologie , Mâle , Femelle , Microbiome gastro-intestinal
15.
Endocrine ; 85(2): 626-637, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38424350

RÉSUMÉ

AIM: Much focus of immunotherapy for type 1 diabetes (T1D) has been devoted on selectively boosting regulatory T (Treg) cells using low dose IL-2 due to their constitutive expression of IL-2Rα, CD25. However, several clinical trials using a low dose of IL-2 only showed a limited improvement of metabolic control. It can therefore be hypothesized that further decreasing IL-2 dosage may increase the selective responsiveness of Treg cells. METHODS: We induced experimental T1D using multiple low dose streptozotocin (STZ) injections and treated the mice with an ultra-low dose IL-2 (uIL-2, approximately 7-fold lower than low dose). Immune response was studied using multicolor flow cytometry. RESULTS: We found that uIL-2 did not protect STZ mice from developing hyperglycemia. It did neither increase Treg cell proportions, nor did it correct the phenotypic shift of Treg cells seen in T1D. It only partially decreased the proportion of IFN-γ+ T cells. Likewise, uIL-2 also did not protect the dysfunction of regulatory B (Breg) cells. Strikingly, when administered in combination with an anti-inflammatory cytokine IL-35, uIL-2 abrogated IL-35's protective effect. Low dose IL-2, on the other hand, protected half of the STZ mice from developing hyperglycemia. No difference was found in the Treg and Breg response, and it only tended to decrease CD80 expression in macrophages and dendritic cells. CONCLUSION: In conclusion, further decreasing IL-2 dosage may not be a suitable approach for T1D therapy, and the limited success suggests that an alternative low dose IL-2 therapy strategy or other immunotherapies should be considered.


Sujet(s)
Diabète expérimental , Diabète de type 1 , Interleukine-2 , Lymphocytes T régulateurs , Animaux , Diabète de type 1/traitement médicamenteux , Diabète de type 1/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Souris , Diabète expérimental/traitement médicamenteux , Diabète expérimental/immunologie , Interleukines , Mâle , Souris de lignée C57BL , Immunothérapie/méthodes
16.
Immunol Res ; 72(4): 644-653, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38347341

RÉSUMÉ

Diabetes mellitus (DM) is a major risk factor for tuberculosis (TB), though the underlying mechanisms linking DM and TB remain ambiguous. Macrophages are a key player in the innate immune response and their phagocytic ability is enhanced in response to microbial infections. Upon infection or inflammation, they also repel invading pathogens by generating; reactive oxygen species (ROS), reactive nitrogen species (RNS), pro-inflammatory cytokines (IL-1ß and IL-6), and anti-inflammatory cytokines (IL-10). However, the robustness of these innate defensive capabilities of macrophages when exposed to hyperglycemia remains unclear. In our current work, we explored the production of these host defense molecules in response to challenge with Mycobacterium tuberculosis (Mtb) infection and lipopolysaccharide (LPS) stimulation. Utilizing peritoneal macrophages from high-fat diet + streptozotocin induced diabetic mice and hyperglycemic THP-1-derived macrophages as model systems; we found that LPS stimulation and Mtb infection were ineffective in stimulating the production of ROS, RNS, and pro-inflammatory cytokines in cells exposed to hyperglycemia. On the contrary, an increase in production of anti-inflammatory cytokines was observed. To confirm the mechanism of decreased anti-bacterial activity of the diabetic macrophage, we explored activation status of these compromised macrophages and found decreased surface expression of activation (TLR-4) and differentiation markers (CD11b and CD11c). We postulate that this could be the cause for higher susceptibility for Mtb infection among diabetic individuals.


Sujet(s)
Cytokines , Hyperglycémie , Mycobacterium tuberculosis , Espèces réactives de l'oxygène , Tuberculose , Animaux , Mycobacterium tuberculosis/immunologie , Souris , Tuberculose/immunologie , Tuberculose/microbiologie , Hyperglycémie/immunologie , Humains , Cytokines/métabolisme , Espèces réactives de l'oxygène/métabolisme , Diabète expérimental/immunologie , Lipopolysaccharides/immunologie , Mâle , Souris de lignée C57BL , Macrophages/immunologie , Cellules THP-1 , Activation des macrophages/immunologie , Macrophages péritonéaux/immunologie , Maladie chronique , Espèces réactives de l'azote/métabolisme , Immunité innée , Récepteur de type Toll-4/métabolisme
17.
Transplantation ; 108(5): 1115-1126, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38192025

RÉSUMÉ

BACKGROUND: The utilization of islet-like cells derived from pluripotent stem cells may resolve the scarcity of islet transplantation donors. The subcutaneous space is a promising transplantation site because of its capacity for graft observation and removal, thereby ensuring safety. To guarantee subcutaneous islet transplantation, physicians should ensure ample blood supply. Numerous methodologies, including prevascularization, have been investigated to augment blood flow, but the optimal approach remains undetermined. METHODS: From C57BL/6 mice, 500 syngeneic islets were transplanted into the prevascularized subcutaneous site of recipient mice by implanting agarose rods with basic fibroblast growth factor at 1 and 2 wk. Before transplantation, the blood glucose levels, cell infiltration, and cytokine levels at the transplant site were evaluated. Furthermore, we examined the impact of the extracellular matrix capsule on graft function and the inflammatory response. RESULTS: Compared with the 1-wk group, the 2-wk group exhibited improved glycemic control, indicating that longer prevascularization enhanced transplant success. Flow cytometry analysis detected immune cells, such as neutrophils and macrophages, in the extracellular matrix capsules, whereas cytometric bead array analysis indicated the release of inflammatory and proinflammatory cytokines. Treatment with antitumor necrosis factor and anti-interleukin-6R antibodies in the 1-wk group improved graft survival, similar to the 2-wk group. CONCLUSIONS: In early prevascularization before subcutaneous transplantation, neutrophil and macrophage accumulation prevented early engraftment owing to inflammatory cytokine production.


Sujet(s)
Glycémie , Cytokines , Survie du greffon , Transplantation d'ilots de Langerhans , Souris de lignée C57BL , Transplantation d'ilots de Langerhans/méthodes , Transplantation d'ilots de Langerhans/immunologie , Animaux , Glycémie/métabolisme , Cytokines/métabolisme , Souris , Mâle , Facteurs temps , Diabète expérimental/immunologie , Diabète expérimental/chirurgie , Tissu sous-cutané/vascularisation , Tissu sous-cutané/immunologie , Matrice extracellulaire/métabolisme , Ilots pancréatiques/immunologie , Ilots pancréatiques/vascularisation , Néovascularisation physiologique
18.
Biochem Pharmacol ; 216: 115764, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37634595

RÉSUMÉ

Development of specific therapies that target and accelerate diabetic wound repair is an urgent need to alleviate pain and suffering and the huge socioeconomic burden of this debilitating disease. C-X-C Motif Chemokine Ligand 12 (CXCL12) also know an stromal cell-derived factor 1α (SDF-1α) is a chemokine that binds the CXC chemokine receptor type 4 (CXCR4) and activates downstream signaling resulting in recruitment of hematopoietic cells to locations of tissue injury and promotes tissue repair. In diabetes, low expression of CXCL12 correlates with impaired wound healing. Activation of CXCR4 receptor signaling with agonists or positive allosteric modulators (PAMs) provides a potential for small molecule therapeutic discovery and development. We recently reported high throughput screening and identification of the CXCR4 partial agonist UCUF-728, characterization of in vitro activity and reduced wound closure time in diabetic mice at 100 µM as a proof-of-concept study. We report here, the discovery of a second chemical scaffold demonstrating increased agonist potency and represented by thiadiazine derivative, UCUF-965. UCUF-965 is a potent partial agonist of ß-arrestin recruitment in CXCR4 receptor overexpressing cell line. Furthermore, UCUF-965 potentiates the CXCL12 maximal response in cAMP signaling pathway, activates CXCL12 stimulated migration in lymphoblast cells and modulates the levels of specific microRNA involved in the complex wound repair process, specifically in mouse fibroblasts. Our results indicate that UCUF-965 acts as a PAM agonist of the CXCR4 receptor. Furthermore, UCUF-965 enhanced angiogenesis markers and reduced wound healing time by 36% at 10.0 µM in diabetic mice models compared to untreated control.


Sujet(s)
Diabète expérimental , Récepteurs CXCR4 , Cicatrisation de plaie , Animaux , Souris , Mouvement cellulaire/physiologie , Chimiokine CXCL12/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/génétique , Diabète expérimental/immunologie , Cellules souches hématopoïétiques , Récepteurs CXCR4/agonistes , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , Transduction du signal , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Cicatrisation de plaie/génétique , Cicatrisation de plaie/physiologie
19.
Nutr Diabetes ; 13(1): 4, 2023 04 08.
Article de Anglais | MEDLINE | ID: mdl-37031163

RÉSUMÉ

BACKGROUND: Lentinan (LNT) is a complex fungal component that possesses effective antitumor and immunostimulating properties. However, there is a paucity of studies regarding the effects and mechanisms of LNT on type 1 diabetes. OBJECTIVE: In the current study, we investigated whether an intraperitoneal injection of LNT can diminish the risk of developing type 1 diabetes (T1D) in non-obese diabetic (NOD) mice and further examined possible mechanisms of LNT's effects. METHODS: Pre-diabetic female NOD mice 8 weeks of age, NOD mice with 140-160 mg/dL, 200-230 mg/dL or 350-450 mg/dL blood glucose levels were randomly divided into two groups and intraperitoneally injected with 5 mg/kg LNT or PBS every other day. Then, blood sugar levels, pancreas slices, spleen, PnLN and pancreas cells from treatment mice were examined. RESULTS: Our results demonstrated that low-dosage injections (5 mg/kg) of LNT significantly suppressed immunopathology in mice with autoimmune diabetes but increased the Foxp3+ regulatory T cells (Treg cells) proportion in mice. LNT treatment induced the production of Tregs in the spleen and PnLN cells of NOD mice in vitro. Furthermore, the adoptive transfer of Treg cells extracted from LNT-treated NOD mice confirmed that LNT induced Treg function in vivo and revealed an enhanced suppressive capacity as compared to the Tregs isolated from the control group. CONCLUSION: LNT was capable of stimulating the production of Treg cells from naive CD4 + T cells, which implies that LNT exhibits therapeutic values as a tolerogenic adjuvant and may be used to reverse hyperglycaemia in the early and late stages of T1D.


Sujet(s)
Diabète de type 1 , Lentinane , État prédiabétique , Lymphocytes T régulateurs , Animaux , Femelle , Souris , Diabète expérimental/immunologie , Diabète expérimental/prévention et contrôle , Diabète de type 1/traitement médicamenteux , Diabète de type 1/immunologie , Diabète de type 1/prévention et contrôle , Injections péritoneales , Lentinane/administration et posologie , Lentinane/immunologie , Lentinane/pharmacologie , Lentinane/usage thérapeutique , Souris de lignée NOD , État prédiabétique/traitement médicamenteux , État prédiabétique/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie
20.
Int J Mol Sci ; 23(21)2022 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-36362050

RÉSUMÉ

Diabetic kidney disease (DKD) frequently leads to end-stage renal disease and other life-threatening illnesses. The dysregulation of glomerular cell types, including mesangial cells, endothelial cells, and podocytes, appears to play a vital role in the development of DKD. Myeloid-derived suppressor cells (MDSCs) exhibit immunoregulatory and anti-inflammatory properties through the depletion of L-arginine that is required by T cells, through generation of oxidative stress, interference with T-cell recruitment and viability, proliferation of regulatory T cells, and through the promotion of pro-tumorigenic functions. Under hyperglycemic conditions, mouse mesangial cells reportedly produce higher levels of fibronectin and pro-inflammatory cytokines. Moreover, the number of MDSCs is noticeably decreased, weakening inhibitory immune activities, and creating an inflammatory environment. In diabetic mice, immunotherapy with MDSCs that were induced by a combination of granulocyte-macrophage colony-stimulating factor, interleukin (IL)-1ß, and IL-6, reduced kidney to body weight ratio, fibronectin expression, and fibronectin accumulation in renal glomeruli, thus ameliorating DKD. In conclusion, MDSCs exhibit anti-inflammatory activities that help improve renal fibrosis in diabetic mice. The therapeutic targeting of the proliferative or immunomodulatory pathways of MDSCs may represent an alternative immunotherapeutic strategy for DKD.


Sujet(s)
Diabète expérimental , Néphropathies diabétiques , Cellules myéloïdes suppressives , Animaux , Souris , Diabète expérimental/immunologie , Diabète expérimental/métabolisme , Néphropathies diabétiques/immunologie , Néphropathies diabétiques/métabolisme , Cellules endothéliales/métabolisme , Fibronectines/métabolisme , Souris de lignée C57BL , Cellules myéloïdes suppressives/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE