Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Zhonghua Yu Fang Yi Xue Za Zhi ; 57(8): 1231-1237, 2023 Aug 06.
Article in Zh | MEDLINE | ID: mdl-37574317

ABSTRACT

Objective: To investigate the effect of microRNA (miR-148b) targeting decoy receptor 3 (DcR3) on macrophage polarization in sepsis. Methods: Experimental study. From December 2019 to December 2022, serum microRNA expression was detected in 3 patients with sepsis and 3 healthy controls in the clinical laboratory of Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine. Phorbol 12-myristate 13-acetate (PMA) was used to induce the differentiation of human acute monocytic leukemia cells THP-1 into macrophages, and then lipopolysaccharide (LPS) was added to stimulate the establishment of a sepsis cell model, and the expression changes of miR-148b and DcR3 were detected by RT-PCR and Western blot. Overexpression of DcR3 was used to detect the expression levels of TNF-α, CD163 and IL-10 in macrophages stimulated by LPS (100 ng/ml). Overexpression of miR-148b was used to observe the changes of molecular markers of macrophage polarization. The targeting regulation effect of miR-148b on DcR3 was determined by dual-luciferase reporter assay. t test was used to analyze whether there were statistical differences among the groups. Results: The expression of miR-148b was down-regulated (P<0.05) and the expression of DcR3 was up-regulated (P<0.01) in THP-1 macrophages stimulated by LPS. Overexpression of DcR3 inhibited the expression of TNF-α (P<0.05) and promoted the expression of CD163 (P<0.01) and IL-10 (P<0.01). When miR-148b mimics was added, the opposite effect was observed. The dual-luciferase reporter assay confirmed that miR-148b targets and binds to DcR3, inhibiting its transcription and expression. The results of flow cytometry showed that DcR3 could reverse the promoting effect of miR-148b on the CD86/CD163 ratio of macrophages (P<0.05). Conclusion: miR-148b inhibits the expression of DcR3, thereby inhibiting M2 polarization in LPS-stimulated macrophage cells.


Subject(s)
Lipopolysaccharides , MicroRNAs , Receptors, Tumor Necrosis Factor, Member 6b , Humans , Interleukin-10 , Lipopolysaccharides/pharmacology , Macrophages , MicroRNAs/genetics , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Tumor Necrosis Factor-alpha
2.
Mediators Inflamm ; 2020: 1237281, 2020.
Article in English | MEDLINE | ID: mdl-32587467

ABSTRACT

PURPOSE: Interleukin-1α (IL-1α) is a potent cytokine that plays a role in inflammatory arthritis and bone loss. Decoy receptor 3 (DCR3) is an immune modulator of monocytes and macrophages. The aim of this study was to investigate the mechanism of DCR3 in IL-1α-induced osteoclastogenesis. METHODS: We treated murine macrophages with DCR3 during receptor activator of nuclear factor kappa Β ligand- (RANKL-) plus IL-1α-induced osteoclastogenesis to monitor osteoclast formation by tartrate-resistant acid phosphatase (TRAP) staining. Osteoclast activity was assessed using a pit formation assay. The mechanisms of inhibition were studied by biochemical analyses, including RT-PCR, immunofluorescent staining, flow cytometry, an apoptosis assay, immunoblotting, and ELISA. RESULTS: DCR3 suppresses IL-1α-induced osteoclastogenesis in both primary murine bone marrow-derived macrophages (BMM) and RAW264.7 cells as it inhibits bone resorption. DCR3 induces RANKL-treated osteoclast precursor cells to express IL-1α, secretory IL-1ra (sIL-1ra), intracellular IL-1ra (icIL-1ra), reactive oxygen species (ROS), and Fas ligand and to activate IL-1α-induced interleukin-1 receptor-associated kinase 4 (IRAK4). The suppression of DCR3 during RANKL- or IL-1α-induced osteoclastogenesis may be due to the abundant secretion of IL-1ra, accumulation of ROS, and expression of Fas ligand in apoptotic osteoclast precursor cells. CONCLUSIONS: We concluded that there is an inhibitory effect of DCR3 on osteoclastogenesis via ROS accumulation and ROS-induced Fas ligand, IL-1α, and IL-1ra expression. Our results suggested that the upregulation of DCR3 in preosteoclasts might be a therapeutic target in inflammatory IL-1α-induced bone resorption.


Subject(s)
Fas Ligand Protein/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/metabolism , Osteoclasts/metabolism , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Death/genetics , Cell Death/physiology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Osteoclasts/cytology , RAW 264.7 Cells , Tartrate-Resistant Acid Phosphatase/metabolism
3.
J Pathol ; 244(2): 189-202, 2018 02.
Article in English | MEDLINE | ID: mdl-29057478

ABSTRACT

Endometriosis is a multifactorial inflammatory disease with persistent activation of the nuclear factor-κB (NF-κB) signalling pathway. Aberrant adhesion of endometrium is the essential step in the progression of endometriosis, but the molecular mechanism of ectopic growth of endometrium is still unclear. Decoy receptor 3 (DcR3)/TNFRSF6B, a pleiotropic immunomodulator regulated by oestrogen, is able to activate focal adhesion kinase to promote cell adhesion. We found that DcR3 is upregulated in human ectopic endometrial cells via activation of the Akt-NF-κB signalling pathway, and its expression level correlates positively with that of the adhesion molecules intercellular adhesion molecule 1 (ICAM-1) and homing cell adhesion molecule (HCAM; CD44). In a multivariate regression model, DcR3 expression level was the most significant parameter associated with endometriosis severity. Knockdown of DcR3 not only downregulated the expression of ICAM-1 and HCAM, but also reduced cell adhesion and migration. In vivo investigation further showed that DcR3 promoted the growth and spread of endometrium, whereas knockdown of DcR3 by lentivirus-delivered short hairpin RNA inhibited ectopic adhesion of endometrium and abrogated endometriosis progression. These observations are in support of DcR3 playing a critical role in the pathogenesis of endometriosis, and the inhibition of DcR3 expression being a promising approach for the treatment of endometriosis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Adhesion , Endometriosis/metabolism , Endometrium/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Animals , Case-Control Studies , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Disease Progression , Endometriosis/pathology , Endometriosis/physiopathology , Endometriosis/surgery , Endometrium/pathology , Endometrium/physiopathology , Endometrium/surgery , Female , Heterografts , Humans , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Signal Transduction
4.
BMC Biochem ; 19(1): 13, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30587127

ABSTRACT

BACKGROUND: Sepsis is a severe condition characterised by the body's systemic inflammatory response to infection. The specific sepsis-related biomarkers should be used in clinical diagnosis, therapeutic response monitoring, rational use of antibiotics, and prognosis (risk stratification), etc. RESULTS: In this study, we investigated the expression level of Decoy Receptor 3 (DcR3) and the mechanism of high expression in sepsis patients. Septic cell model experiments were performed by treating human umbilical vein endothelial cells (HUVECs) and Jurkat cells with lipopolysaccharide (LPS), lipoteichoic acid (LTA) and zymosan, respectively. SP600125, SB203580 and ammonium pyrrolidinedithiocarbamate (PDTC) were used to inhibit JNK1/2, p38MAPK and NF-κB signalling pathways in septic cell model, respectively. These results showed that DcR3 levels were higher in sepsis group than control. DcR3 mRNA and protein levels in HUVECs were increased following treatment with LPS, LTA and zymosan, and also increased in Jurkat cells treated by LPS, but not by LTA or zymosan. When HUVECs were treated with the NF-κB inhibitor PDTC, DcR3 expression was decreased compared with controls. However, SP600125 and SB203580 had no effect on DcR3 mRNA or protein levels. CONCLUSIONS: The results indicated that DcR3 secretion proceeded through the NF-κB signalling pathway in HUVECs.


Subject(s)
Lipopolysaccharides/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects , Anthracenes/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/pharmacology , Jurkat Cells , NF-kappa B/metabolism , Proline/analogs & derivatives , Proline/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Teichoic Acids/pharmacology , Thiocarbamates/pharmacology , Zymosan/pharmacology
5.
Mod Rheumatol ; 28(2): 287-292, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28696795

ABSTRACT

OBJECTIVES: Decoy receptor 3 (DcR3) competitively binds to Fas ligand, lymphotoxin-related inducible ligand that competes for glycoprotein D binding to herpes virus entry mediator on T cells (LIGHT) and TNF-like ligand 1A (TL1A), thereby preventing their effects. Using a microarray assay, we previously newly identified centrosomal protein 70 kDa (CEP70) as one of the genes whose expression in fibroblast-like synoviocytes from patients with rheumatoid arthritis (RA-FLS) is reduced by DcR3. Here, we investigated the significance of DcR3 regulation of CEP70 for RA-FLS. METHODS: Synovial samples were obtained from RA patients who had never been treated with biologics and from osteoarthritis (OA) patients. CEP70 mRNA expression was quantified using RT-qPCR analysis. CEP70 protein expression was assessed using immunohistochemical and western blot analyses. RESULTS: CEP70 was expressed predominantly in the superficial lining layer in RA synovial tissue. CEP70 expression was dose-dependently downregulated by DcR3-Fc in RA-FLS but was not downregulated in OA-FLS. TL1A antibody prevented the DcR3-Fc inhibitory effects on CEP70 expression in RA-FLS. CONCLUSIONS: These results indicated that DcR3 reduces CEP70 expression in RA-FLS by binding to membrane-bound TL1A and may suppress RA-FLS proliferation. The reduction in CEP70 expression by DcR3/TL1A signaling may control the hyperplasia of RA synovium.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cell Cycle Proteins/metabolism , Fibroblasts/metabolism , Microtubule-Associated Proteins/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Synovial Membrane/metabolism , Aged , Cell Cycle Proteins/genetics , Cells, Cultured , Down-Regulation , Female , Humans , Male , Microtubule-Associated Proteins/genetics , Middle Aged , Synovial Membrane/cytology
6.
J Biomed Sci ; 24(1): 39, 2017 Jun 19.
Article in English | MEDLINE | ID: mdl-28629361

ABSTRACT

Decoy receptor 3 (DcR3), also known as tumor necrosis factor receptor (TNFR) superfamily member 6b (TNFRSF6B), is a soluble decoy receptor which can neutralize the biological functions of three members of tumor necrosis factor superfamily (TNFSF): Fas ligand (FasL), LIGHT, and TL1A. In addition to 'decoy' function, recombinant DcR3.Fc is able to modulate the activation and differentiation of dendritic cells (DCs) and macrophages via 'non-decoy' action. DcR3-treated DCs skew T cell differentiation into Th2 phenotype, while DcR3-treated macrophages behave M2 phenotype. DcR3 is upregulated in various cancer cells and several inflammatory tissues, and is regarded as a potential biomarker to predict inflammatory disease progression and cancer metastasis. However, whether DcR3 is a pathogenic factor or a suppressor to attenuate inflammatory reactions, has not been discussed comprehensively yet. Because mouse genome does not have DcR3, it is not feasible to investigate its physiological functions by gene-knockout approach. However, DcR3-mediated effects in vitro are determined via overexpressing DcR3 or addition of recombinant DcR3.Fc fusion protein. Moreover, CD68-driven DcR3 transgenic mice are used to investigate DcR3-mediated systemic effects in vivo. Upregulation of DcR3 during inflammatory reactions exerts negative-feedback to suppress inflammation, while tumor cells hijack DcR3 to prevent apoptosis and promote tumor growth and invasion. Thus, 'switch-on' of DcR3 expression may be feasible for the treatment of inflammatory diseases and enhance tissue repairing, while 'switch-off' of DcR3 expression can enhance tumor apoptosis and suppress tumor growth in vivo.


Subject(s)
Apoptosis , Gene Expression Regulation , Immunologic Factors/genetics , Inflammation/genetics , Neoplasms/genetics , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Animals , Animals, Genetically Modified/genetics , Humans , Immunologic Factors/metabolism , Inflammation/etiology , Mice/genetics , Neoplasms/etiology , Receptors, Tumor Necrosis Factor, Member 6b/metabolism
7.
J Pathol ; 240(1): 25-37, 2016 09.
Article in English | MEDLINE | ID: mdl-27218286

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an irreversible lethal lung disease with an unknown etiology. IPF patients' lung fibroblasts express inappropriately high Akt activity, protecting them in response to an apoptosis-inducing type I collagen matrix. FasL, a ligand for Fas, is known to be increased in the lung tissues of patients with IPF, implicated with the progression of IPF. Expression of Decoy Receptor3 (DcR3), which binds to FasL, thereby subsequently suppressing the FasL-Fas-dependent apoptotic pathway, is frequently altered in various human disease. However, the role of DcR3 in IPF fibroblasts in regulating their viability has not been examined. We found that enhanced DcR3 expression exists in the majority of IPF fibroblasts on collagen matrices, resulting in the protection of IPF fibroblasts from FasL-induced apoptosis. Abnormally high Akt activity suppresses GSK-3ß function, thereby accumulating the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) in the nucleus, increasing DcR3 expression in IPF fibroblasts. This alteration protects IPF cells from FasL-induced apoptosis on collagen. However, the inhibition of Akt or NFATc1 decreases DcR3 mRNA and protein levels, which sensitizes IPF fibroblasts to FasL-mediated apoptosis. Furthermore, enhanced DcR3 and NFATc1 expression is mainly present in myofibroblasts in the fibroblastic foci of lung tissues derived from IPF patients. Our results showed that when IPF cells interact with collagen matrix, aberrantly activated Akt increases DcR3 expression via GSK-3ß-NFATc1 and protects IPF cells from the FasL-dependent apoptotic pathway. These findings suggest that the inhibition of DcR3 function may be an effective approach for sensitizing IPF fibroblasts in response to FasL, limiting the progression of lung fibrosis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Apoptosis/physiology , Fas Ligand Protein/metabolism , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Collagen/metabolism , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
8.
Cytokine ; 72(2): 185-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25647275

ABSTRACT

OBJECTIVE: To measure the levels of Tumor necrosis factor (TNF)-like ligand 1A (TL1A) and decoy receptor 3 (DcR3) in serum and synovial fluid (SF) of patients with rheumatoid arthritis (RA). To evaluate the effect of recombinant human (rh) TL1A on interleukin (IL)-17 production and IL-17mRNA expression. METHODS: The serum and SF levels of TL1A and DcR3, and the production of IL-17 by rhTL1A-treated PBMC were measured by enzyme-linked immunosorbent assay (ELISA). The expression of IL-17 mRNA by rhTL1A-treated PBMC was measured by real-time reverse transcriptase polymerase chain reaction (RT-PCR). We also tested the change of TL1A and DcR3 level following TNF-α blockade therapy. RESULTS: Serum TL1A and DcR3 levels were higher in RA patients. This increase was more significant in RF and anti-CCP positive patients. TL1A and DcR3 levels were higher in SF samples than in paired sera. TL1A and DcR3 decreased after anti-TNF treatment. rhTL1A increased the production of IL-17 protein and the expression of IL-17mRNA. CONCLUSION: TL1A and DcR3 may be of pathogenic and potentially of therapeutic importance in RA patients.


Subject(s)
Arthritis, Rheumatoid/immunology , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Synovial Fluid/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Adult , Aged , Anti-Inflammatory Agents, Non-Steroidal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infliximab/administration & dosage , Infliximab/therapeutic use , Interleukin-17/genetics , Interleukin-17/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Member 6b/blood , Tumor Necrosis Factor Ligand Superfamily Member 15/blood , Tumor Necrosis Factor-alpha/antagonists & inhibitors
9.
Respirology ; 20(1): 147-54, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25355638

ABSTRACT

BACKGROUND AND OBJECTIVE: Early diagnosis of tuberculous pleural effusion (TPE) remains difficult. While some inflammatory markers in pleural effusion (PE) are helpful in diagnosis, the roles of anti-inflammatory cytokines and effector molecules of cytotoxic T lymphocytes have not been investigated. METHODS: Lymphocyte-predominant exudative PE samples were assayed for inflammatory and anti-inflammatory cytokines and effector molecules of cytotoxic T lymphocytes. Logistic regression analysis was used to predict the probability of TPE and identify independently associated factors. Receiver operating characteristic (ROC) curve analysis was applied to determine the optimal cut-off value for the predicted probability. RESULTS: Of 95 patients enrolled, 35 had TPE, 46 had malignant PE and 14 had PE due to other aetiologies. Interferon-γ (IFN-γ), adenosine deaminase (ADA), decoy receptor (DcR) 3, monocyte chemo-attractant protein (MCP)-1, IFN-induced protein (IP)-10, granzyme A and perforin were higher in TPE than in PE of other aetiologies. By logistic regression analysis, IFN-γ ≥ 75 pg/mL, ADA ≥ 40 IU/mL, DcR3 ≥ 9.3 ng/mL and soluble tumour necrosis factor receptor 1 (TNF-sR1) ≥ 3.2 ng/mL were independent factors associated with TPE. The predicted probability based on the four predictors had an area under the ROC curve of 0.920, with 82.9% sensitivity and 86.7% specificity under the cut-off value of 0.303. In the TPE group, patients with positive PE/pleural culture for Mycobacterium tuberculosis had higher pleural IFN-γ, MCP-1, IP-10 and perforin than those with positive sputum but negative PE culture. CONCLUSIONS: While pleural interferon-γ and ADA are conventional markers for diagnosing TPE, simultaneous measurements of DcR3 and TNF-sR1 can improve the diagnostic efficacy.


Subject(s)
Mycobacterium tuberculosis , Pleural Effusion , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Receptors, Tumor Necrosis Factor, Type I/blood , T-Lymphocytes, Cytotoxic/pathology , Tuberculosis, Pleural , Adenosine Deaminase/metabolism , Aged , Biomarkers/metabolism , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/metabolism , Male , Middle Aged , Mycobacterium tuberculosis/isolation & purification , Mycobacterium tuberculosis/pathogenicity , Perforin/metabolism , Pleural Effusion/diagnosis , Pleural Effusion/etiology , Pleural Effusion/metabolism , Pleural Effusion/physiopathology , ROC Curve , Sensitivity and Specificity , Tuberculosis, Pleural/complications , Tuberculosis, Pleural/diagnosis , Tuberculosis, Pleural/metabolism , Tuberculosis, Pleural/physiopathology
10.
Biochim Biophys Acta ; 1832(10): 1538-48, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23707413

ABSTRACT

Decoy receptor 3 (DcR3) is a soluble receptor of Fas ligand (FasL), LIGHT (TNFSF14) and TNF-like molecule 1A (TL1A) and plays pleiotropic roles in many inflammatory and autoimmune disorders and malignant diseases. In cutaneous biology, DcR3 is expressed in primary human epidermal keratinocytes and is upregulated in skin lesions in psoriasis, which is characterized by chronic inflammation and angiogenesis. However, the regulatory mechanisms of DcR3 over-expression in skin lesions of psoriasis are unknown. Here, we demonstrate that DcR3 can be detected in both dermal blood vessels and epidermal layers of psoriatic skin lesions. Analysis of serum samples showed that DcR3 was elevated, but FasL was downregulated in psoriatic patients compared with normal individuals. Additional cell studies revealed a central role of epidermal growth factor receptor (EGFR) in controlling the basal expression of DcR3 in keratinocytes. Activation of EGFR by epidermal growth factor (EGF) and transforming growth factor (TGF)-α strikingly upregulated DcR3 production. TNF-αenhanced DcR3 expression in both keratinocytes and endothelial cells compared with various inflammatory cytokines involved in psoriasis. Additionally, TNF-α-enhanced DcR3 expression in keratinocytes was inhibited when EGFR was knocked down or EGFR inhibitor was used. The NF-κB pathway was critically involved in the molecular mechanisms underlying the action of EGFR and inflammatory cytokines. Collectively, the novel regulatory mechanisms of DcR3 expression in psoriasis, particularly in keratinocytes and endothelial cells, provides new insight into the pathogenesis of psoriasis and may also contribute to the understanding of other diseases that involve DcR3 overexpression.


Subject(s)
ErbB Receptors/physiology , Keratinocytes/metabolism , Psoriasis/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Up-Regulation/physiology , Cells, Cultured , Humans , NF-kappa B/metabolism , Signal Transduction
11.
Am J Nephrol ; 40(3): 191-9, 2014.
Article in English | MEDLINE | ID: mdl-25323840

ABSTRACT

BACKGROUND: Protein-energy wasting (PEW) is common and associated with poor outcome in hemodialysis patients. In hemodialysis patients, geriatric nutritional risk index (GNRI) and decoy receptor 3 (DcR3) have been shown as the nutritional and inflammatory markers, respectively. The present study aimed to assess the predictive ability of GNRI and DcR3 for PEW status and long-term outcomes in chronic hemodialysis patients. METHODS: A prospective cohort of 318 hemodialysis patients was conducted with a median follow-up of 54 months. Malnutrition-inflammation score (MIS) was used as the reference standard for the presence of PEW. Endpoints were cardiovascular and all-cause mortality. RESULTS: Baseline GNRI had a strong negative correlation with DcR3 and MIS score. For patients with age < or ≥60, high DcR3 and low GNRI were independent predictors for the presence of PEW at baseline. At the end of the study, 81 patients died (27 cardiovascular deaths). The adjusted hazard ratios (95% confidence intervals) of low GNRI and high DcR3 were 1.93 (1.1-4.8) and 2.53 (1.2-5.5) for cardiovascular mortality and 1.85 (1.1-3.2) and 2.37 (1.5-3.7) for all-cause mortality, respectively. While integrated into a model of conventional risk factors, GNRI together with DcR3 further significantly improved the predictability for overall mortality (c statistic, 0.823). CONCLUSIONS: Low GNRI and high DcR3 were the alternatives for identifying hemodialysis patients at risk of PEW and overall mortality. Further studies are needed to verify whether timely recognition of hemodialysis patients with a high malnutrition-inflammation risk could reduce their mortality by appropriate interventional strategies.


Subject(s)
Kidney Failure, Chronic/blood , Kidney Failure, Chronic/physiopathology , Nutritional Status , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Adult , Aged , Female , Humans , Inflammation , Kaplan-Meier Estimate , Kidney Failure, Chronic/mortality , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Renal Dialysis/mortality , Risk Assessment , Risk Factors , Severity of Illness Index
12.
Clin Exp Rheumatol ; 32(5): 715-23, 2014.
Article in English | MEDLINE | ID: mdl-25084482

ABSTRACT

OBJECTIVES: The decoy receptor 3 (DcR3) is a member of the tumour necrosis factor (TNF) receptor superfamily and may regulate inflammation. The aim of this study was to investigate the role of DcR3 in B cell functions and its correlation to disease activity in patients with rheumatoid arthritis (RA). METHODS: The concentrations of DcR3 and TNF-α were measured by ELISA. B cell proliferation was assessed by quantification of 3H-thymidine uptake. Staphylococcus aureus Cowan (SAC) strain were used to stimulate B cell proliferation and TNF-α production. RESULTS: Compared to the osteoarthritis (OA) patients, the RA group had higher synovial DcR3 levels (3273.6±1623.2 vs. 1594.8±1190.0 pg/ml, p=0.003), which were negatively correlated with the serum erythrocyte sedimentation rate and Disease Activity Score using 28 joint counts (DAS28) scores (r=-0.560, p=0.002; r=-0.579, p<0.001, respectively). Although the RA B cells have more active characteristics, B cell proliferation induced by SAC was successfully suppressed by recombinant DcR3.Fc fusion protein with an average inhibition of 44.8%. Moreover, DcR3.Fc fusion protein was found to suppress SAC-induced TNF-α production by B cells in 8 RA patients (average inhibition 47.0%). CONCLUSIONS: The results of our study indicated that the inhibition of B cell functions by DcR3 may partially explain the negative correlation between DcR3 level and disease activity in RA patients. Our findings imply that DcR3 may be used as a biomarker for disease activity and a potential therapeutic agent in the treatment of RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , B-Lymphocytes/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Aged , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , Biomarkers/metabolism , Case-Control Studies , Cell Proliferation , Cells, Cultured , Female , Humans , Lymphocyte Activation , Male , Middle Aged , Osteoarthritis, Knee/diagnosis , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/metabolism , Severity of Illness Index , Signal Transduction , Synovial Fluid/immunology , Synovial Fluid/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
13.
J Immunol ; 188(12): 5867-76, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22581854

ABSTRACT

Decoy receptor 3 (DcR3) is a soluble protein in the TNFR superfamily. Its known ligands include Fas ligand, homologous to lymphotoxin, showing inducible expression, and competing with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, TNF-like molecule 1A, and heparan sulfate proteoglycans. DcR3 has been reported to modulate the functions of T cells, dendritic cells, and macrophages; however, its role in regulating B cell activation is largely unknown. In this study, we found that the DcR3.Fc fusion protein bound to human and mouse B cells and suppressed the activation of B cells. DcR3.Fc attenuated Staphylococcus aureus, IgM-, Pam(3)CSK(4)-, and LPS-mediated B cell proliferation but did not affect cytokine-induced B cell growth. In the presence of these mitogens, DcR3.Fc did not induce B cell apoptosis, suggesting that DcR3 may inhibit the signal(s) important for B cell activation. Because the combination of Fas.Fc, LT-ßR.Fc (homologous to lymphotoxin, showing inducible expression, and competing with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes receptor), and DR3.Fc (TNF-like molecule 1A receptor) did not suppress B cell proliferation and because the biological effect of DcR3.Fc on B cells was not blocked by heparin, we hypothesize that a novel ligand(s) of DcR3 mediates its inhibitory activity on B cells. Moreover, we found that TLR2-stimulated NF-κB p65 activation and NF-κB-driven luciferase activity were attenuated by DcR3.Fc. The TLR2-induced cytokine production by B cells was consistently reduced by DcR3. These results imply that DcR3 may regulate B cell activation by suppressing the activation of NF-κB.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Activation/immunology , NF-kappa B/immunology , Receptors, Tumor Necrosis Factor, Member 6b/immunology , Toll-Like Receptor 2/immunology , Animals , Apoptosis/immunology , B-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Humans , Mice , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Toll-Like Receptor 2/metabolism
14.
ScientificWorldJournal ; 2014: 605236, 2014.
Article in English | MEDLINE | ID: mdl-24741354

ABSTRACT

BACKGROUND: Overexpression of decoy receptor 3 (DcR3) have been reported in various classes of malignancies. However, its expression and clinicopathological contribution in gliomas has not been fully elucidated. OBJECTIVE: To explore the expression and clinical significance of DcR3 protein in relation to tumor cell differentiation and proliferation in glioma cell lines and tissues. METHODS: One hundred and twenty-five samples of glioma patients and 18 cases of normal brain tissues were recruited. The expression of DcR3 protein was detected using immunohistochemistry. Tumor differentiation was assessed by histologic characters and the status of glial fibrillary acidic protein (GFAP). Tumor cell labeling indexes (LIs) of Ki-67 and PCNA were also obtained. The relationship between the DcR3 level and clinicopathological features was investigated, including tumor differentiation, LIs, and survival. Meanwhile, the expression of DcR3 protein was also measured in the supernatants of 8 glioma cell lines and glioma cells freshly prepared from 8 human glioblastoma specimens by using western blot. RESULTS: The level of DcR3 protein in gliomas was significantly higher than that in normal brain tissues (P < 0.01). DcR3 expression showed positive correlations with tumor pathological grade (r = 0.621, P < 0.01) and negative with GFAP expression (r = -0.489, P < 0.01). Furthermore, there were positive correlations between DcR3 expression and Ki-67, PCNA LIs (r = 0.529, P < 0.01; r = 0.556, P < 0.01). The survival in the DcR3 negative group was 50 ± 1.79 months, longer than that of the DcR3 positive group (48.36 ± 2.90), however, without significance (P = 0.149). Different levels of DcR3 could also be detected in the culturing supernatants of all the 8 glioma cell lines and glioma cells freshly obtained from 8 human glioblastoma specimens. CONCLUSIONS: The overexpression of DcR3 might play a crucial role in the tumorigenesis, differentiation, and proliferation of glioma.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Expression , Glioma/genetics , Glioma/pathology , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Adult , Aged , Brain Neoplasms/mortality , Case-Control Studies , Cell Proliferation , Female , Glioma/mortality , Humans , Ki-67 Antigen/metabolism , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Proliferating Cell Nuclear Antigen/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Young Adult
15.
Eur J Obstet Gynecol Reprod Biol ; 296: 185-193, 2024 May.
Article in English | MEDLINE | ID: mdl-38458034

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the expression and clinical significance of HIF-1α and DcR3 in endometriosis by analysing clinical case data. Tissue samples were collected for tissue chip analysis and staining, and human endometrial stromal cells were isolated and cultured for cell experiments. Additionally, experiments were conducted on collected peritoneal fluid to explore the association and role of HIF-1α and DcR3 in endometriosis. STUDY DESIGN: Patients who visited the Department of Obstetrics and Gynaecology at Central Hospital in Fengxian District, Shanghai, from January 2018 to December 2021 were recruited for this controlled study. Clinical data and tissue chip staining results were collected for multiple regression analysis on the clinical significance of HIF-1α and DcR3. Endometrial tissue, ovarian cysts, and pelvic fluid were collected, and human endometrial stromal cells were cultured. The impact of HIF-1α on DcR3 in different oxygen environments and its role in endometriosis were investigated through PCR, Western blotting, enzyme-linked immunosorbent assay, as well as adhesion and migration assays. RESULTS: In patients with endometriosis, the expression of DcR3 and HIF-1α was found to be upregulated and correlated in ectopic endometrium. The expression of DcR3 served as an indicator of the severity of endometriosis. Hypoxia induced the expression of DcR3, which was regulated by HIF-1α and promoted migration and adhesion. CONCLUSION: DcR3 can be used as a clinical indicator to assess the severity of endometriosis. The hypoxic environment in endometriosis enhances disease progression by regulating DcR3 through HIF-1α.


Subject(s)
Endometriosis , Hypoxia-Inducible Factor 1, alpha Subunit , Receptors, Tumor Necrosis Factor, Member 6b , Female , Humans , Endometriosis/metabolism , Endometrium/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Stromal Cells/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism
16.
Int J Biol Macromol ; 275(Pt 2): 133703, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38986982

ABSTRACT

Despite the high mortality rate associated with sepsis, no specific drugs are available. Decoy receptor 3 (DcR3) is now considered a valuable biomarker and therapeutic target for managing inflammatory conditions. DcR3-SUMO, an analog of DcR3, has a simple production process and high yield. However, its precise underlying mechanisms in sepsis remain unclear. This study investigated the protective effects of DcR3-SUMO on lipopolysaccharide (LPS)-induced inflammatory cells and septic mice. We evaluated the effects of DcR3 intervention and overexpression on intracellular inflammatory cytokine levels in vitro. DcR3-SUMO significantly reduced cytokine levels within inflammatory cells, and notably increased DcR3 protein and mRNA levels in LPS-induced septic mice, confirming its anti-inflammatory efficacy. Our in vitro and in vivo results demonstrated comparable anti-inflammatory effects between DcR3-SUMO and native DcR3. DcR3-SUMO protein administration in septic mice notably enhanced tissue morphology, decreased sepsis scores, and elevated survival rates. Furthermore, DcR3-SUMO treatment effectively lowered inflammatory cytokine levels in the serum, liver, and lung tissues, and mitigated the extent of tissue damage. AlphaFold3 structural predictions indicated that DcR3-SUMO, similar to DcR3, effectively interacts with the three pro-apoptotic ligands, namely TL1A, LIGHT, and FasL. Collectively, DcR3-SUMO and DcR3 exhibit comparable anti-inflammatory effects, making DcR3-SUMO a promising therapeutic agent for sepsis.


Subject(s)
Cytokines , Lipopolysaccharides , Receptors, Tumor Necrosis Factor, Member 6b , Sepsis , Animals , Sepsis/metabolism , Sepsis/drug therapy , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Mice , Cytokines/metabolism , Inflammation/metabolism , Male , Humans , Recombinant Fusion Proteins/pharmacology , Anti-Inflammatory Agents/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Mice, Inbred C57BL
17.
mBio ; 15(6): e0052124, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38700314

ABSTRACT

Despite its high mortality, specific and effective drugs for sepsis are lacking. Decoy receptor 3 (DcR3) is a potential biomarker for the progression of inflammatory diseases. The recombinant human DcR3-Fc chimera protein (DcR3.Fc) suppresses inflammatory responses in mice with sepsis, which is critical for improving survival. The Fc region can exert detrimental effects on the patient, and endogenous peptides are highly conducive to clinical application. However, the mechanisms underlying the effects of DcR3 on sepsis are unknown. Herein, we aimed to demonstrate that DcR3 may be beneficial in treating sepsis and investigated its mechanism of action. Recombinant DcR3 was obtained in vitro. Postoperative DcR3 treatment was performed in mouse models of lipopolysaccharide- and cecal ligation and puncture (CLP)-induced sepsis, and their underlying molecular mechanisms were explored. DcR3 inhibited sustained excessive inflammation in vitro, increased the survival rate, reduced the proinflammatory cytokine levels, changed the circulating immune cell composition, regulated the gut microbiota, and induced short-chain fatty acid synthesis in vivo. Thus, DcR3 protects against CLP-induced sepsis by inhibiting the inflammatory response and apoptosis. Our study provides valuable insights into the molecular mechanisms associated with the protective effects of DcR3 against sepsis, paving the way for future clinical studies. IMPORTANCE: Sepsis affects millions of hospitalized patients worldwide each year, but there are no sepsis-specific drugs, which makes sepsis therapies urgently needed. Suppression of excessive inflammatory responses is important for improving the survival of patients with sepsis. Our results demonstrate that DcR3 ameliorates sepsis in mice by attenuating systematic inflammation and modulating gut microbiota, and unveil the molecular mechanism underlying its anti-inflammatory effect.


Subject(s)
Cecum , Disease Models, Animal , Receptors, Tumor Necrosis Factor, Member 6b , Sepsis , Animals , Sepsis/drug therapy , Sepsis/microbiology , Mice , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Cecum/surgery , Humans , Ligation , Punctures , Male , Mice, Inbred C57BL , Gastrointestinal Microbiome , Cytokines/metabolism , Lipopolysaccharides , Apoptosis/drug effects , Inflammation
18.
Genes Immun ; 14(7): 447-52, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23965943

ABSTRACT

Genome-wide association studies have implicated common variation at the 20q13 locus in inflammatory bowel disease, particularly for the pediatric Crohn's form. This locus harbors tumor necrosis factor receptor superfamily (TNFRSF6B), encoding a secreted protein, decoy receptor 3 (DcR3), which binds to and neutralizes pro-inflammatory cytokines of the tumor necrosis factor superfamily. We sought to further the evidence of DcR3's role in pediatric IBD by identifying missense mutations with functional significance within TNFRSF6B. We sequenced the exons of the gene in 528 Caucasian pediatric IBD cases and 549 Caucasian healthy controls to establish the frequency of such events in each population. Sequencing revealed that our IBD cohort harbored a greater number of missense variants, yielding an odds ratio of 3.9 (P-value=0.005). Using functional assays, we established that the frequency of mutants defective in secretion from cultured cells was greater in the Crohn's category than in the controls, yielding an odds ratio of 7.1 (P-value=0.004). These results suggest that rare defective variants in TNFRSF6B have a role in the pathogenesis of some cases of IBD and that interventions targeting this group of tumor necrosis factor-family members may benefit patients with IBD.


Subject(s)
Colitis, Ulcerative/genetics , Crohn Disease/genetics , Mutation, Missense , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Adolescent , Black or African American , Case-Control Studies , Child , Child, Preschool , Colitis, Ulcerative/ethnology , Crohn Disease/ethnology , Exocytosis , Exons , Female , Gene Frequency , HEK293 Cells , Humans , Male , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Sequence Analysis, DNA , White People
19.
Mol Cancer ; 12(1): 120, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24107265

ABSTRACT

BACKGROUND: Overexpression of Decoy Receptor 3 (DcR3), a soluble member of the tumor necrosis factor receptor superfamily, is a common event in several types of cancer. In renal cell carcinoma (RCC), DcR3 overexpression is associated with lymph node and distant metastasis as well as a poor prognosis. However, the functional role and regulation of DcR3 expression in RCC is so far unknown. METHODS: Modulation of DcR3 expression by siRNA and ectopic gene expression, respectively, was performed in ACHN and 769-P RCC cell lines. Functional effects of a modulated DcR3 expression were analyzed with regard to migration, invasion, adhesion, clonogenicity, and proliferation. Furthermore, quantitative RT-PCR and immunoblot analyses were performed to evaluate the expression of downstream mediators of DcR3. In further experiments, luciferase assays, quantitative RT-PCR and immunoblot analyses were applied to study the regulation of DcR3 expression in RCC. Additionally, an ex vivo tissue slice culture technique combined with immunohistochemistry was used to study the regulation of DcR3 expression in human RCC specimens. RESULTS: Here, we show that DcR3 promotes adhesion, migration and invasiveness of RCC cells. The DcR3-dependent increase in cellular invasiveness is accompanied with an up-regulation of integrin alpha 4, matrixmetalloproteinase 7 and urokinase plasminogen activator (uPA). Further, we identified a signaling pathway regulating DcR3 expression in RCC. Using in vitro experiments as well as an ex vivo RCC tissue slice culture model, we demonstrate that expression of DcR3 is regulated in a PI3K/AKT-dependent manner involving the transcription factor nuclear factor of activated T-cells (NFAT). CONCLUSIONS: Taken together, our results identify DcR3 as a key driver of tumor cell dissemination and suggest DcR3 as a promising target for rational therapy of RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Movement , Kidney Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/physiology , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Carcinoma, Renal Cell/pathology , Cell Adhesion , Cell Line, Tumor , Chromones/pharmacology , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Kidney Neoplasms/pathology , Morpholines/pharmacology , NFATC Transcription Factors/metabolism , Neoplasm Invasiveness , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Signal Transduction , Transcription, Genetic
20.
Ann Hematol ; 92(10): 1359-67, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23652586

ABSTRACT

UNLABELLED: The soluble decoy receptor 3 (DcR3) is a member of the tumor necrosis factor receptor superfamily whose overexpression has been observed in several human malignancies. Survivin is one of the inhibitors of apoptosis proteins that are thought to play an important role in the pathogenesis of malignancies. We aimed to evaluate the expression of DcR3 in relation to survivin in B cell non-Hodgkin`s lymphoma (NHL) and then we focused on patients with diffuse large B cell non-Hodgkin's lymphoma (DLBCL) (50 cases) and correlated DcR3 expression with survivin expression and other prognostic parameters. Fifteen subjects with reactive lymphoid hyperplasia were included as controls. The expression of DcR3 and survivin were analyzed by immunohistochemistry on formalin-fixed paraffin-embedded lymph node sections from 80 cases of B cell NHL and 15 controls. Bone marrow biopsy sections of patients were also immunostained with the previous markers. RESULTS: DcR3 expression was found in 32.5% of B cell NHL patients versus 6.7% of controls (p <0.001) and was associated with the aggressive/highly aggressive subtypes. DcR3 was strongly expressed in 30% of DLBCL patients, where it was associated with survivin expression, high international prognostic index (IPI), the presence of extra nodal disease, ECOG performance status >1, reduced remission rates and shorter event-free survival. The expression of survivin was 40% in B cell NHL patients versus 13.3% in the control group (p <0.001). The expression of survivin in aggressive/highly aggressive B cell NHL was significantly higher than that in indolent B cell NHL. Survivin expression has been detected in 44% of the DLBCL patients and was associated with their clinical stage and shorter event-free survival (p = 0.026). Bone marrow biopsy sections from DLBCL patients showed significant DcR3 and survivin over expressions in sections with infiltration by lymphoma cells than sections with no infiltration. CONCLUSION: DcR3 expression was associated with other prognostic factors including survivin, reduced remission rates, and shorter event-free survival. Survivin is closely related to aggressive/highly aggressive subtypes of B cell NHL and is associated with shorter event-free survival. Both DcR3 and survivin expressions on bone marrow sections can be of help in diagnosing bone marrow infiltration.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, Non-Hodgkin/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Adolescent , Adult , Aged , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Survivin , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL