Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.555
Filtrer
1.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125655

RÉSUMÉ

Pancreatic cancer is a very aggressive disease with a dismal prognosis. The tumor microenvironment exerts immunosuppressive activities through the secretion of several cytokines, including interleukin (IL)-1. The IL-1/IL-1 receptor (IL-1R) axis is a key regulator in tumor-promoting T helper (Th)2- and Th17-type inflammation. Th2 cells are differentiated by dendritic cells endowed with Th2-polarizing capability by the thymic stromal lymphopoietin (TSLP) that is secreted by IL-1-activated cancer-associated fibroblasts (CAFs). Th17 cells are differentiated in the presence of IL-1 and other IL-1-regulated cytokines. In pancreatic cancer, the use of a recombinant IL-1R antagonist (IL1RA, anakinra, ANK) in in vitro and in vivo models has shown efficacy in targeting the IL-1/IL-1R pathway. In this study, we have developed sphingomyelin nanosystems (SNs) loaded with ANK (ANK-SNs) to compare their ability to inhibit Th2- and Th17-type inflammation with that of the free drug in vitro. We found that ANK-SNs inhibited TSLP and other pro-tumor cytokines released by CAFs at levels similar to ANK. Importantly, inhibition of IL-17 secretion by Th17 cells, but not of interferon-γ, was significantly higher, and at lower concentrations, with ANK-SNs compared to ANK. Collectively, the use of ANK-SNs might be beneficial in reducing the effective dose of the drug and its toxic effects.


Sujet(s)
Antagoniste du récepteur à l'interleukine-1 , Interleukine-1 , Tumeurs du pancréas , Sphingomyéline , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Humains , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Interleukine-1/métabolisme , Sphingomyéline/métabolisme , Cytokines/métabolisme , Lignée cellulaire tumorale , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Lymphocytes auxiliaires Th2/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th2/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques
2.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 494-500, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38952088

RÉSUMÉ

Objective To investigate the effect of Terminalia chebula water extract (TCWE) on the cellular immunity and PD-1/PD-L1 pathway in rats with collagen-induced arthritis (CIA). Methods SD rats were randomly divided into four groups: a control group, a CIA group, a TCWE group and a methotrexate (MTX) group, with 15 rats in each group. Except for the control group, SD rats in other groups were subcutaneously injected with type II collagen to establish the model of collagen-induced arthritis (CIA). The rats in the TCWE group were treated with 20 mg/(kg.d) TCWE and the rats in the MTX group were treated with 1.67 mg/(kg.d) MTX. After 14 days of treatment, the cartilage morphology was examined using hematoxylin-eosin (HE) staining, and splenic T lymphocyte apoptosis and Treg/Th17 cell ratio were detected by flow cytometry. The mRNA expressions of retinoid-related orphan nuclear receptor γt (RORγt), forkhead box P3 (FOXP3), PD-1 and PD-L1 in spleen were detected by reverse transcription PCR. The expression and localization of RORγt and FOXP3 were detected by immunohistochemical staining. The protein expressions of PD-1 and PD-L1 in splenic lymphocytes were detected by Western blot, and the levels of serum interleukin 17 (IL-17) and transforming growth factor ß (TGF-ß) in rats were detected by ELISA. Results Compared with CIA group, the pathological changes of cartilage and synovium were significantly alleviated in the TCWE group and the MTX group. Both the apoptosis rate of T lymphocytes in spleen and the ratio of Treg/Th17 cells increased. The expression of RORγt decreased, while the expressions of FOXP3, PD-1 and PD-L1 increased in spleen lymphocytes. The level of serum IL-17 decreased, while the level of serum TGF-ß increased. Conclusion TCWE treatment may activate PD-1/PD-L1 pathway in spleen cells to regulate cellular immunity, thus reducing cartilage injury in CIA rats.


Sujet(s)
Arthrite expérimentale , Antigène CD274 , Récepteur-1 de mort cellulaire programmée , Rat Sprague-Dawley , Rate , Terminalia , Animaux , Arthrite expérimentale/immunologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Rate/effets des médicaments et des substances chimiques , Rate/immunologie , Rate/métabolisme , Antigène CD274/génétique , Antigène CD274/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , Rats , Terminalia/composition chimique , Mâle , Immunité cellulaire/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/génétique , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/métabolisme , Inflammation/traitement médicamenteux , Inflammation/immunologie , Inflammation/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/métabolisme , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme
3.
Front Cell Infect Microbiol ; 14: 1362773, 2024.
Article de Anglais | MEDLINE | ID: mdl-39081865

RÉSUMÉ

Coumarin, a phenolic compound, is a secondary metabolite produced by plants such as Tanga and Lime. Coumarin derivatives were prepared via Pechmann condensation. In this study, we performed in vitro and in vivo experiments to determine the antimicrobial and gut immune-regulatory functions of coumarin derivatives. For the in vitro antimicrobial activity assay, coumarin derivatives C1 and C2 were selected based on their pathogen-killing activity against various pathogenic microbes. We further demonstrated that the selected coumarin derivatives disrupted bacterial cell membranes. Next, we examined the regulatory function of the coumarin derivatives in gut inflammation using an infectious colitis model. In an in vivo infectious colitis model, administration of selected C1 coumarin derivatives reduced pathogen loads, the number of inflammatory immune cells (Th1 cells and Th17 cells), and inflammatory cytokine levels (IL-6 and IL-1b) in the intestinal tissue after pathogen infection. In addition, we found that the administration of C1 coumarin derivatives minimized abnormal gut microbiome shift-driven pathogen infection. Potential pathogenic gut microbes, such as Enterobacteriaceae and Staphylococcaceae, were increased by pathogen infection. However, this pathogenic microbial expansion was minimized and beneficial bacteria, such as Ligilactobacillus and Limosilactobacillus, increased with C1 coumarin derivative treatment. Functional gene enrichment assessment revealed that the relative abundance of genes associated with lipid and nucleotide metabolism was reduced by pathogen infection; however, this phenomenon was not observed in C1 coumarin derivative-treated animals. Collectively, our data suggest that C1 coumarin derivative is effective antibacterial agents that minimize pathogen-induced gut inflammation and abnormal gut microbiome modulation through their antibacterial activity.


Sujet(s)
Antibactériens , Colite , Coumarines , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Coumarines/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Animaux , Colite/microbiologie , Colite/traitement médicamenteux , Antibactériens/pharmacologie , Souris , Cytokines/métabolisme , Bactéries/effets des médicaments et des substances chimiques , Bactéries/classification , Souris de lignée C57BL , Inflammation/traitement médicamenteux , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Mâle
4.
Int Immunopharmacol ; 138: 112597, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38955025

RÉSUMÉ

BACKGROUND: Guillain-Barré syndrome (GBS) is an auto-inflammatory peripheral nerve disease. Dendritic cell-mediated T cell polarization is of pivotal importance in demyelinating lesions of peripheral nerves and nerve roots. However, the regulatory function of VX-509 (Decernotinib)-modified tolerogenic dendritic cells (VX-509-tolDCs) during immune remodeling following GBS remains unclear. Here, we used experimental autoimmune neuritis (EAN) as a model to investigate these aspects of GBS. METHODS: DCs were treated with varying concentrations of VX-509 (0.25, 1, and 4 µM) or served as a control using 10-8 M 1,25-(OH)2D3. Flow cytometry was employed to assess the apoptosis, phenotype, and capacity to induce T cell responses of the treated DCs. In the in vivo experiments, EAN mice received administration of VX-509-tolDCs or 1,25-(OH)2D3-tolDCs via the tail vein at a dose of 1x106 cells/mouse on days 5, 9, 13, and 17. RESULTS: VX-509 inhibited the maturation of DCs and promoted the development of tolDCs. The function of antigen-specific CD4 + T cells ex vivo was influenced by VX-509-tolDCs. Furthermore, the adoptive transfer of VX-509-tolDCs effectively alleviated inflammatory demyelinating lesions in EAN by promoting Th17/Treg (T helper 17 and regulatory T cells) rebalance. CONCLUSION: The adoptive transfer of VX-509-tolDCs alleviated inflammatory demyelinating lesions in a mouse model of GBS, known as the EAN mouse, by partially restoring the balance between Treg and Th17 cells.


Sujet(s)
Cellules dendritiques , Souris de lignée C57BL , Névrite auto-immune expérimentale , Lymphocytes T régulateurs , Cellules Th17 , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/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 , Névrite auto-immune expérimentale/immunologie , Névrite auto-immune expérimentale/traitement médicamenteux , Souris , Tolérance immunitaire/effets des médicaments et des substances chimiques , Cellules cultivées , Femelle , Modèles animaux de maladie humaine , Mâle , Humains
5.
Skelet Muscle ; 14(1): 16, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026344

RÉSUMÉ

BACKGROUND: This study aims to investigate the involvement of acid sphingomyelinase (ASM) in the pathology of dermatomyositis (DM), making it a potential therapeutic target for DM. METHODS: Patients with DM and healthy controls (HCs) were included to assess the serum level and activity of ASM, and to explore the associations between ASM and clinical indicators. Subsequently, a myositis mouse model was established using ASM gene knockout and wild-type mice to study the significant role of ASM in the pathology and to assess the treatment effect of amitriptyline, an ASM inhibitor. Additionally, we investigated the potential treatment mechanism by targeting ASM both in vivo and in vitro. RESULTS: A total of 58 DM patients along with 30 HCs were included. The ASM levels were found to be significantly higher in DM patients compared to HCs, with median (quartile) values of 2.63 (1.80-4.94) ng/mL and 1.64 (1.47-1.96) ng/mL respectively. The activity of ASM in the serum of DM patients was significantly higher than that in HCs. Furthermore, the serum levels of ASM showed correlations with disease activity and muscle enzyme levels. Knockout of ASM or treatment with amitriptyline improved the severity of the disease, rebalanced the CD4 T cell subsets Th17 and Treg, and reduced the production of their secreted cytokines. Subsequent investigations revealed that targeting ASM could regulate the expression of relevant transcription factors and key regulatory proteins. CONCLUSION: ASM is involved in the pathology of DM by regulating the differentiation of naive CD4 + T cells and can be a potential treatment target.


Sujet(s)
Amitriptyline , Différenciation cellulaire , Dermatomyosite , Souris knockout , Sphingomyeline phosphodiesterase , Lymphocytes T régulateurs , Cellules Th17 , Dermatomyosite/traitement médicamenteux , Dermatomyosite/immunologie , Dermatomyosite/génétique , Humains , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Mâle , Femelle , Adulte d'âge moyen , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Amitriptyline/pharmacologie , Amitriptyline/usage thérapeutique , Adulte , Souris , Sphingomyeline phosphodiesterase/génétique , Sphingomyeline phosphodiesterase/métabolisme , Souris de lignée C57BL
6.
Sci Rep ; 14(1): 17241, 2024 07 26.
Article de Anglais | MEDLINE | ID: mdl-39060348

RÉSUMÉ

Studies have demonstrated that prior to puberty, girls have a lower incidence and severity of asthma symptoms compared to boys. This study aimed to explore the role of progesterone (P4), a sex hormone, in reducing inflammation and altering the immune microenvironment in a mouse model of allergic asthma induced by OVA. Female BALB/c mice with or without ovariectomy to remove the influence of sex hormones were used for the investigations. Serum, bronchoalveolar lavage fluid (BALF), and lung tissue samples were collected for analysis. The results indicated that P4 treatment was effective in decreasing inflammation and mucus secretion in the lungs of OVA-induced allergic asthma mice. P4 treatment also reduced the influx of inflammatory cells into the BALF and increased the levels of Th1 and Th17 cytokines while decreasing the levels of Th2 and Treg cytokines in both BALF and lung microenvironment CD45+ T cells. Furthermore, P4 inhibited the infiltration of inflammatory cells into the lungs, suppressed NETosis, and reduced the number of pulmonary CD4+ T cells while increasing the number of regulatory T cells. The neutrophil elastase inhibitor GW311616A also suppressed airway inflammation and mucus production and modified the secretion of immune Th1, Th2, Th17, and Treg cytokines in lung CD45+ immune cells. These changes led to an alteration of the immunological milieu with increased Th1 and Th17 cells, accompanied by decreased Th2, Treg, and CD44+ T cells, similar to the effects of P4 treatment. Treatment with P4 inhibited NETosis by suppressing the p38 pathway activation, leading to reduced reactive oxygen species production. Moreover, P4 treatment hindered the release of double-stranded DNA during NETosis, thereby influencing the immune microenvironment in the lungs. These findings suggest that P4 treatment may be beneficial in reducing inflammation associated with allergic asthma by modulating the immune microenvironment. In conclusion, this research indicates the potential of P4 as a therapeutic agent for ameliorating inflammation in OVA-induced allergic asthma mice.


Sujet(s)
Asthme , Ovalbumine , Progestérone , Animaux , Femelle , Souris , Asthme/immunologie , Asthme/traitement médicamenteux , Asthme/métabolisme , Liquide de lavage bronchoalvéolaire , Microenvironnement cellulaire/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Modèles animaux de maladie humaine , Pièges extracellulaires/effets des médicaments et des substances chimiques , Pièges extracellulaires/métabolisme , Pièges extracellulaires/immunologie , Inflammation/traitement médicamenteux , Inflammation/immunologie , Inflammation/métabolisme , Poumon/immunologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Souris de lignée BALB C , Ovalbumine/immunologie , Progestérone/pharmacologie , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme
7.
Biochem Pharmacol ; 227: 116428, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39009096

RÉSUMÉ

Sepsis-associated encephalopathy (SAE) is the main cause of cognitive impairment in patients with sepsis. The infiltration of inflammatory signals into the central nervous system (CNS) via the compromised blood-brain barrier (BBB) represents a crucial step in the pathological progression of SAE. In particular, T-helper 17 cell (Th17 cells) has been suggested to be highly correlated with the activation of central immune responses. Thus, preventing Th17 cell infiltration into the CNS may be a possible strategy to alleviate cognitive decline in SAE. Dipsacoside B (DB) is one of the primary active components in Chuan Xu Duan (Dipsacus asper Wall). We speculate that DB may be a potential candidate for the treatment of SAE-related cognitive deficits. In the present study, we demonstrated that DB could effectively alleviate cognitive impairment in SAE mice. DB significantly suppressed the central inflammatory response induced by repeated lipopolysaccharide (LPS) injection. The mechanism underlying its therapeutic effect should be attributed to the reduction of BBB impairment and pathogenic Th17 cell infiltration into the CNS by inhibition of vascular endothelial growth factor A (VEGFA)/ Vascular endothelial growth factor receptor 2(VEGFR2)/ Endothelial nitric oxide synthase (eNOS) signaling. Our findings suggest that DB is a potential candidate for the treatment of SAE-related cognitive dysfunction.


Sujet(s)
Dysfonctionnement cognitif , Souris de lignée C57BL , Maladies neuro-inflammatoires , Encéphalopathie associée au sepsis , Cellules Th17 , Animaux , Souris , Encéphalopathie associée au sepsis/traitement médicamenteux , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Cellules Th17/métabolisme , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/étiologie , Mâle , Maladies neuro-inflammatoires/traitement médicamenteux , Saponines/pharmacologie , Saponines/usage thérapeutique
8.
Environ Sci Pollut Res Int ; 31(35): 48758-48772, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39039370

RÉSUMÉ

Aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are mycotoxins widely found as cereal contaminants, and their co-consumption is associated with liver cancer. Both are immunotoxic, but their interactions have been little studied. This work was aimed to evaluate in mouse spleen mononuclear cells (SMC) the effects of the exposure to AFB1 (5-50 µM), FB1 (25-250 µM), and AFB1-FB1 mixtures (MIX) on the in vitro differentiation of regulatory T cells (Treg and Tr1-like) and Th17 cells, as well as elucidate the contribution of aryl hydrocarbon receptor (Ahr) in such effects. AFB1 and mainly MIX induced cytotoxicity in activated CD4 cells via Ahr signaling. AFB1 (5 µM) increased the Treg cell differentiation, but its combination with FB1 (25 µM) also reduced Th17 cell expansion by Ahr-dependent mechanisms. Therefore, this mixture could enhance the Treg/Th17 cell ratio and favor immunosuppression and escape from tumor immunosurveillance to a greater extent than individual mycotoxins. Whereas, AFB1-FB1 mixtures at medium-high doses inhibited the Tr1-like cell expansion induced by the individual mycotoxins and affected Treg and Th17 cell differentiation in Ahr-independent and dependent manners, respectively, which could alter anti-inflammatory and Th17 immune responses. Moreover, individual FB1 altered regulatory T and Th17 cell development independently of Ahr. In conclusion, AFB1 and FB1 interact by modifying Ahr signaling, which is involved in the immunotoxicity as well as in the alteration of the differentiation of Treg, Tr1-like, and Th17 cells induced by AFB1-FB1 mixtures. Therefore, Ahr is implicated in the regulation of the anti- and pro-inflammatory responses caused by the combination of AFB1 and FB1.


Sujet(s)
Aflatoxine B1 , Différenciation cellulaire , Fumonisines , Récepteurs à hydrocarbure aromatique , Lymphocytes T régulateurs , Cellules Th17 , Récepteurs à hydrocarbure aromatique/métabolisme , Aflatoxine B1/toxicité , Animaux , Cellules Th17/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Fumonisines/toxicité , Souris , Différenciation cellulaire/effets des médicaments et des substances chimiques
9.
J Neuroinflammation ; 21(1): 181, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068463

RÉSUMÉ

Treating Multiple sclerosis (MS), a well-known immune-mediated disease characterized by axonal demyelination, is challenging due to its complex causes. Naphthalenedione, present in numerous plants, is being explored as a potential medicine for MS due to its immunomodulatory properties. However, its effects on lymphocytes can vary depending on factors such as the specific compound, concentration, and experimental conditions. In this study, we aim to explore the therapeutic potential of 2-bromo-1,4-naphthalenedione (BrQ), a derivative of naphthalenedione, in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, and to elucidate its underlying mechanisms. We observed that mice treated with BrQ exhibited reduced severity of EAE symptoms, including lower clinical scores, decreased leukocyte infiltration, and less extensive demyelination in central nervous system. Furthermore, it was noted that BrQ does not directly affect the remyelination process. Through cell-chat analysis based on bulk RNA-seq data, coupled with validation of flow analysis, we discovered that BrQ significantly promotes the expansion of CD8+ T cells and their interactions with other immune cells in peripheral immune system in EAE mice. Subsequent CD8+ T cell depletion experiments confirmed that BrQ alleviates EAE in a CD8+ T cell-dependent manner. Mechanistically, expanded CD8+ cells were found to selectively reduce antigen-specific CD4+ cells and subsequently inhibit Th1 and Th17 cell development in vivo, ultimately leading to relief from EAE. In summary, our findings highlight the crucial role of BrQ in modulating the pathogenesis of MS, suggesting its potential as a novel drug candidate for treating MS and other autoimmune diseases.


Sujet(s)
Lymphocytes T CD8+ , Encéphalomyélite auto-immune expérimentale , Souris de lignée C57BL , Lymphocytes auxiliaires Th1 , Cellules Th17 , Animaux , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Souris , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th1/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Femelle , Naphtalènes/pharmacologie , Naphtalènes/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques
10.
Mol Cell Probes ; 76: 101969, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964425

RÉSUMÉ

The progression and pathogenesis of membranous glomerulonephritis (MGN) are inextricably linked to chronic inflammation. Despite improving clinical remission rates due to the application of cyclophosphamide (CYC), treatment of MGN still requires further exploration. Ruxolitinib (Ruxo) negatively affects the signaling pathways participating in the production of pro-inflammatory cytokines. Hence, we investigated whether the combination of CYC and Ruxo can modulate inflammation through influencing T helper 17 (Th17) lineages and regulatory T cells (Tregs). Passive Heymann nephritis (PHN), an experimental model of MGN, was induced in a population of rats. Then, the animals were divided into five groups: PHN, CYC-receiving, Ruxo-receiving, CYC-Ruxo-receiving PHN rats, and healthy controls. After 28 days of treatment, biochemistry analysis was performed and splenocytes were isolated for flowcytometry investigation of Th17 cells and Tregs. The correlative transcription factors of the cells, alongside their downstream cytokine gene expressions, were also assessed using real-time PCR. Furthermore, serum cytokine signatures for the lymphocytes were determined through ELISA. The combination of CYC and Ruxo significantly reduced the serum values of urea in rats versus the PHN group (24.62 ± 7.970 vs. 40.60 ± 10.81 mg/dL). In contrast to Treg's activities, the functionality of Th17 cells noticeably increased not only in PHN rats but also in CYC or Ruxo-receiving PHN animals when compared with the control (10.60 ± 2.236, 8.800 ± 1.465, 8.680 ± 1.314 vs. 4.420 ± 1.551 %). However, in comparison to the PHN group, the incidence of Th17 cells notably fell in rats receiving CYC and Ruxo (10.60 ± 2.236 vs. 6.000 ± 1.373 %) in favor of the Treg's percentage (5.020 ± 1.761 vs. 8.980 ± 1.178 %), which was verified by the gene expressions and cytokine productions correlative to these lymphocytes. The combination of CYC and Ruxo was able to decline Th17 cells in favor of Tregs improvement in PHN rats, suggesting an innovative combination therapy in MGN treatment approaches.


Sujet(s)
Cyclophosphamide , Cytokines , Glomérulonéphrite extra-membraneuse , Nitriles , Pyrazoles , Pyrimidines , Lymphocytes T régulateurs , Cellules Th17 , Animaux , Glomérulonéphrite extra-membraneuse/traitement médicamenteux , Glomérulonéphrite extra-membraneuse/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Cyclophosphamide/pharmacologie , Cyclophosphamide/usage thérapeutique , Nitriles/pharmacologie , Nitriles/usage thérapeutique , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Rats , Pyrazoles/pharmacologie , Pyrazoles/usage thérapeutique , Cytokines/métabolisme , Mâle , Modèles animaux de maladie humaine , Association de médicaments
11.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-38928319

RÉSUMÉ

Matrine (MT) possesses anti-inflammatory, anti-allergic and antioxidative properties. However, the impact and underlying mechanisms of matrine on colitis are unclear. The purpose of this research was to examine the protective impact and regulatory mechanism of matrine on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. MT alleviated DSS-induced UC by inhibiting weight loss, relieving colon shortening and reducing the disease activity index (DAI). Moreover, DSS-induced intestinal injury and the number of goblet cells were reversed by MT, as were alterations in the expression of zonula occludens-1 (ZO-1) and occludin in colon. Simultaneously, matrine not only effectively restored DSS-induced oxidative stress in colonic tissues but also reduced the production of inflammatory cytokines. Furthermore, MT could treat colitis mice by regulating the regulatory T cell (Treg)/T helper 17 (Th17) cell imbalance. We observed further evidence that MT alleviated the decrease in intestinal flora diversity, reduced the proportion of Firmicutes and Bacteroidetes, decreased the proportion of Proteobacteria and increased the relative abundance of Lactobacillus and Akkermansia in colitis mice. In conclusion, these results suggest that MT may mitigate DSS-induced colitis by enhancing the colon barrier integrity, reducing the Treg/Th17 cell imbalance, inhibiting intestinal inflammation, modulating oxidative stress and regulating the gut microbiota. These findings provide strong evidence for the development and application of MT as a dietary treatment for UC.


Sujet(s)
Alcaloïdes , Sulfate dextran , Microbiome gastro-intestinal , , Stress oxydatif , Quinolizines , Lymphocytes T régulateurs , Animaux , Alcaloïdes/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Quinolizines/pharmacologie , Quinolizines/usage thérapeutique , Souris , Lymphocytes T régulateurs/métabolisme , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Mâle , Colite/induit chimiquement , Colite/traitement médicamenteux , Colite/métabolisme , Colite/microbiologie , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Protéine-1 de la zonula occludens/métabolisme , Côlon/anatomopathologie , Côlon/métabolisme , Côlon/effets des médicaments et des substances chimiques , Côlon/microbiologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Cellules Th17/immunologie , Modèles animaux de maladie humaine , Cytokines/métabolisme , Souris de lignée C57BL , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/microbiologie , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Occludine/métabolisme
12.
Nat Commun ; 15(1): 5413, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926424

RÉSUMÉ

Diet composition impacts metabolic health and is now recognized to shape the immune system, especially in the intestinal tract. Nutritional imbalance and increased caloric intake are induced by high-fat diet (HFD) in which lipids are enriched at the expense of dietary fibers. Such nutritional challenge alters glucose homeostasis as well as intestinal immunity. Here, we observed that short-term HFD induced dysbiosis, glucose intolerance and decreased intestinal RORγt+ CD4 T cells, including peripherally-induced Tregs and IL17-producing (Th17) T cells. However, supplementation of HFD-fed male mice with the fermentable dietary fiber fructooligosaccharides (FOS) was sufficient to maintain RORγt+ CD4 T cell subsets and microbial species known to induce them, alongside having a beneficial impact on glucose tolerance. FOS-mediated normalization of Th17 cells and amelioration of glucose handling required the cDC2 dendritic cell subset in HFD-fed animals, while IL-17 neutralization limited FOS impact on glucose tolerance. Overall, we uncover a pivotal role of cDC2 in the control of the immune and metabolic effects of FOS in the context of HFD feeding.


Sujet(s)
Cellules dendritiques , Alimentation riche en graisse , Homéostasie , Souris de lignée C57BL , Oligosaccharides , Animaux , Oligosaccharides/pharmacologie , Alimentation riche en graisse/effets indésirables , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/métabolisme , Mâle , Souris , Cellules Th17/immunologie , Cellules Th17/métabolisme , Cellules Th17/effets des médicaments et des substances chimiques , Glucose/métabolisme , Interleukine-17/métabolisme , Fibre alimentaire/pharmacologie , Intolérance au glucose/immunologie , Intolérance au glucose/métabolisme , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/métabolisme , Dysbiose/immunologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques
13.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892051

RÉSUMÉ

Dietary supplementation with n-3 polyunsaturated fatty acids (PUFA) has been found to be beneficial in rodent rheumatoid arthritis models and human trials. However, the molecular targets of n-3 PUFAs and their beneficial effects on rheumatoid arthritis are under-researched. Free fatty acid receptor 4 (FFA4, also known as GPR120) is a receptor for n-3 PUFA. We aim to investigate whether FFA4 activation reduces collagen-induced rheumatoid arthritis (CIA) by using an FFA4 agonist, compound A (CpdA), in combination with DBA-1J Ffa4 gene wild-type (WT) and Ffa4 gene knock-out (KO) mice. CIA induced an increase in the arthritis score, foot edema, synovial hyperplasia, pannus formation, proteoglycan loss, cartilage damage, and bone erosion, whereas the administration of CpdA significantly suppressed those increases in Ffa4 WT mice but not Ffa4 gene KO mice. CIA increased mRNA expression levels of pro-inflammatory Th1/Th17 cytokines, whereas CpdA significantly suppressed those increases in Ffa4 WT mice but not Ffa4 gene KO mice. CIA induced an imbalance between Th1/Th17 and Treg cells, whereas CpdA rebalanced them in spleens from Ffa4 WT mice but not Ffa4 gene KO mice. In SW982 synovial cells, CpdA reduced the LPS-induced increase in pro-inflammatory cytokine levels. In summary, the present results suggest that the activation of FFA4 in immune and synovial cells could suppress the characteristics of rheumatoid arthritis and be an adjuvant therapy.


Sujet(s)
Arthrite expérimentale , Souris knockout , Récepteurs couplés aux protéines G , Lymphocytes T régulateurs , Lymphocytes auxiliaires Th1 , Cellules Th17 , Animaux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/immunologie , Arthrite expérimentale/métabolisme , Arthrite expérimentale/traitement médicamenteux , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Cellules Th17/immunologie , Cellules Th17/métabolisme , Cellules Th17/effets des médicaments et des substances chimiques , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/agonistes , Souris , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/métabolisme , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Souris de lignée DBA , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/anatomopathologie , Mâle , Cytokines/métabolisme
14.
Ecotoxicol Environ Saf ; 281: 116662, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38944008

RÉSUMÉ

OBJECTIVE: This study aimed to investigate the mechanism that Lactobacillus murinus (L. murinus) alleviated lung inflammation induced by polycyclic aromatic hydrocarbons (PAHs) exposure based on metabolomics. METHODS: Female mice were administrated with PAHs mix, L. murinus and indoleacrylic acid (IA) or indolealdehyde (IAId). Microbial diversity in feces was detected by 16 S rRNA gene sequencing. Non-targeted metabolomics analysis in urine samples and targeted analysis of tryptophan metabolites in serum by UPLC-Orbitrap-MS and short-chain fatty acids (SCFA) in feces by GC-MS were performed, respectively. Flow cytometry was used to determine T helper immune cell differentiation in gut and lung tissues. The levels of IgE, IL-4 and IL-17A in the bronchoalveolar lavage fluid (BALF) or serum were detected by ELISA. The expressions of aryl hydrocarbon receptor (Ahr), cytochrome P450 1A1 (Cyp1a1) and forkheadbox protein 3 (Foxp3) genes and the histone deacetylation activity were detected by qPCR and by ELISA in lung tissues, respectively. RESULTS: PAHs exposure induced lung inflammation and microbial composition shifts and tryptophan metabolism disturbance in mice. L. murinus alleviated PAHs-induced lung inflammation and inhibited T helper cell 17 (Th17) cell differentiation and promoted regulatory T cells (Treg) cell differentiation. L. murinus increased the levels of IA and IAId in the serum and regulated Th17/Treg imbalance by activating AhR. Additionally, L. murinus restored PAHs-induced decrease of butyric acid and valeric acid which can reduce the histone deacetylase (HDAC) level in the lung tissues, enhancing the expression of the Foxp3 gene and promoting Treg cell differentiation. CONCLUSION: our study illustrated that L. murinus alleviated PAHs-induced lung inflammation and regulated Th17/Treg cell differentiation by regulating host tryptophan metabolism and SCFA levels. The study provided new insights into the reciprocal influence between gut microbiota, host metabolism and the immune system, suggesting that L. murinus might have the potential as a novel therapeutic strategy for lung diseases caused by environmental pollution in the future.


Sujet(s)
Lactobacillus , Pneumopathie infectieuse , Hydrocarbures aromatiques polycycliques , Animaux , Souris , Femelle , Hydrocarbures aromatiques polycycliques/toxicité , Pneumopathie infectieuse/induit chimiquement , Pneumopathie infectieuse/traitement médicamenteux , Récepteurs à hydrocarbure aromatique/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/immunologie , Tryptophane , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Probiotiques/pharmacologie , Probiotiques/usage thérapeutique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Liquide de lavage bronchoalvéolaire/cytologie , Liquide de lavage bronchoalvéolaire/composition chimique , Métabolomique , Cytochrome P-450 CYP1A1/métabolisme , Cytochrome P-450 CYP1A1/génétique
15.
Toxicol Appl Pharmacol ; 488: 116980, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823456

RÉSUMÉ

Multiple sclerosis (MS) is a class of autoimmune diseases mainly caused by the immune system attacking the myelin sheath of the axons in the nervous system. Although the pathogenesis of MS is complex, studies have shown that dendritic cells (DCs) play a vital role in the pathogenesis of MS. Quercetin (QU) has a unique advantage in clinical application, especially for treating autoimmune diseases. However, the mechanism of QU in the treatment of experimental autoimmune encephalomyelitis (EAE) remains unclear. In this study, we explore the potential role of QU in EAE. Finally, we find that QU has anti-inflammatory activities and neural protective effects in EAE. The experimental results suggest that the cellular basis for QU's function is to inhibit the activation of DCs while modulating the Th17 cell differentiation in the co-culture system. Further, QU may target STAT4 to inhibit its activation in DCs. This work will be of great significance for the future development and utilization of QU.


Sujet(s)
Cellules dendritiques , Encéphalomyélite auto-immune expérimentale , Souris de lignée C57BL , Quercétine , Facteur de transcription STAT-4 , Cellules Th17 , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/métabolisme , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Animaux , Quercétine/pharmacologie , Facteur de transcription STAT-4/métabolisme , Femelle , Souris , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Cellules Th17/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Techniques de coculture , Anti-inflammatoires/pharmacologie
16.
J Ethnopharmacol ; 334: 118463, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-38908493

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Wutou Decoction (WTD) is a classic traditional Chinese medicine formula, which has shown clinical efficacy in treating rheumatoid arthritis (RA). The Treg stability and Th17/Treg imbalance is an important immunological mechanism in RA progression. Whether WTD regulates CD4+ T cell subsets has not been thoroughly investigated yet. AIM OF THE STUDY: This study aimed to explore the potential role and mechanisms of WTD in regulating the diminished stability of Treg cells and the imbalance of CD4+ T cell subsets via in vivo and in vitro experiments. MATERIALS AND METHODS: Firstly, the therapeutic effects of WTD on the collagen-induced arthritis (CIA) mouse and its potential regulatory function on CD4+ T cell subsets were evaluated in vivo. Animal specimens were collected after 31 days of treatment with WTD. The anti-arthritic and anti-inflammatory effects of WTD were assessed through arthritis scoring, body weight, spleen index, serum IL-6 levels, and micro-PET/CT imaging. Gene enrichment analysis was performed to evaluate the activation T cell-related signaling pathway. Flow cytometry was used to determine the proportions of CD4+ T cell subsets in vitro and in vitro. Additionally, ELISA was used to assess the secretion of IL-10 and TGF-ß by Treg cells under inflammatory conditions. The suppressive function of Treg cells on cell proliferation under inflammatory conditions was examined using CFSE labeling. Immunofluorescence staining was performed to detect the phosphorylation levels of STAT3 in CD4+ T cells from mouse spleen tissues. Western blotting was used to evaluate the phosphorylation levels of JAK2/STAT3 in Treg cells. RESULTS: WTD significantly alleviated joint inflammation in CIA mice. WTD reduced serum IL-6 levels in CIA mice, improved their body weight and spleen index. WTD treatment inhibited the activation of CD4+ T cell subgroup-related signaling in the joint tissues of CIA mice. In vitro and in vitro experiments showed that WTD increased the proportion of Treg cells and decreased the proportion of Th17 cells in CIA mice spleen. Furthermore, WTD promoted the secretion of IL-10 and TGF-ß by Treg cells and enhanced the inhibitory capacity of Treg cells on cell proliferation under inflammatory conditions. Immunofluorescence detected decreased STAT3 phosphorylation levels in CD4+ T cells from CIA mice spleen, while western blotting revealed a decrease in JAK2/STAT3 phosphorylation levels in Treg cells in vitro. CONCLUSIONS: Inhibiting JAK2/STAT3 phosphorylation is a potential mechanism through which WTD improves Treg cell stability, balances CD4+ T cell subsets, and attenuates RA joint inflammation.


Sujet(s)
Arthrite expérimentale , Médicaments issus de plantes chinoises , Kinase Janus-2 , Souris de lignée DBA , Facteur de transcription STAT-3 , Transduction du signal , Lymphocytes T régulateurs , Cellules Th17 , Animaux , Facteur de transcription STAT-3/métabolisme , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Kinase Janus-2/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique
17.
Neuroscience ; 552: 65-75, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-38885894

RÉSUMÉ

Multiple sclerosis (MS) is an autoimmune inflammatory condition affecting the central nervous system, and experimental autoimmune encephalomyelitis (EAE) animal models have been extensively used to study it. T-helper 17 cells, which produce interleukin-17(IL-17), play crucial roles in MS pathogenesis, and the JAK2/STAT3 pathway has an essential function in their differentiation from naive CD4 + T cells. This study investigated the effects of the JAK2/STAT3 pathway inhibitor AG490 on EAE in vivo and in vitro, as well as the underlying mechanisms. AG490 ameliorated EAE severity and attenuated its typical symptoms by downregulating proteins associated with the JAK2/STAT3 pathway. Furthermore, it decreased T-helper 17 cell differentiation from naive CD4 + T cells by inactivating STAT3. In addition, it conferred protective effects against EAE by restoring autophagy. These findings indicate the potential of AG490 as a candidate anti-MS therapeutic.


Sujet(s)
Autophagie , Encéphalomyélite auto-immune expérimentale , Kinase Janus-2 , Souris de lignée C57BL , Facteur de transcription STAT-3 , Transduction du signal , Cellules Th17 , Tyrphostines , Animaux , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/métabolisme , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Facteur de transcription STAT-3/métabolisme , Tyrphostines/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Autophagie/physiologie , Kinase Janus-2/métabolisme , Kinase Janus-2/antagonistes et inhibiteurs , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Souris
18.
Cells ; 13(12)2024 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-38920670

RÉSUMÉ

Proinflammatory T-lymphocytes recruited into the brain and spinal cord mediate multiple sclerosis (MS) and currently there is no cure for MS. IFN-γ-producing Th1 cells induce ascending paralysis in the spinal cord while IL-17-producing Th17 cells mediate cerebellar ataxia. STAT1 and STAT3 are required for Th1 and Th17 development, respectively, and the simultaneous targeting of STAT1 and STAT3 pathways is therefore a potential therapeutic strategy for suppressing disease in the spinal cord and brain. However, the pharmacological targeting of STAT1 and STAT3 presents significant challenges because of their intracellular localization. We have developed a STAT-specific single-domain nanobody (SBT-100) derived from camelids that targets conserved residues in Src homolog 2 (SH2) domains of STAT1 and STAT3. This study investigated whether SBT-100 could suppress experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We show that SBT-100 ameliorates encephalomyelitis through suppressing the expansion of Th17 and Th1 cells in the brain and spinal cord. Adoptive transfer experiments revealed that lymphocytes from SBT-100-treated EAE mice have reduced capacity to induce EAE, indicating that the immunosuppressive effects derived from the direct suppression of encephalitogenic T-cells. The small size of SBT-100 makes this STAT-specific nanobody a promising immunotherapy for CNS autoimmune diseases, including multiple sclerosis.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Anticorps à domaine unique , Cellules Th17 , Animaux , Femelle , Souris , Camélidés du Nouveau Monde , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/thérapie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Souris de lignée C57BL , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/traitement médicamenteux , Anticorps à domaine unique/pharmacologie , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/usage thérapeutique , Moelle spinale/anatomopathologie , Moelle spinale/effets des médicaments et des substances chimiques , Moelle spinale/immunologie , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-3/métabolisme , 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
19.
J Interferon Cytokine Res ; 44(5): 208-220, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38691831

RÉSUMÉ

Intestinal damage and secondary bacterial translocation are caused by the inflammatory response induced by sepsis. Tongfu Lifei (TLF) decoction has a protective effect on sepsis-related gastrointestinal function injury. However, the relation between gut microbiota, immune barrier, and sepsis under the treatment of TLF have not been well clarified yet. Here, rats were subjected to cecal ligation and puncture (CLP) to create a sepsis model. Subsequently, the TLF decoction was given to CLP rats by gavage, fecal microbiota transplantation (FMT), and antibiotic were used as positive control. TLF suppressed the inflammatory response and improved the pathological changes in the intestines of CLP rats. Besides, TLF promoted the balance of the percentage of the Th17 and Treg cells. Intestinal barrier function was also improved by TLF through enhancing ZO-1, and Occludin and Claudin 1 expression, preventing the secondary translocation of other gut microbiota. TLF dramatically boosted the gut microbiota's alpha- and beta-diversity in CLP rats. Moreover, it increased the relative abundance of anti-inflammatory gut microbiota and changed the progress of the glucose metabolism. In short, TLF regulated the gut microbiota to balance the ratio of Th17/Treg cells, reducing the inflammation in serum and intestinal mucosal injury in rats.


Sujet(s)
Médicaments issus de plantes chinoises , Microbiome gastro-intestinal , Muqueuse intestinale , Sepsie , Lymphocytes T régulateurs , Cellules Th17 , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Sepsie/immunologie , Sepsie/traitement médicamenteux , Sepsie/complications , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Rats , Médicaments issus de plantes chinoises/pharmacologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/immunologie , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/microbiologie , Mâle , Rat Sprague-Dawley
20.
Balkan Med J ; 41(3): 174-185, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38700313

RÉSUMÉ

Background: Psoriasis is a chronic inflammatory skin disease that has no cure. While the specific cause of psoriasis is unknown, interactions between immune cells and inflammatory cytokines are believed to be important in its pathogenesis. Thymic stromal lymphopoietin (TSLP) is a cytokine produced by epithelial cells that profoundly affects dendritic cells (DCs) and is involved in allergy and inflammatory diseases. In some studies, its expression is higher in the skin of psoriasis patients, whereas it is increased in treated psoriasis patients when compared with untreated patients in others. Aims: To investigate the role of TSLP in the pathogenesis of psoriasis. Study Design: In vitro and in vivo study. Methods: To investigate the effect of TSLP on psoriasis in vivo, a mouse psoriasis model and shRNA targeting TSLP to reduce its expression were used. Mouse primary bone marrow dendritic cells (BMDCs) were cultured in vitro and used to investigate the signaling pathways activated by TSLP. Results: We found that reducing TSLP expression in psoriasis skin alleviated disease severity. TSLP activated the Janus kinase (JAK)/SYK pathway in psoriatic skin. In vitro studies with BMDCs demonstrated that TSLP increased DC maturation through the JAK/SYK pathway and activated DCs-secreted cytokines that stimulated CD4+ T cells to develop into T helper 17 (Th17) cells by activating STAT3 signaling. The JAK/SYK pathway inhibitor reduced the effect of TSLP on activating BMDCs and promoting Th17 differentiation by CD4+ T cells. Conclusion: These findings indicated that TSLP exerted its immune-modulating effect in psoriasis through the JAK/SYK pathway.


Sujet(s)
Cytokines , Cellules dendritiques , Psoriasis , Cellules Th17 , Lymphopoïétine stromale thymique , Animaux , Humains , Souris , Cytokines/métabolisme , Cytokines/effets des médicaments et des substances chimiques , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Modèles animaux de maladie humaine , Janus kinases , Transduction du signal/effets des médicaments et des substances chimiques , Syk kinase , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE