Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Immunol ; 211(12): 1756-1761, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37888952

ABSTRACT

CXCR5 is a hallmark of T follicular helper (Tfh) cells. The mechanism of CXCR5 induction, however, is still incompletely understood. In this study, we report that in mice with the absence of transcription factor Bach2, the Th17-inducing cytokines IL-6 and TGF-ß together induced CXCR5 expression in vitro. Mechanistically, IL-6/STAT3 drove Cxcr5 promoter activity via the upstream site 1 regulatory element, whereas TGF-ß enhanced permissive histone modifications, and the STAT3 binding to the site 1 regulatory element was higher in the absence of Bach2. Subsequently, despite previous studies showing enhanced Th17 cell differentiation in the absence of Bach2 in vitro, we found that in vivo, the Bach2 deficiency led to an enhanced Tfh cell response at the expense of the Th17 cell response. These findings suggest that Bach2 helps integrate cytokine signals to arbitrate differentiation decisions between Tfh and Th17 lineages.


Subject(s)
Cytokines , Th17 Cells , Mice , Animals , Cytokines/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Interleukin-6/metabolism , Cell Differentiation , Transforming Growth Factor beta/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism
2.
Diabetes Metab Syndr Obes ; 16: 1567-1576, 2023.
Article in English | MEDLINE | ID: mdl-37283621

ABSTRACT

Purpose: The relationship between blood lipids and fibroblast growth factor (FGF) 21 in the postprandial period remains unclear. To investigate this, we observed the changes in blood lipid levels after an oral fat tolerance test (OFTT) and examined the short-term effects on FGF21. Patients and Methods: A total of 158 non-diabetic adult volunteers who underwent OFTT were randomly recruited from the Hebei General Hospital. Participants were stratified into three groups according to fasting and 4-h postprandial triglyceride levels: normal fat tolerance (NFT), impaired fat tolerance (IFT), and hypertriglyceridemia (HTG). Blood samples were collected at 2-h intervals for 6 h. Circulating total cholesterol levels, triglycerides, high-density lipoprotein-cholesterol, low-density lipoprotein-cholesterol, free fatty acids (FFA), and FGF21 were assessed. Results: Fasting FGF21 levels increased progressively in the NFT, IFT, and HTG groups and were strongly correlated with FFA levels (r = 0.531, P < 0.001). During the OFTT, the FFA and FGF21 levels decreased and then increased after reaching a nadir at 2 and 4 h, respectively. After adjusting for potential risk factors, the FFA incremental area under the curve (iAUC) was an independent influencing factor of FGF21 iAUC (P = 0.005). Conclusion: Fasting FGF21 levels showed a strong positive correlation with FFA. During OFTT, changes in FGF21 levels were closely associated with alterations in FFA exogenously changed by OFTT. Moreover, they were linearly related to each other. Therefore, the serum FGF21 level is positively correlated to the FFA level in the postprandial period.

3.
Diabetes Obes Metab ; 24(11): 2232-2240, 2022 11.
Article in English | MEDLINE | ID: mdl-35791646

ABSTRACT

AIM: To evaluate the efficacy and safety of DBPR108 (prusogliptin), a novel dipeptidyl peptidase-4 (DPP-4) inhibitor, as an add-on therapy in patients with type 2 diabetes (T2D) that is inadequately controlled with metformin. MATERIALS AND METHODS: In this 24-week, multi-centre, randomized, double-blind, placebo-controlled, superiority, phase III study, adult T2D patients with HbA1c levels ranging from 7.0% to 9.5% on stable metformin were enrolled and randomized (2:1) into the DBPR108 + metformin and placebo + metformin groups. The primary endpoint was the change from baseline in HbA1c at week 24 of DBPR108 versus placebo as an add-on therapy to metformin. RESULTS: At week 24, the least-square mean (standard error) change from baseline in HbA1c was significantly greater in the DBPR108 group (-0.70% [0.09%]) than in the placebo group (-0.07% [0.11%]) (P < .001), with a treatment difference of -0.63% (95% confidence interval: -0.87%, -0.39%) on the full analysis set. A higher proportion of patients achieved an HbA1c of 6.5% or less (19.7% vs. 8.5%) and an HbA1c of 7.0% or less (50.0% vs. 21.1%) at week 24 in the DBPR108 + metformin group. Furthermore, add-on DBPR108 produced greater reductions from baseline in fasting plasma glucose and 2-hour postprandial plasma glucose without causing weight gain. The overall frequency of adverse events was similar between the two groups. CONCLUSIONS: DBPR108 as add-on therapy to metformin offered a significant improvement in glycaemic control, was superior to metformin monotherapy (placebo) and was safe and well-tolerated in patients with T2D that is inadequately controlled with metformin.


Subject(s)
Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Metformin , Adult , Blood Glucose , Butanes , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/therapeutic use , Double-Blind Method , Drug Therapy, Combination , Glycated Hemoglobin , Humans , Hypoglycemic Agents/adverse effects , Metformin/adverse effects , Nitriles , Pyrrolidines , Treatment Outcome
4.
Article in English | MEDLINE | ID: mdl-35845595

ABSTRACT

Objective: The aim of the study is to evaluate the application value of three-dimensional speckle tracking imaging (3D-STI) and combined detection of thyroid autoantibodies and hormones in the diagnosis and treatment of Graves' disease. Methods: A total of 60 patients with Graves' disease enrolled in our hospital from February 2020 to February 2021 were included in the experimental group, and 60 healthy patients after a physical examination during the same period were selected as the control group. No intervention was performed on the control group, and the experimental group received conventional Graves' disease treatment. The levels of thyroid autoantibodies and hormones in the two groups before and after the treatment were measured, and the 3D-STI was performed to compare the 3D-STI strain parameters of the research objects. Results: A significantly higher level of thyroid autoantibodies in the experimental group than that in the control group before and after the treatment was found (P < 0.001), with a remarkable decline observed after the treatment (P < 0.001). The positive rate of thyroid autoantibodies in the experimental group before the treatment was significantly higher than that in the control group (P < 0.05). After the treatment, the positive rate of TRAb and TPOAb was higher than that of the control group (P < 0.05), and the positive rate of TPOAb was higher than before the treatment. The two groups showed no significant difference in the positive rate of TGAb (P > 0.05). Significant differences were observed in the thyroid hormone levels between the two groups and also between before and after the treatment (P < 0.001). The experimental group garnered significantly higher 3D-STI strain parameters than the control group before the treatment (P < 0.05); after the treatment, the hyperthyroidism of the patients was relieved with a decreased 3D-STI value, but it was still notably higher than the control group (P < 0.05). Remarkably higher positive rates of combined detection before and after the treatment in the experimental group than those in the control group were obtained (P < 0.05). Conclusion: The combined detection of 3D-STI and thyroid autoantibodies and hormones ensures a better detection rate of Graves' disease and monitors the treatment effect of patients in real time, which provides a basis for clinical diagnosis and treatment and merits clinical promotion and application.

5.
Kidney Blood Press Res ; 47(8): 514-522, 2022.
Article in English | MEDLINE | ID: mdl-35717941

ABSTRACT

BACKGROUND: Diabetic kidney disease is a major cause of global end-stage renal diseases. Ectopic lipid deposition in the renal tissues of diabetic kidney disease is one major factor leading to renal fibrosis and chronic kidney disease. Pterostilbene has been reported to display lipid-lowing activity and participate in many kidney diseases. However, the influence of pterostilbene on the ectopic lipid deposition is unclear. We intend to explore the influence of pterostilbene on the ectopic lipid deposition in the kidneys of mice induced by high fat. METHODS: A high-fat diet-induced diabetic mouse model was established to detect the alleviative effect of pterostilbene on the ectopic lipid deposition in the kidneys of diabetic mice. A biochemical analysis was performed to examine the levels of urine albumin, urine creatinine, serum creatinine, and blood urea nitrogen in mice after pterostilbene treatment. Histological analysis was conducted to detect the degree of renal injury and fibrosis. Oil red O staining and immunohistochemical staining were carried out to evaluate lipid droplets and the expression of adipose differentiation-related protein in renal tissues of the mice treated by pterostilbene. The protein levels were assessed by Western blotting. RESULTS: Pterostilbene inhibits the expression of the TGF-ß1 and p-smad3 and suppresses the protein levels of SREBP-1 and FAS, and it ultimately reduces the ectopic lipid deposition, alleviates the renal tubular damage and renal fibrosis in the kidneys of diabetic mice induced by high fat, and improves kidney function. CONCLUSION: Pterostilbene alleviates renal fibrosis and ectopic lipid deposition in the kidneys of diabetic mice induced by high-fat diet by inhibiting the TGF-ß1/smad3 signaling.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Diet, High-Fat/adverse effects , Fibrosis , Kidney/pathology , Lipids , Mice , Resveratrol/metabolism , Resveratrol/pharmacology , Stilbenes , Transforming Growth Factor beta1/metabolism
6.
Int J Clin Pract ; 2022: 9734738, 2022.
Article in English | MEDLINE | ID: mdl-35685602

ABSTRACT

Background and Aims: Previous studies on the effects of resveratrol on metabolic indicators reported contradictory findings, and these indicators have not been frequently studied in patients with type 2 diabetes. In this study, we aimed to examine the effects of resveratrol on metabolic indicators in a specific group of people with type 2 diabetes using the most recent literature. Methods: We used RevMan 5.4 and Stata 14.0 software to identify randomized controlled studies on the impact of resveratrol on metabolic indicators in patients with type 2 diabetes using relevant search terms and keywords such as "resveratrol" and "type 2 diabetes" in the China National Knowledge Infrastructure, PubMed, Cochrane, and Embase. Data were expressed as the weighted mean difference (WMD) and 95% confidence interval (CI). Results: This meta-analysis included 19 studies involving 1151 patients with type 2 diabetes, including 584 patients treated with resveratrol and 567 patients who received placebo. Compared with the control data, large doses of resveratrol (≥1000 mg) reduced fasting blood glucose levels (WMD: -18.76 mg/dL, 95% CI: -23.43, -14.09; P < 0.00001). Additionally, resveratrol reduced systolic blood pressure (WMD: -7.97 mmHg, 95% CI: -10.63, -5.31; P < 0.00001) and diastolic blood pressure (WMD: -3.55 mmHg, 95% CI: -5.18, -1.93; P < 0.00001) in patients with type 2 diabetes but did not improve waist circumference (WMD: 0.05 cm, 95% CI: -1.77, 1.88; P=0.95), triglyceride levels (WMD: -4.49 mg/dL, 95% CI: -24.23, 15.25; P=0.66), or high-density lipoprotein cholesterol levels (WMD: -1.05 mg/dL, 95% CI: -2.44, 0.33; P=0.14) in patients with type 2 diabetes. Conclusion: This systematic review and meta-analysis updated the most recent literature and provided new evidence, proving that resveratrol treatment can reduce systolic blood pressure and diastolic blood pressure. High-dose resveratrol can reduce fasting blood glucose in patients with type 2 diabetes, although it has no effect on waist circumference, triglyceride, and high-density lipoprotein cholesterol.


Subject(s)
Diabetes Mellitus, Type 2 , Blood Glucose , Cholesterol, HDL , Humans , Resveratrol/therapeutic use , Triglycerides
7.
Eur J Pharmacol ; 918: 174776, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35090936

ABSTRACT

Diabetic kidney disease (DKD) is a common microvascular complication of diabetes, and previous studies have shown that lipid deposits in the kidneys can lead to diabetic kidney damage. Resveratrol reduces circulating glucose and lipid concentrations, but it is unknown whether it can reduce renal lipid deposition and lipotoxic damage by regulating local lipid metabolism. We first showed that abnormal lipid metabolism is closely related to DKD in patients. There were excessive lipid deposits in the kidneys of patients with various stages of DKD, alongside abnormal expression of the junctional adhesion molecule-like (JAML)/sirtuin 1 (Sirt1) lipid synthesis pathway (P < 0.05). Next, we fed C57BL/6J mice a high-fat diet for 12 weeks, which caused an increase in body mass, blood glucose concentration, and blood lipid concentrations; and abnormalities in renal function (P < 0.05). Resveratrol administration ameliorated the defects in circulating lipid and glucose concentrations, renal dysfunction, the renal expression of components of the JAML/Sirt1 lipid synthesis pathway, and the expression of the adipose differentiation-related protein in the mice (P < 0.05). Histological staining also showed less lipid deposition and kidney damage. Thus, resveratrol regulates the JAML/Sirt1 lipid synthesis pathway, reduces lipid deposition in the kidney, and ameliorates diabetic kidney damage.


Subject(s)
Cell Adhesion Molecules/metabolism , Diabetic Nephropathies , Lipid Metabolism/drug effects , Lipids , Resveratrol/pharmacology , Sirtuin 1/metabolism , Animals , Antioxidants/pharmacology , Blood Glucose/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Kidney/drug effects , Kidney/pathology , Lipids/biosynthesis , Lipids/blood , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Tissue Distribution/drug effects
8.
Diabetes Obes Metab ; 23(5): 1111-1120, 2021 05.
Article in English | MEDLINE | ID: mdl-33417292

ABSTRACT

AIM: To evaluate henagliflozin, a novel sodium-glucose co-transporter-2 inhibitor, as monotherapy in patients with type 2 diabetes and inadequate glycaemic control with diet and exercise. MATERIALS AND METHODS: This multicentre trial included a 24-week, randomized, double-blind, placebo-controlled period, followed by a 28-week extension period. Four hundred and sixty-eight patients with an HbA1c of 7.0%-10.5% were randomly assigned (1:1:1) to receive once-daily placebo, or 5 or 10 mg henagliflozin. After 24 weeks, patients on placebo were switched to 5 or 10 mg henagliflozin, and patients on henagliflozin maintained the initial therapy. The primary endpoint was the change in HbA1c from baseline after 24 weeks. RESULTS: At Week 24, the placebo-adjusted least squares (LS) mean changes from baseline in HbA1c were -0.91% (95% CI: -1.11% to -0.72%; P < .001) and -0.94% (-1.13% to -0.75%; P < .001) with henagliflozin 5 and 10 mg, respectively; the placebo-adjusted LS mean changes were -1.3 (-1.8 to -0.9) and -1.5 (-2.0 to -1.1) kg in body weight, and -5.1 (-7.2 to -3.0) and -4.4 (-6.5 to -2.3) mmHg in systolic blood pressure (all P < .05). The trends of these improvements were sustained for an additional 28 weeks. Adverse events occurred in 81.0%, 78.9% and 78.9% of patients in the placebo, henagliflozin 5 and 10 mg groups, respectively. No diabetic ketoacidosis or major episodes of hypoglycaemia occurred. CONCLUSIONS: Henagliflozin 5 mg and 10 mg as monotherapy provided effective glycaemic control, reduced body weight and blood pressure, and was generally well tolerated.


Subject(s)
Diabetes Mellitus, Type 2 , Blood Glucose , Bridged Bicyclo Compounds, Heterocyclic , Diabetes Mellitus, Type 2/drug therapy , Diet , Double-Blind Method , Drug Therapy, Combination , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/therapeutic use , Treatment Outcome
9.
Sci Bull (Beijing) ; 66(15): 1571-1580, 2021 08 15.
Article in English | MEDLINE | ID: mdl-36654286

ABSTRACT

Chiglitazar (Carfloglitazar) is a novel non-thiazolidinedione (TZD) structured peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes in previous clinical studies. This randomized phase 3 trial aimed to compare the efficacy and safety of chiglitazar with placebo in patients with type 2 diabetes with insufficient glycemic control by strict diet and exercise alone. Eligible patients were randomly assigned to receive chiglitazar 32 mg (n = 167), chiglitazar 48 mg (n = 166), or placebo (n = 202) once daily. The primary endpoint was the change in glycosylated hemoglobin A1c (HbA1c) at week 24 with superiority of chiglitazar over placebo. The results showed that both chiglitazar 32 and 48 mg resulted in significant and clinically meaningful reductions in HbA1c, and placebo-adjusted estimated treatment differences at week 24 for chiglitazar 32 and 48 mg were -0.87% (95% confidential interval (CI): -1.10 to -0.65; P < 0.0001) and -1.05% (95% CI: -1.29 to -0.81; P < 0.0001), respectively. Secondary efficacy parameters including glycemic control, insulin sensitivity and triglyceride reduction were also significantly improved in the chiglitazar groups. The overall frequency of adverse events and study discontinuation attributable to adverse events were similar among the groups. Low incidences of mild edema and body weight gain were reported in the chiglitazar dose groups. The results from this phase 3 trial demonstrated that the PPAR pan-agonist chiglitazar possesses an overall good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/drug therapy , Peroxisome Proliferator-Activated Receptors/therapeutic use , Hypoglycemic Agents/adverse effects , Carbazoles
10.
Cell Rep ; 30(6): 1898-1909.e4, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049019

ABSTRACT

Understanding metabolic pathways that regulate Th17 development is important to broaden therapeutic options for Th17-mediated autoimmunity. Here, we report a pivotal role of mitochondrial oxidative phosphorylation (OXPHOS) for lineage specification toward pathogenic Th17 differentiation. Th17 cells rapidly increase mitochondrial respiration during development, and this is necessary for metabolic reprogramming following T cell activation. Surprisingly, specific inhibition of mitochondrial ATP synthase ablates Th17 pathogenicity in a mouse model of autoimmunity by preventing Th17 pathogenic signature gene expression. Notably, cells activated under OXPHOS-inhibited Th17 conditions preferentially express Foxp3, rather than Th17 genes, and become suppressive Treg cells. Mechanistically, OXPHOS promotes the Th17 pioneer transcription factor, BATF, and facilitates T cell receptor (TCR) and mTOR signaling. Correspondingly, overexpression of BATF rescues Th17 development when ATP synthase activity is restricted. Together, our data reveal a regulatory role of mitochondrial OXPHOS in dictating the fate decision between Th17 and Treg cells by supporting early molecular events necessary for Th17 commitment.


Subject(s)
Mitochondria/metabolism , Oxidative Phosphorylation , Receptors, Antigen, T-Cell/metabolism , Th17 Cells/immunology , Animals , Cell Differentiation , Mice , Signal Transduction
11.
J Immunol ; 202(10): 2991-2998, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30971440

ABSTRACT

T follicular helper (Tfh) cells are essential for germinal center B cell responses. The molecular mechanism underlying the initial Tfh cell differentiation, however, is still incompletely understood. In this study, we show that in vivo, despite enhanced non-Tfh cell effector functions, the deletion of transcription factor Bach2 results in preferential Tfh cell differentiation. Mechanistically, the deletion of Bach2 leads to the induction of CXCR5 expression even before the upregulation of Ascl2. Subsequently, we have identified a novel regulatory element in the murine CXCR5 locus that negatively regulates CXCR5 promoter activities in a Bach2-dependent manner. Bach2 deficiency eventually results in a collapsed CD4+ T cell response with severely impaired CD4+ T cell memory, including Tfh cell memory. Our results demonstrate that Bach2 critically regulates Tfh cell differentiation and CD4+ T cell memory.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Immunologic Memory , T-Lymphocytes, Helper-Inducer/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Gene Expression Regulation/immunology , Mice , Mice, Transgenic , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , T-Lymphocytes, Helper-Inducer/cytology
12.
J Immunol ; 200(2): 586-594, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29212910

ABSTRACT

T follicular helper (Tfh) cells play an essential role in the formation of germinal centers (GC) and generation of high-affinity Abs. The homing of activated CD4+ T cells into B cell follicles and the involvement of key costimulatory and coinhibitory molecules are critical in controlling both the initiation and the magnitude of GC responses. Meanwhile, studies have shown that a high number of single clone B cells leads to intraclonal competition, which inhibits the generation of high-affinity Abs. Our previous work has shown that transcription factor Foxp1 is a critical negative regulator of Tfh cell differentiation. In this study, we report that the deletion of Foxp1 leads to a high proportion of activated CD4+ T cells homing into B cell follicles with faster kinetics, resulting in earlier GC formation. In addition, we show that Foxp1-deficient Tfh cells restore the generation of high-affinity Abs when cotransferred with high numbers of single clone B cells. We find that Foxp1 regulates the expression levels of cytotoxic T lymphocyte-associated Ag-4 (CTLA-4) in activated CD4+ T cells and that Ctla4 is a direct Foxp1 target. Finally, we demonstrate that CTLA-4 expression on conventional CD4+ T cells plays a cell-intrinsic role in Tfh cell differentiation in vivo, and CTLA-4 blockade helps abolish the intraclonal competition of B cells in generating high-affinity Abs.


Subject(s)
CTLA-4 Antigen/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Forkhead Transcription Factors/metabolism , Germinal Center/immunology , Repressor Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/genetics , Immunomodulation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/cytology
13.
J Immunol ; 196(9): 3537-41, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27001958

ABSTRACT

Previously we have shown that transcription factor Foxp1 plays an essential role in maintaining naive T cell quiescence; in the absence of Foxp1, mature naive CD8(+) T cells proliferate in direct response to homeostatic cytokine IL-7. In this study, we report that the deletion of Foxp1 in naive CD8(+) T cells leads to enhanced activation of the PI3K/Akt/mammalian target of rapamycin signaling pathway and its downstream cell growth and metabolism targets in response to IL-7. We found that Foxp1 directly regulates PI3K interacting protein 1, a negative regulator of PI3K. Additionally, we found that deletion of Foxp1 in naive CD8(+) T cells results in increased expression levels of E2fs, the critical components for cell cycle progression and proliferation, in a manner that is not associated with increased phosphorylation of retinoblastoma protein. Taken together, our studies suggest that Foxp1 enforces naive CD8(+) T cell quiescence by simultaneously repressing key pathways in both cellular metabolism and cell cycle progression.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Cycle , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Interleukin-7/metabolism , Repressor Proteins/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle/physiology , Cell Proliferation , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Homeostasis , Interleukin-7/immunology , Interleukin-7/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Repressor Proteins/deficiency , Repressor Proteins/genetics , Retinoblastoma Protein/immunology , Retinoblastoma Protein/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
14.
J Biol Chem ; 289(48): 33311-9, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25315772

ABSTRACT

Human Galectin-3 (Gal-3), a ß-galactoside-binding protein expressed by tumor cells, has been reported to act as an immune regulator in antitumor T cells. However, its effect on natural killer (NK) cells is elusive. Using a recombinant human NK cell-activating receptor, NKp30 fusion protein (NKp30-Fc), we found that soluble NKp30-Fc could immunoprecipitate Galectin-3. The direct interaction between NKp30 and Galectin-3 was further confirmed using surface plasmon resonance experiments. Because Galectin-3 was mainly released from tumor cells in a soluble form in our study, the binding assay was performed to show that soluble Galectin-3 specifically bound to NK cells and NKp30 on the surface of the NK cells. Functionally, when soluble Galectin-3 was added to the NK-tumor cell coculture system, the NKp30-mediated, but not NKG2D-mediated, cytolysis and CD107a expression in the NK cells were inhibited, and these phenotypes could be restored by preincubation of soluble Galectin-3 with NKp30-Fc fusion protein or the addition of anti-Gal-3 antibody alone. Moreover, genetic down-regulation of Galectin-3 (shGal-3) resulted in tumor cells being more sensitive to NK cell lysis, and, reversely, Galectin-3-overexpressing HeLa cells (exGal-3) became less sensitive to NK cell killing. The results of these in vitro experiments were supported by studies in shGal-3-HeLa or exGal-3-HeLa xenograft non-obese diabetic/severe combined immunodeficiency mice after NK cell adoptive immunotherapy, indicating that Galectin-3 strongly antagonizes human NK cell attack against tumors in vivo. These findings indicate that Galectin-3 may function as an immune regulator to inhibit NK cell function against tumors, therefore providing a new therapeutic target for tumor treatment.


Subject(s)
Galectin 3/immunology , Immunity, Cellular , Natural Cytotoxicity Triggering Receptor 3/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Tumor Escape , Animals , Blood Proteins , Down-Regulation/genetics , Down-Regulation/immunology , Galectin 3/genetics , Galectins , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , HeLa Cells , Heterografts , Humans , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Mice , Mice, Inbred NOD , Mice, SCID , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Natural Cytotoxicity Triggering Receptor 3/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
15.
Nat Immunol ; 15(7): 667-75, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24859450

ABSTRACT

CD4(+) follicular helper T cells (T(FH) cells) are essential for germinal center (GC) responses and long-lived antibody responses. Here we report that naive CD4(+) T cells deficient in the transcription factor Foxp1 'preferentially' differentiated into T(FH) cells, which resulted in substantially enhanced GC and antibody responses. We found that Foxp1 used both constitutive Foxp1A and Foxp1D induced by stimulation of the T cell antigen receptor (TCR) to inhibit the generation of T(FH) cells. Mechanistically, Foxp1 directly and negatively regulated interleukin 21 (IL-21); Foxp1 also dampened expression of the costimulatory molecule ICOS and its downstream signaling at early stages of T cell activation, which rendered Foxp1-deficient CD4(+) T cells partially resistant to blockade of the ICOS ligand (ICOSL) during T(FH) cell development. Our findings demonstrate that Foxp1 is a critical negative regulator of T(FH) cell differentiation.


Subject(s)
Cell Differentiation , Forkhead Transcription Factors/physiology , Repressor Proteins/physiology , T-Lymphocytes, Helper-Inducer/cytology , Animals , CD4-Positive T-Lymphocytes/immunology , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukins/genetics , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/physiology
16.
Liver Int ; 34(7): e246-56, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24125589

ABSTRACT

BACKGROUND & AIMS: Liver disease, such as malignancy and hepatitis, often correlates with several genetic disorders. We aimed to construct a hepatocyte-specific vector that could manipulate multiple genes simultaneously. METHODS: We selected a highly efficient hepatocyte-specific α-foetoprotein (AFP) enhancer/albumin promoter (an RNA polymerase II promoter) to express our gene of interest and transcribe microRNA-based shRNAs (shRNAmir). Multiple shRNAmirs were assembled together in tandem to enhance the gene-silencing effect. By employing the AFP enhancer/albumin promoter and inserting an internal ribosome entry site (IRES), a hepatocyte-specific, multi-reporter vector that overexpressed both ß-galactosidase (LacZ) and DsRed2 while simultaneously knocking down both EGFP and luciferase expression was successfully constructed and functionally tested in vitro. RESULTS: The reporter genes in the multireporter vector were easily replaced by immune-related genes to construct the Multi-Vector, which overexpressed human interleukin 10 and silenced both CCL5 and CX3CL1 (FKN) simultaneously in vivo; visualization of DsRed2 coexpressed to monitor vector function in vivo confirmed that the Multi-Vector was successfully introduced into the host. Simultaneous manipulation of these multiple genes by the Multi-Vector synergistically inhibited acute liver injury induced by Poly I:C/D-GalN injection in mice. The multifunctional cassette was also packaged in and successfully delivered by an adenoviral vector. CONCLUSIONS: We successfully engineered a vector that can simultaneously regulate multiple genes from a single multigene-containing vector in a hepatocyte-specific manner, suggesting the possibility that this method could be extensively and practically utilized in liver gene therapy.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Gene Expression Regulation/genetics , Gene Knockdown Techniques/methods , Genes, Reporter/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Hepatocytes/metabolism , Adenoviridae , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Enhancer Elements, Genetic/genetics , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , MicroRNAs/genetics , Poly I-C , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , RNA, Small Interfering/genetics , alpha-Fetoproteins/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
17.
J Immunol ; 190(9): 4821-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23554312

ABSTRACT

Adenovirus or adenoviral vectors were reported to induce serious liver inflammation in an NK cell-dependent manner, which limits its clinical applicability for liver gene therapy. We tried to develop an efficient liver-directed therapeutic approach to control hepatic NK cell function via simultaneously manipulating multiple immune genes. Based on our previous study, we found that CCL5 knockdown synergistically enhanced the attenuating effect of silencing CX3CL1 (fractalkine [FKN]) in adenovirus-induced acute liver injury. In addition, the combined treatment of human IL-10 expression with FKN knockdown would further strengthen the protective effect of silencing FKN. We used a hepatocyte-specific promoter to construct a hepatocyte-specific multiple function vector, which could simultaneously overexpress human IL-10 and knock down CCL5 and FKN expression. This vector could attenuate adenovirus-induced acute hepatitis highly efficiently by reducing liver NK cell recruitment and serum IFN-γ and TNF-α. The multiple function vectors could be delivered by nonviral (hydrodynamic injection) and viral (adenovirus) approaches, and maintained long-term function (more than 1 month in mice). Our results suggest a possible strategy to ameliorate the acute liver injury induced by adenovirus by modulating multiple immune genes. The novel multifunction vector has an extensive and practical use for polygenic and complex liver diseases such as malignancies and hepatitis, which correlate with multiple gene disorders.


Subject(s)
Genetic Vectors/genetics , Genetic Vectors/immunology , Killer Cells, Natural/immunology , Liver Diseases/genetics , Liver Diseases/immunology , Liver/immunology , Adenoviridae/immunology , Adenoviridae Infections/genetics , Adenoviridae Infections/immunology , Animals , Cell Line, Tumor , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CX3CL1/genetics , Chemokine CX3CL1/immunology , Genetic Therapy/methods , Hepatocytes/immunology , Hepatocytes/virology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Liver/virology , Liver Diseases/virology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
18.
Cancer Sci ; 100(3): 529-36, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19154402

ABSTRACT

Toll-like receptor 3 and RIG-I like receptors (RLRs; MDA5, RIG-I) are involved in cell growth inhibition and apoptosis. However, the toll-like receptor 3-related apoptotic pathway is insensitive to direct polyinosinic-polycytidylic acid (dsRNA analog) stimulation in hepatoma cells. To determine whether the strategy of transferring polyinosinic-polycytidylic acid into cells (polyinosinic-polycytidylic acid-liposome) could induce apoptosis in hepatoma cells through cytoplasm receptors, we examined the responses of innate immune receptors RLRs and toll-like receptor 3 in response to different stimulation. We found that the apoptosis could exclusively be detected under polyinosinic-polycytidylic acid-liposome stimulation, which involved the activation of the caspase pathway. Besides, the expression of RIG-I, MDA5, IFNbeta and interferon-stimulated gene 15 was increased significantly at an early stage. Moreover, the growth inhibition of polyinosinic-polycytidylic acid-liposome was confirmed in a mouse model. Taken together, these results suggest polyinosinic-polycytidylic acid-liposome could be used as a potential apoptotic agent in hepatocellular carcinoma cells and imply a potential therapeutic strategy.


Subject(s)
Apoptosis/physiology , Carcinoma, Hepatocellular/metabolism , DEAD-box RNA Helicases/drug effects , Interferon Inducers/administration & dosage , Liver Neoplasms/metabolism , Poly I-C/administration & dosage , Animals , Caspases/drug effects , Cell Line, Tumor , DEAD Box Protein 58 , Flow Cytometry , Humans , Interferon-Induced Helicase, IFIH1 , Interferon-beta/drug effects , Liposomes , Male , Mice , Mice, Nude , RNA, Messenger/analysis , Receptors, Immunologic , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...