Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Dokl Biochem Biophys ; 513(Suppl 1): S67-S74, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38379077

ABSTRACT

In the current study the effects of metformin and cyanidin on the immune system and intestinal flora in rats with type-2 diabetes was investigated. The findings showed that metformin or cyanidin treatment considerably reduced the rise in body weight and glucose levels induced by type-2 diabetes. The type-2 diabetic rats' glucose tolerance was significantly increased by cyanidin administration comparable to that of metformin. Cyanidin administration resulted in a significant reduction in serum cholesterol and low-density lipoprotein (LDL) levels in rats with type-2 diabetes. Treatment with cyanidin significantly increased the ratio of high-density lipoprotein to low-density lipoprotein in type-2 diabetes rats. Cyanidin administration significantly raised the ratio of Firmicutes to Bacteroidetes in the fecal samples of type-2 diabetic rats compared to the model group. In comparison to the model group, it also significantly raised the levels of Lactobacillus intestinalis, Lactobacillus gasseri, and Lactobacillus reuteri in the type-2 diabetes rats. In type-2 diabetes rat fecal samples, the abundance of Christensenellaceae significantly increased while Enterobacteriaceae and Proteobacteria were found to decrease upon cyanidin administration. Furthermore, cyanidin administration to the rats with type-2 diabetes significantly improved the glucose homeostasis. In conclusion, the study demonstrates that cyanidin enhances glucose homeostasis in rats with type-2 diabetes, potentially through controlling intestinal flora. Thus, cyanidin may be looked into more as a possible therapeutic agent for type 2 diabetes.


Subject(s)
Anthocyanins , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Metformin , Rats , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Metformin/pharmacology , Lipoproteins, LDL/therapeutic use , Immunity , Glucose/therapeutic use , Blood Glucose , Hypoglycemic Agents/pharmacology
2.
Front Psychol ; 13: 928231, 2022.
Article in English | MEDLINE | ID: mdl-35783716

ABSTRACT

Based on event system theory, we explore the effect of the strength of channel conflict on cross-channel integration from the perspective of manufacturers, and then analyzes the mediating effect of channel fluency and channel stability. Taking manufactures who implement cross-channel integration as research samples, and basing the data collected from 229 respondents, the study uses multiple regression analysis and Bootstrap method to test the research hypotheses. The empirical research findings show that: the strength of channel conflict plays a negative role on channel fluency and channel stability; the strength of channel conflict has a double-edged sword effect on cross-channel integration: it can reduce cross-channel integration by destroying channel fluency, and at the same time can improve cross-channel integration by destroying and reducing channel stability.

3.
Endocr Connect ; 11(2)2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35107084

ABSTRACT

Hashimoto's thyroiditis (HT) is a very common organ-specific autoimmune disease characterized by lymphocyte infiltration and the destruction of thyroid follicular cells (TFCs), in which IFN-γ and chemokines play pivotal roles. Moreover, ß-catenin has been implicated in the regulation of T cell infiltration. However, whether ß-catenin is involved in Hashimoto's thyroiditis is unknown. Here, we examined ß-catenin expression in thyroid tissues and investigated its role in the pathogenesis of HT. The results showed that ß-catenin expression was markedly reduced in the thyroid tissues of HT patients; more importantly, IFN-γ treatment markedly reduced the expression of ß-catenin and was accompanied by the secretion of chemokines such as CCL5, CXCL16, GRO-ß, and GRO-γ in TFCs in vitro, which was attributed to GSK-3ß/ß-catenin signaling pathway activation. Collectively, the decreased expression of ß-catenin might contribute to IFNγ-induced chemokine secretion and lymphocyte infiltration in the development of HT.

4.
Article in English | MEDLINE | ID: mdl-31789139

ABSTRACT

BACKGROUND: Hashimoto's thyroiditis (HT) is characterized by lymphocytic infiltration of the thyroid parenchyma, which ultimately leads to tissue destruction and loss of function. Caveolin-1 (Cav-1) is an essential structural constituent of lipid rafts in the plasma membrane of cells and is reported to be significantly reduced in thyrocytes from HT patients. However, the mechanism of Cav-1 involvement in HT pathogenesis is still largely unclear. METHODS: Cav-1 expression in thyroid tissues from HT patients and euthyroid nodular goiter tissues was detected by immunohistochemistry staining. Cav-1 knockdown and overexpression were constructed by lentiviral transfection in the human thyroid follicular epithelial cell (TFC) line of Nthy-ori 3-1. The mRNA expression levels of chemokines in TFCs were determined by quantitative real-time PCR (qPCR). Cav-1 and peroxisome proliferator-activated receptor gamma (PPARγ) levels were analysed by qPCR and Western blot analysis. The migration ability of peripheral blood mononuclear cells (PBMCs) was detected by the Transwell assay. RESULTS: In this study, Cav-1 and PPARγ expression was reduced in the thyroid tissues from HT patients. In vitro experiments showed that the expressions of chemokine (C-C motif) ligand 5 (CCL5) and migration of PBMCs were markedly increased, while the level of PPARγ was significantly decreased after the lentivirus-mediated knockdown of Cav-1 in Nthy-ori 3-1 cells. Interestingly, pioglitazone, a PPARγ agonist, not only upregulated PPARγ and Cav-1 proteins significantly, but also effectively reversed the Cav-1-knockdown-induced upregulation of CCL5 in Nthy-ori 3-1 cells and reduced the infiltration of lymphocytes. CONCLUSION: The inhibition of Cav-1 upregulated the CCL5 expression and downregulated the PPARγ expression in TFC while pioglitazone, a PPARγ agonist, reversed the detrimental consequence. This outcome might be a potential target for the treatment of lymphocyte infiltration into the thyroid gland and HT development.


Subject(s)
Caveolin 1/biosynthesis , Chemokine CCL5/biosynthesis , Hashimoto Disease/metabolism , PPAR gamma/biosynthesis , Caveolin 1/genetics , Cell Line , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CCL5/genetics , Gene Expression , Hashimoto Disease/genetics , Hashimoto Disease/pathology , Humans , PPAR gamma/agonists , PPAR gamma/genetics , Pioglitazone/pharmacology , Thyroid Gland/drug effects , Thyroid Gland/metabolism
5.
Endocr J ; 65(9): 893-901, 2018 Sep 27.
Article in English | MEDLINE | ID: mdl-29877208

ABSTRACT

Hashimoto's thyroiditis (HT) is considered a T helper-type 1 (Th1) cytokine-dominant autoimmune thyroid disease. Caveolin-1 (Cav-1), a part of the thyroxisome multiprotein complex, is localized at the apical pole of thyrocytes and is indispensable for synthesis of thyroid hormones and modulation of oxidative stress in order to avoid cell damage and apoptosis. Reduced autophagy induces thyroid follicular cells (TFC) apoptosis by activating reactive oxygen species (ROS) in HT patients. Nevertheless, whether Cav-1 has roles in the regulation of autophagy remains largely unclear. In this study, we examined Th1 cytokines and Cav-1 expression in HT thyroid tissues, determined the effects of interleukin-1beta (IL-1ß) and interferon-gamma (IFN-γ) on Cav-1 and autophagy activity in TFC, and investigated the association between Cav-1 and autophagy activity in vitro. Our results indicate that higher levels of IL-1ß and IFN-γ and lower levels of Cav-1 were expressed in thyroid tissues of HT patients than in those of normal controls. Cav-1 mRNA and protein levels were significantly decreased in TFC exposed to IL-1ß and IFN-γ, accompanied by decreased expression of autophagy-related protein LC3B-II. Interestingly, small interfering RNA (siRNA)-mediated Cav-1 knockdown in TFC reduced LC3B-II protein expression. Taken together, these results suggest that lack of Cav-1 expression inhibited autophagy activity in TFC exposed to Th1 cytokines (IL-1ß and IFN-γ), which might be a novel pathogenetic mechanism of HT.


Subject(s)
Autophagy/genetics , Caveolin 1/physiology , Hashimoto Disease/genetics , Thyroid Epithelial Cells/physiology , Autophagy/drug effects , Case-Control Studies , Caveolin 1/genetics , Cells, Cultured , Gene Knockdown Techniques , Hashimoto Disease/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Th1 Cells/metabolism , Th1 Cells/physiology , Thyroid Epithelial Cells/drug effects , Thyroid Epithelial Cells/metabolism
6.
Endocr Connect ; 7(6): 850-858, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29764904

ABSTRACT

Myeloid-related protein 14 (MRP14) is responsible for inflammatory reactions. However, the correlation between MRP14 and Hashimoto's thyroiditis (HT) is still not clear. In this study, we examined the status of MRP14 in thyroid tissues and sera of HT patients and explored the mechanism of IL-1ß-mediated regulation of MRP14 expression, as well as the effects of MRP14 on pro-inflammatory chemokine secretion in thyroid follicular cells (TFCs), to elucidate the role of MRP14 in HT development. Our results showed dramatically increased MRP14 expression in thyroid tissues and sera from HT patients. In addition, IL-1ß significantly promoted the expression of MRP14 in TFCs, which was mediated by activation of the MAPK/NF-κB signalling pathway. More importantly, IL-1ß induced the secretion of the chemokines GRO-2, CXCL9 and CCL22, which was dependent on the regulation of MRP14 in TFCs. Therefore, these findings suggested that under pro-inflammatory conditions, TFCs secreted chemokines with the help of MRP14 regulation, which might suggest a potential pathological mechanism of lymphocyte infiltration into the thyroid gland in HT.

7.
Nat Commun ; 9(1): 1136, 2018 03 19.
Article in English | MEDLINE | ID: mdl-29555915

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized by uncontrolled secretion of autoantibodies by plasma cells. Although the functional importance of plasma cells and autoantibodies in SLE has been well established, the underlying molecular mechanisms of controlling autoantibody production remain poorly understood. Here we show that Peli1 has a B cell-intrinsic function to protect against lupus-like autoimmunity in mice. Peli1 deficiency in B cells induces autoantibody production via noncanonical NF-κB signaling. Mechanically, Peli1 functions as an E3 ligase to associate with NF-κB inducing kinase (NIK) and mediates NIK Lys48 ubiquitination and degradation. Overexpression of Peli1 inhibits noncanonical NF-κB activation and alleviates lupus-like disease. In humans, PELI1 levels negatively correlate with disease severity in SLE patients. Our findings establish Peli1 as a negative regulator of the noncanonical NF-κB pathway in the context of restraining the pathogenesis of lupus-like disease.


Subject(s)
Lupus Erythematosus, Systemic/etiology , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Animals , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Models, Animal , Female , HEK293 Cells , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination , NF-kappaB-Inducing Kinase
8.
Front Immunol ; 9: 96, 2018.
Article in English | MEDLINE | ID: mdl-29434604

ABSTRACT

Hashimoto's thyroiditis (HT) represents the most common organ-specific autoimmune disease. Inflammatory factors and reactive oxygen species (ROS) play detrimental roles during the pathogenesis of HT. In this study, we found that thyroid follicular cells (TFCs) from HT patients expressed an elevated level of interleukin-23 (IL-23), which contributed to autophagy suppression and ROS accumulation. Additionally, IL-23-induced autophagy suppression and ROS accumulation in human TFCs was attributed to AKT/mTOR/NF-κB signaling pathway activation. Inhibition of either IL-23 by a specific neutralization antibody, or mTOR by rapamycin, or NF-κB by IKK-16, significantly reversed the autophagy suppression and ROS accumulation. These results demonstrate a key role for IL-23 in HT pathogenesis and provide a potential therapeutic strategy against IL-23 or its signaling pathway in HT.


Subject(s)
Autophagy , Hashimoto Disease/metabolism , Interleukin-23/metabolism , Reactive Oxygen Species/metabolism , Adult , Biomarkers , Cell Line , Cytokines/metabolism , Female , Gene Expression , Hashimoto Disease/genetics , Hashimoto Disease/immunology , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Thyroid Gland/immunology , Thyroid Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...