Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 271
Filter
1.
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
2.
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
3.
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
4.
Zhonghua Yu Fang Yi Xue Za Zhi ; 57(8): 1231-1237, 2023 Aug 06.
Article in Chinese | 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
5.
Sci Rep ; 13(1): 6523, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37085701

ABSTRACT

The ligand-receptor interaction is fundamental to many cellular processes in eukaryotic cells such as cell migration, proliferation, adhesion, signaling and so on. Cell migration is a central process in the development of organisms. Receptor induced chemo-tactic sensitivity plays an important role in cell migration. However, recently some receptors identified as decoy receptors, obstruct some mechanisms of certain regular receptors. DcR3 is one such important decoy receptor, generally found in glioma cell, RCC cell and many various malignant cells which obstruct some mechanism including apoptosis cell-signaling, cell inflammation, cell migration. The goal of our work is to mathematically formulate ligand-receptor interaction induced cell migration in the presence of decoy receptors. We develop here a novel mathematical model, consisting of four coupled partial differential equations which predict the movement of glioma cells due to the reaction-kinetic mechanism between regular receptors CD95, its ligand CD95L and decoy receptors DcR3 as obtained in experimental results. The aim is to measure the number of cells in the chamber's filter for different concentrations of ligand in presence of decoy receptors and compute the distance travelled by the cells inside filter due to cell migration. Using experimental results, we validate our model which suggests that the concentration of ligands plays an important role in cell migration.


Subject(s)
Glioma , Humans , Ligands , Signal Transduction , Cell Movement , Inflammation , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Apoptosis
6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1045860

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)
Humans , Interleukin-10 , Lipopolysaccharides/pharmacology , Macrophages , MicroRNAs/genetics , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Tumor Necrosis Factor-alpha
7.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1046183

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)
Humans , Interleukin-10 , Lipopolysaccharides/pharmacology , Macrophages , MicroRNAs/genetics , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Tumor Necrosis Factor-alpha
8.
Front Immunol ; 13: 879648, 2022.
Article in English | MEDLINE | ID: mdl-35720343

ABSTRACT

Background: Decoy receptor 3 (DcR3) belongs to the tumor necrosis factor (TNF) receptor superfamily and neutralizes TNF ligands, including FasL and TRAIL, to prevent T activation during T-cell priming. However, the cellular mechanisms underlying acute cell-mediated rejection (ACMR) remain unknown. Methods: We generated DcR3 transgenic (Tg) mice and mice with high DcR3 expression (HDE) to study both in vivo and in vitro. FasR RNA knockdown in immortalized CD4+CD8+ T-cells was used to survey the role of DcR3 on FasR/Fas-associated protein with death domain (FADD)/caspase 8 pathway and its cross-link to TNF receptor-associated factor 1 (TNFR1)-associated death domain protein (TRADD) in suppressing TNFR1. TNF/TRADD knockout mice were used to show the importance of TNF adaptor protein. Results: DcR3.Fc suppressed C57BL/6 female T-cell activation and transformation into CD4+CD69+, CD4+CD44+, and CD4+CD25+Foxp3+ when compared with isotype IgG1 and its co-treatment with FasL/TRAIL after exposing to bone marrow-derived dendritic cells (BMDCs) that carried alloantigen with male H-Y and minor antigenic determinant. Interleukin-17 and interferon-γ productions by BMDC-activated T-cells were lowered after co-treating with DcR3.Fc. DcR3.Fc induced effector T-cells (Teffs) and was susceptible to FasR-mediated apoptosis through the FADD/TRADD/caspase 8 pathway. After exposing to DcR3.Fc, TRADD was silenced, likely turning down the inflammatory response. The systemic effects of DcR3 Tg mice and HDE phenotype induced by the promoter of cytomegalovirus not only attenuated ACMR severity but also ameliorated the high serum creatinine and blood urea nitrogen levels even with high T-cell exposure frequencies. Besides this, DcR3 has minor biological effects on both MHC-matched and MHC-mismatched models. Conclusions: High DcR3 doses protect renal tubular epithelial cells from acute T-cell attack during the T-cell priming stage via interfering with TNF ligand-mediated reverse signaling and possibly promoting Teff apoptosis through FasR upregulation. Our findings supported that the decoy receptor is involved in T-cell modulation in kidney transplant rejection.


Subject(s)
Receptors, Tumor Necrosis Factor, Member 6b , Receptors, Tumor Necrosis Factor, Type I , Animals , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism
9.
Exp Mol Med ; 54(4): 542-551, 2022 04.
Article in English | MEDLINE | ID: mdl-35478210

ABSTRACT

Decoy receptor 3 (DcR3) is a soluble receptor for Fas ligand, LIGHT and TL1A, but it also exerts effector functions. Previously, we found that DcR3 is upregulated in the serum and lesional skin of patients with psoriasis and is upregulated by EGFR activation in proliferating primary human epidermal keratinocytes. However, the functional role of intracellular DcR3 in keratinocyte differentiation is still incompletely defined. Herein, primary cultured human epidermal keratinocytes were differentiated by phorbol 12-myristate 13-acetate (PMA) treatment, calcium treatment and cell confluence, which are three standard in vitro differentiation models. We found that the constitutive expression of the DcR3 gene and protein was progressively suppressed during terminal differentiation of keratinocytes. These changes were correlated with downregulation of EGFR activation during keratinocyte differentiation. EGFR inhibition by gefitinib further decreased confluence-induced suppression of DcR3 mRNA expression, and, vice versa, knocking down DcR3 expression attenuated EGFR and EGFR ligand expression as well as EGFR activation. Under conditions without a change in cell growth, DcR3 silencing reduced the expression of involucrin and transglutaminase 1 but enhanced the induction of the terminal differentiation markers keratin 10 and loricrin. Of note, DcR3 interacted with PKCα and PKCδ and enhanced PKC activity. In keratinocytes with PKCα and PKCδ silencing, differentiation markers were differentially affected. In conclusion, DcR3 expression in keratinocytes is regulated by EGFR and forms a positive feedback loop to orchestrate constitutive EGFR and PKC activity. During differentiation, DcR3 is downregulated and involved in modulating the pattern of terminal differentiation.


Subject(s)
Keratinocytes , Protein Kinase C-alpha , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Antigens, Differentiation/metabolism , Cell Differentiation , Cells, Cultured , Enzyme Activation , Epidermis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Keratinocytes/metabolism , Protein Kinase C/metabolism , Protein Kinase C-alpha/metabolism
10.
Immunol Res ; 69(6): 568-575, 2021 12.
Article in English | MEDLINE | ID: mdl-34383195

ABSTRACT

Bronchial asthma is a common chronic inflammatory disease with high prevalence and morbidity, particularly in school-aged children. Decoy receptor 3 (DcR3) is a soluble decoy receptor that belongs to the tumor necrosis factor receptor superfamily and has been reported to be elevated in several allergic and inflammatory diseases. This study was designed to determine the role of DcR3 in pediatric asthma. The serum DcR3 levels were analyzed in 85 subjects (60 pediatric patients with bronchial asthma and 25 age- and sex-matched healthy control children) using the enzyme-linked immunosorbent assay technique. Patients with asthma had higher serum DcR3 levels than healthy control subjects (p = 0.007). In the atopic group of patients with asthma, the serum DcR3 levels were inversely correlated with the asthma control test score (R = - 0.392, p = 0.039). Overall, DcR3 could be a promising biomarker of atopic asthma, specifically in pediatric patients.


Subject(s)
Asthma/blood , Receptors, Tumor Necrosis Factor, Member 6b/blood , Asthma/immunology , Biomarkers/blood , Child , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Receptors, Tumor Necrosis Factor, Member 6b/immunology
11.
J Burn Care Res ; 42(5): 925-933, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34213565

ABSTRACT

The clinical value of Decoy receptor 3 (DcR3) in severe burn is investigated. Ten patients with severe burns were monitored for DcR3, PCT, CRP, IL6, SOFA score, white blood cell (WBC), and platelet. The correlations were analyzed. DcR3 increased on day 1. The nonsurvivors had a steady high level of DcR3 while the survivors had a relatively low level of DcR3. The peak magnitude of DcR3 was high in five nonsurvivors and low in five survivors without overlap. Three patients had a continuously increasing DcR3 level and then died. In the other two nonsurvivors, DcR3 reached the peak and then decreased before death. DcR3 correlated well with PCT (ρ = 0.4469, P < .0001), less with CRP, platelet, IL6, SOFA score and WBC (ρ = 0.4369, 0.4078, 0.3995, 0.2631, 0.1504, respectively, all P < .001). To explore the mechanisms, the HaCaT or THP-1 cells were stimulated by the plasma of burn patients, 45°C, LPS or stimulators of TLRs or NOD2 (PGN, CL264, MDP, iE-DAP, Gardiquimod), and their DcR3 was increased, which could be reduced by GDC-0941 or BEZ235 (inhibitors of PI3K and mTOR). The levels of DcR3 appeared to be a useful biomarker for monitoring the clinical severity and a predictor of mortality of severe burns.


Subject(s)
Burns/blood , Receptors, Tumor Necrosis Factor, Member 6b/blood , Severity of Illness Index , Biomarkers/blood , Burns/metabolism , Humans , Platelet Count
12.
Front Immunol ; 12: 638676, 2021.
Article in English | MEDLINE | ID: mdl-33746978

ABSTRACT

Gout is a common inflammatory arthritis caused by the deposition of monosodium urate (MSU) crystals in the joints. This activates the macrophages into a proinflammatory state by inducing NLRP3-dependent interleukin-1ß (IL-1ß) secretion, resulting in neutrophil recruitment. Soluble decoy receptor 3 (DcR3) is an immune modulator and can exert biological functions via decoy and non-decoy actions. Previously, we showed that DcR3 suppresses lipopolysaccharides (LPS)- and virus-induced inflammatory responses in the macrophages and promotes the macrophages into the M2 phenotype. In this study, we clarified the actions of DcR3 and its non-decoy action motif heparin sulfate proteoglycan (HSPG) binding domain (HBD) in the MSU crystal-induced NLRP3 inflammasome activation in the macrophages and in mice. In bone marrow-derived macrophages, THP-1 and U937 cells, we found that the MSU crystal-induced secretion of IL-1ß and activation of NLRP3 were suppressed by both DcR3.Fc and HBD.Fc. The suppression of the MSU-induced NLRP3 inflammasome activation is accompanied by the inhibition of lysosomal rupture, mitochondrial production of the reactive oxygen species (ROS), expression of cathepsins, and activity of cathepsin B, without affecting the crystal uptake and the expression of NLRP3 or pro-IL-1ß. In the air pouch mice model of gout, MSU induced less amounts of IL-1ß and chemokines secretion, an increased M2/M1 macrophage ratio, and a reduction of neutrophil recruitment in DcR3-transgenic mice, which expresses DcR3 in myeloid cells. Similarly, the mice intravenously treated with DcR3.Fc or HBD.Fc displayed less inflammation response. These findings indicate that HBD of DcR3 can reduce MSU crystal-induced NLRP3 inflammasome activation via modulation of mitochondrial and lysosomal functions. Therefore, we, for the first time, demonstrate a new therapeutic potential of DcR3 for the treatment of gout.


Subject(s)
Gout/immunology , Inflammasomes/metabolism , Lysosomes/metabolism , Macrophages/immunology , Neutrophils/immunology , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Animals , Disease Models, Animal , Humans , Interleukin-1beta/metabolism , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophil Infiltration , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/genetics , THP-1 Cells , Uric Acid/metabolism
13.
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
14.
BMC Cancer ; 20(1): 572, 2020 Jun 18.
Article in English | MEDLINE | ID: mdl-32552802

ABSTRACT

BACKGROUND: Despite recent advances in cancer immunotherapy, the efficacy of these therapies for the treatment of human prostate cancer patients is low due to the complex immune evasion mechanisms (IEMs) of prostate cancer and the lack of predictive biomarkers for patient responses. METHODS: To understand the IEMs in prostate cancer and apply such understanding to the design of personalized immunotherapies, we analyzed the RNA-seq data for prostate adenocarcinoma from The Cancer Genome Atlas (TCGA) using a combination of biclustering, differential expression analysis, immune cell typing, and machine learning methods. RESULTS: The integrative analysis identified eight clusters with different IEM combinations and predictive biomarkers for each immune evasion cluster. Prostate tumors employ different combinations of IEMs. The majority of prostate cancer patients were identified with immunological ignorance (89.8%), upregulated cytotoxic T lymphocyte-associated protein 4 (CTLA4) (58.8%), and upregulated decoy receptor 3 (DcR3) (51.6%). Among patients with immunologic ignorance, 41.4% displayed upregulated DcR3 expression, 43.26% had upregulated CTLA4, and 11.4% had a combination of all three mechanisms. Since upregulated programmed cell death 1 (PD-1) and/or CTLA4 often co-occur with other IEMs, these results provide a plausible explanation for the failure of immune checkpoint inhibitor monotherapy for prostate cancer. CONCLUSION: These findings indicate that human prostate cancer specimens are mostly immunologically cold tumors that do not respond well to mono-immunotherapy. With such identified biomarkers, more precise treatment strategies can be developed to improve therapeutic efficacy through a greater understanding of a patient's immune evasion mechanisms.


Subject(s)
Biomarkers, Tumor/genetics , Immune Evasion/genetics , Immunotherapy/methods , Precision Medicine/methods , Prostatic Neoplasms/therapy , Biomarkers, Tumor/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Datasets as Topic , Gene Expression Regulation, Neoplastic/immunology , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , RNA-Seq , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Receptors, Tumor Necrosis Factor, Member 6b/immunology
15.
J Neuroimmunol ; 335: 577020, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31445379

ABSTRACT

TL1A/DR3/DcR3 pathway is an important mediator of inflammatory responses and contributes to the pathogenesis of several chronic inflammatory diseases. Therefore, we analysed PBMC gene expression of these molecules in 30 relapsing-remitting multiple sclerosis (RRMS) patients, 8 secondary progressive MS (SPMS), 9 primary progressive MS (PPMS), 11 clinically isolated syndrome (CIS) patients, and 16 healthy controls (HCs), to evaluate their biomarker potential in MS. The results showed significant decrease in TL1A expression in RRMS compared to other study groups. TL1A as a marker of inflammation, we found its higher expression among treatment näive RRMS patients as compared to HCs and among patients who were treated with DMTs. Moreover, TL1A expression was found to be associated with the clinical and MRI findings of MS patients suggesting its possible involvement in the establishment or preservation of immune system homeostasis or in the regulation of inflammatory activity. Taken together, these findings suggest the TL1A should be evaluated further for its potential as a candidate biomarker of inflammatory activity and the marker of therapeutic response to immunomodulatory treatments in MS.


Subject(s)
Multiple Sclerosis/immunology , Receptors, Tumor Necrosis Factor, Member 25/biosynthesis , Receptors, Tumor Necrosis Factor, Member 6b/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Adult , Biomarkers/analysis , Female , Follow-Up Studies , Humans , Male , Middle Aged , Receptors, Tumor Necrosis Factor, Member 25/analysis , Receptors, Tumor Necrosis Factor, Member 6b/analysis , Transcriptome , Tumor Necrosis Factor Ligand Superfamily Member 15/analysis
16.
J Chin Med Assoc ; 82(9): 680-684, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31356560

ABSTRACT

Decoy receptor 3 (DcR3), also known as tumor necrosis factor receptor superfamily member 6b (TNFRSF6B), was recently identified as a novel biomarker for predicting progression of kidney diseases with potential immune modulation. The purpose of this review is to discuss the current evidence related to DcR3 in kidney diseases and to compare the differences between human and animal studies both in vivo and in vitro. High serum DcR3 predicts the occurrence of peritonitis in patients receiving chronic peritoneal dialysis and is positively correlated with inflammatory markers such as interleukin-6, high-sensitivity C-reactive protein, and adhesion molecules in patients on maintenance hemodialysis (HD). Higher serum DcR3 levels not only independently predict cardiovascular and all-cause mortality in HD patients but also identify older adults on HD at risk of protein-energy wasting in combination with a low geriatric nutritional risk index. Recently, renal tubular epithelial cells (RTECs) expressing DcR3 have also been used to predict progression of chronic kidney disease. Expression of DcR3 was correlated with a 2-fold increase in serum creatinine or failure of kidney allograft. DcR3 could protect renal myofibroblasts against Fas-induced apoptosis and subsequently lead to renal fibrosis. Locally expressed DcR3 in the RTECs may suppress the FasL-Fas-mediated apoptosis of T cells, resulting in an accumulation of allo-reactive T cells. In addition to traditional biological functions, recombinant DcR3.Fc and cytomegalovirus promoter-driven human DcR3 plasmid are able to modulate the activation and differentiation of dendritic cells and macrophages via "non-decoy" action. Both progressive IgA nephropathy and autoimmune crescentic glomerulonephritis in mice can be suppressed after hydrodynamics-based gene delivery of DcR3 plasmid. DcR3-mediated effects in vitro could be surveyed via over-expressing DcR3 or addition of recombinant DcR3.Fc, and CD68-driven DcR3 transgenic mice are suitable for investigating systemic effect in vivo. Inhibition of DcR3 expression in human may be a promising approach for pathomechanism.


Subject(s)
Kidney Diseases/complications , Receptors, Tumor Necrosis Factor, Member 6b/physiology , Animals , Biomarkers/blood , Disease Progression , Humans , Immunomodulation , Kidney Diseases/immunology , Kidney Diseases/mortality , Prognosis , Receptors, Tumor Necrosis Factor, Member 6b/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 6b/blood , Receptors, Tumor Necrosis Factor, Member 6b/genetics
17.
Biomed Res Int ; 2019: 9145736, 2019.
Article in English | MEDLINE | ID: mdl-31317042

ABSTRACT

AIMS: To explore the expression level and clinical significance of decoy receptor 3 (DcR3) in patients with acute-on-chronic liver failure (ACLF). METHODS: Serum DcR3 levels were measured by enzyme-linked immunosorbent assay (ELISA) in 76 patients with ACLF and 41 non-ACLF patients with chronic liver disease. Blood routine and liver functions were accessed for their correlations with DcR3. RESULTS: Serum DcR3 in ACLF patients was significantly higher than that in non-ACLF patients. It was positively correlated with neutrophilic granulocyte, aspartate aminotransferase, prothrombin time, and international standardized ratio, but negatively correlated with platelet and serum albumin. At the early stage, the level of DcR3 was not significantly different between the survival and nonsurvival group of ACLF. However, at the late stage, DcR3 increased in nonsurvival and gradually decreased in survivals. The baseline DcR3 could not sufficiently predict the outcome of ACLF, while the change of DcR3 within the first week displayed a better predictive value than model for end-stage liver disease (MELD) score. CONCLUSIONS: DcR3 was highly expressed in patients with ACLF and correlated with several clinical indices. Dynamic change of DcR3 might predict the prognosis of ACLF.


Subject(s)
Acute-On-Chronic Liver Failure/blood , Prognosis , Receptors, Tumor Necrosis Factor, Member 6b/blood , Acute-On-Chronic Liver Failure/genetics , Acute-On-Chronic Liver Failure/pathology , Blood Platelets/metabolism , Blood Platelets/pathology , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/genetics , Humans , Liver/metabolism , Liver/pathology , Male , Middle Aged , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Serum Albumin/genetics
18.
Cytokine ; 123: 154744, 2019 11.
Article in English | MEDLINE | ID: mdl-31255916

ABSTRACT

BACKGROUND: Sickle cell anemia (SCA), a disorder with an important inflammatory component, where vasoocclusion is major contributor to the disease pathophysiology. Pro-inflammatory cytokines play an important regulatory role in the process of inflammation. We investigated the expression TL1A/DR3/DcR3 cytokine signaling pathway in peripheral blood mononuclear cells (PBMC) and their corresponding plasma levels in SCA subjects who presented with acute painful episodes. MATERIALS AND METHODS: PBMC were isolated from the blood of SCA subjects and normal healthy controls. RNA isolated from PBMC was used for real time gene expression of TL1A/DR3/DcR3. Gene expression was compared in subgroups within SCA subjects with co-inherited fetal hemoglobin (HbF) or alpha-globin gene deletions. Plasma prepared from blood was used for determination of TL1A/DR3/DcR3 proteins by ELISA assays. RESULTS: In the PBMC of SCA subjects, expression of TL1A and DcR3 is elevated, while DR3 expression is lowered in comparison to normal control PBMC. In SCA subjects with HbF > 10%, TL1A/DcR3 expression is lower, while HbF < 10% is associated with increased TL1A/DcR3 expression. Moreover, subjects with HbF > 10% appear to have significantly fewer pain episodes in comparison to those with HbF < 10%. Deletion of alpha-globin genes appears to have no significant effect on TL1A/DR3/DcR3 expression. Circulating levels of TL1A, DR3 and DcR3 in plasma were significantly elevated in SCA subjects. CONCLUSIONS: Elevated TL1A and DcR3 expression in PBMC of SCA subjects during painful vasoocclusive crisis, suggest an altered TL1A expression may contribute to the pathophysiology of vasoocclusive crisis in SCA. HbF > 10% appears to moderate TL1A elevation, while HbF < 10% exacerbates TL1A/DcR3 responses. Furthermore, subjects with HbF > 10% have significantly lower pain episodes reported as compared to subjects with HbF < 10%.


Subject(s)
Anemia, Sickle Cell/blood , Gene Expression Regulation , Leukocytes, Mononuclear/metabolism , Receptors, Tumor Necrosis Factor, Member 25/blood , Receptors, Tumor Necrosis Factor, Member 6b/blood , Tumor Necrosis Factor Ligand Superfamily Member 15/blood , Adolescent , Adult , Anemia, Sickle Cell/pathology , Female , Humans , Leukocytes, Mononuclear/pathology , Male
19.
Egypt J Immunol ; 26(1): 43-54, 2019 Jan.
Article in English | MEDLINE | ID: mdl-31332995

ABSTRACT

Different cytokines play roles in the pathogenesis and tissue damage of Rheumatoid Arthritis (RA) including, Tumor necrosis factor superfamily (TNFSF) and their receptors particularly TNF-like ligand 1A (TL1A), and its decoy receptor DcR3. This study included 150 subjects, of them 50 patients having Rheumatoid Arthritis (RA), 50 patients with Osteoarthritis (OA), and 50 normal controls. Clinical examination was done and data was collected from patient's sheets, routine laboratory investigations included, rheumatoid factor (RF) antibody, anti-cyclic citrullinated peptide (anti-CCP) antibody, erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP). Disease activity score 28 was calculated and used to measure the activity of RA. Serum and synovial fluid (SF) TL1A and DcR3 levels were measured by (ELISA), while IL-17 was measured in supernatant fluid of PBMC culture after stimulation with recombinant human (rh) TL1A. Results showed significantly higher levels of TL1A and its decoy receptor DcR3 in RA patients than the other two groups. It was also found that TL1A is significantly related to the disease activity and enhances IL-17 production after stimulation of PBMC. These results can guide scientists to the future substitutions in the way of treatment of various inflammatory and autoimmune diseases.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Autoantibodies/immunology , Receptors, Tumor Necrosis Factor, Member 6b/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Arthritis, Rheumatoid/immunology , Case-Control Studies , Cells, Cultured , Humans , Interleukin-17/immunology , Leukocytes, Mononuclear
20.
Cell Signal ; 62: 109346, 2019 10.
Article in English | MEDLINE | ID: mdl-31229617

ABSTRACT

Chronic hepatitis B (CHB) is associated with the development of hepatocellular carcinoma (HCC). Decoy receptor 3 (DcR3) is a tumor necrosis factor receptor that promotes tumor cell survival by inhibiting apoptosis and interfering with immune surveillance. Previous studies showed that DcR3 was overexpressed in HCC cells and that short hairpin RNA (shDcR3) sensitizes TRAIL-resistant HCC cells. However, the expression of DcR3 during hepatitis B virus (HBV) infection has not been investigated. Here, we demonstrated that DcR3 was overexpressed in CHB patients and that DcR3 upregulation was positively correlated with the HBV DNA load and liver injury (determined by histological activity index, serum alanine aminotransferase level, and aspartate aminotransferase level). We found that hepatitis B virus X protein (HBx) upregulated DcR3 expression in a dose-dependent manner, but this increase was blocked by NF-κB inhibitors. HBx also induced the activation of NF-κB, and the NF-κB subunits p65 and p50 upregulated DcR3 by directly binding to the DcR3 promoters. Inhibition of PI3K significantly downregulated DcR3 and inhibited the binding of NF-κB to the DcR3 promoters. Our results demonstrate that the HBx induced DcR3 expression via the PI3K/NF-κB pathway; this process may contribute to the development of HBV-mediated HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Receptors, Tumor Necrosis Factor, Member 6b/genetics , Trans-Activators/genetics , Transcription Factor RelA/genetics , Binding Sites/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , NF-kappa B p50 Subunit/genetics , Phosphatidylinositol 3-Kinases/genetics , Promoter Regions, Genetic/genetics , Protein Binding/genetics , RNA, Small Interfering/genetics , Signal Transduction/genetics , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL