Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
PLoS One ; 18(8): e0289798, 2023.
Article in English | MEDLINE | ID: mdl-37552689

ABSTRACT

Liver transplantation is the most effective treatment option for patients with acute or chronic liver failure. However, the applicability and effectiveness of this modality are often limited by a shortage of donors, surgical complications, high medical costs, and the need for continuing immunosuppressive therapy. An alternative approach is liver cell transplantation. LIGHT (a member of the tumor necrosis factor superfamily) could be a promising candidate for promoting the differentiation of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) into hepatocyte-like cells. In this study, we investigated the effect of LIGHT on hBM-MSC differentiation into hepatocyte-like cells. Our previous results showed that LIGHT receptor lymphotoxin-ß receptor (LTßR) is constitutively expressed on the surface of hBM-MSCs. Upon treatment with recombinant human LIGHT (rhLIGHT), the phenotype of hBM-MSCs changed to round or polygonal cells. In addition, the cells exhibited high levels of hepatocyte-specific markers, including albumin, cytokeratin-18 (CK-18), CK-19, cytochrome P450 family 1 subfamily A member 1 (CYP1A1), CYP1A2, CYP3A4, SRY-box transcription factor 17 (SOX17), and forkhead box A2 (FOXA2). These results indicate that rhLIGHT enhances the differentiation of hBM-MSCs into functional hepatocyte-like cells. Furthermore, rhLIGHT-induced hepatocyte-like cells showed a higher ability to store glycogen and uptake indocyanine green compared with control cells, indicating functional progression. Additionally, treatment with rhLIGHT increased the number, viability, and proliferation of cells by inducing the S/G2/M phase and upregulating the expression of various cyclin and cyclin dependent kinase (CDK) proteins. We also found that the hepatogenic differentiation of hBM-MSCs induced by rhLIGHT was mediated by the activation of signal transducer and activator of transcription 3 (STAT3) and STAT5 pathways. Overall, our findings suggest that LIGHT plays an essential role in promoting the hepatogenic differentiation of hBM-MSCs. Hence, LIGHT may be a valuable factor for stem cell therapy.


Subject(s)
Bone Marrow , Mesenchymal Stem Cells , Humans , Bone Marrow Cells , Cell Differentiation , Hepatocytes/metabolism , Cyclin-Dependent Kinases/metabolism , Cells, Cultured , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
2.
Cell Death Dis ; 13(3): 238, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35292636

ABSTRACT

Hepatic stellate cells (HSC) and hydrogen sulfide (H2S) both play important roles in the development of hepatocellar carcinoma (HCC). Whereas, in the microenvironment of HCC, whether HSC participate in regulating the biological process of HCC cells by releasing H2S remains elusive. In vitro, Flow cytometry (FCM), CCK-8, RNA-sequencing, Western blotting, RT-qPCR, immunofluorescence and ChIP assays were carried out in the HCC cells to investigate the effect of H2S on biological functions and JNK/JunB-TNFSF14 signaling pathway. Specimens from HCC patients were analyzed by RT-qPCR and Western blotting assays for evaluating the expression of TNFSF14 and CSE. Statistical analysis was used to analyze the correlation between TNFSF14 expression and clinical data of HCC patients. Based on the FCM and CCK-8 results, we found the LX-2 cells were able to induce HCC cells apoptosis through releasing H2S. RNA-sequencing, RT-qPCR, and Western blotting results showed that TNFSF14 gene was upregulated in both LX-2 and NaHS group. NaHS treated in HCC cells led to JNK/JunB signaling pathway activating and greater binding of p-JunB to its responsive elements on TNFSF14 promoter. Impairment of TNFSF14 induction alleviated LX-2 and NaHS induced apoptosis of HepG2 and PLC/PRF/5 cells. Furthermore, TNFSF14 expression in HCC tissues was lower than the adjacent tissue. HCC patients with low expression of TNFSF14 had higher malignant degree and poor prognosis. In summary, demonstration of the involvement of HSC-derived H2S in JNK/JunB mediated expression of TNFSF14 gene strongly indicates H2S palys an important role in the regulation of HCC apoptosis.


Subject(s)
Carcinoma, Hepatocellular , Hydrogen Sulfide , Liver Neoplasms , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Hepatic Stellate Cells/metabolism , Humans , Hydrogen Sulfide/pharmacology , Liver Neoplasms/pathology , MAP Kinase Signaling System , RNA/metabolism , Sincalide/metabolism , Transcription Factors/metabolism , Tumor Microenvironment , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
3.
PLoS One ; 16(2): e0247368, 2021.
Article in English | MEDLINE | ID: mdl-33606781

ABSTRACT

Osteoporosis is a progressive systemic skeletal disease associated with decreased bone mineral density and deterioration of bone quality, and it affects millions of people worldwide. Currently, it is treated mainly using antiresorptive and osteoanabolic agents. However, these drugs have severe adverse effects. Cell replacement therapy using mesenchymal stem cells (MSCs) could serve as a treatment strategy for osteoporosis in the future. LIGHT (HVEM-L, TNFSF14, or CD258) is a member of the tumor necrosis factor superfamily. However, the effect of recombinant LIGHT (rhLIGHT) on osteogenesis in human bone marrow-derived MSCs (hBM-MSCs) is unknown. Therefore, we monitored the effects of LIGHT on osteogenesis of hBM-MSCs. Lymphotoxin-ß receptor (LTßR), which is a LIGHT receptor, was constitutively expressed on the surface of hBM-MSCs. After rhLIGHT treatment, calcium and phosphate deposition in hBM-MSCs, stained by Alizarin red and von Kossa, respectively, significantly increased. We performed quantitative real-time polymerase chain reaction to examine the expressions of osteoprogenitor markers (RUNX2/CBFA1 and collagen I alpha 1) and osteoblast markers (alkaline phosphatase, osterix/Sp7, and osteocalcin) and immunoblotting to assess the underlying biological mechanisms following rhLIGHT treatment. We found that rhLIGHT treatment enhanced von Kossa- and Alizarin red-positive hBM-MSCs and induced the expression of diverse differentiation markers of osteogenesis in a dose-dependent manner. WNT/ß-catenin pathway activation strongly mediated rhLIGHT-induced osteogenesis of hBM-MSCs, accelerating the differentiation of hBM-MSCs into osteocytes. In conclusion, the interaction between LIGHT and LTßR enhances osteogenesis of hBM-MSCs. Therefore, LIGHT might play an important role in stem cell therapy.


Subject(s)
Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genetic Markers , Humans , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Recombinant Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Wnt Signaling Pathway/drug effects
4.
Front Immunol ; 9: 576, 2018.
Article in English | MEDLINE | ID: mdl-29616048

ABSTRACT

The cytokine TNFSF14 [homologous to Lymphotoxin, exhibits Inducible expression and competes with HSV Glycoprotein D for binding to HVEM, a receptor expressed on T lymphocytes (LIGHT)] has been shown in mouse models to be important for development of lung tissue remodeling that is characteristic of asthma, idiopathic pulmonary fibrosis (IPF), and systemic sclerosis (SSc). However, its cellular targets are not fully delineated. In the present report, we show that LTßR and HVEM, the receptors for LIGHT, are constitutively expressed in primary human lung fibroblasts (HLFs). We asked whether LIGHT could promote inflammatory and remodeling-relevant activity in HLFs and how this was similar to, or distinct from, IL-13 or TGF-ß, two cytokines strongly implicated in the pathogenesis of asthma, IPF, and SSc. Accumulation of myofibroblasts expressing alpha smooth muscle actin is a feature of lung inflammatory diseases. LIGHT promoted cell cycle progression and proliferation of HLFs, but not alpha smooth muscle actin expression. In contrast, TGF-ß upregulated alpha smooth muscle actin but did not drive their proliferation. LIGHT also increased the gene or protein expression of a number of proinflammatory mediators, including ICAM-1 and VCAM-1, IL-6 and GM-CSF, the chemokines CCL5 and 20, and CXCL5, 11, and 12, and lung remodeling-associated proteinases MMP-9 and ADAM8. These were dependent on LTßR but not HVEM. LIGHT displayed overlapping and synergistic activities with IL-13 for a number of the activities, but LIGHT additionally enhanced the gene expression of several molecules, including the innate cytokines IL-33 and TSLP, which were not upregulated by IL-13. Our results highlight the varied and pleiotropic effects of LIGHT in HLFs. LIGHT might then be a therapeutic target for modulation of inflammation and remodeling associated with asthma and other similar diseases of the lung that involve fibroblasts.


Subject(s)
Fibroblasts/metabolism , Inflammation Mediators/metabolism , Interleukin-13/metabolism , Lung/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Biomarkers , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Fibroblasts/drug effects , Gene Expression , Humans , Inflammation Mediators/pharmacology , Interleukin-13/genetics , Lung/pathology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
5.
J Pathol ; 245(2): 209-221, 2018 06.
Article in English | MEDLINE | ID: mdl-29603739

ABSTRACT

High-grade brain cancer such as glioblastoma (GBM) remains an incurable disease. A common feature of GBM is the angiogenic vasculature, which can be targeted with selected peptides for payload delivery. We assessed the ability of micelle-tagged, vascular homing peptides RGR, CGKRK and NGR to specifically bind to blood vessels in syngeneic orthotopic GBM models. By using the peptide CGKRK to deliver the tumour necrosis factor (TNF) superfamily member LIGHT (also known as TNF superfamily member 14; TNFSF14) to angiogenic tumour vessels, we have generated a reagent that normalizes the brain cancer vasculature by inducing pericyte contractility and re-establishing endothelial barrier integrity. LIGHT-mediated vascular remodelling also activates endothelia and induces intratumoural high endothelial venules (HEVs), which are specialized blood vessels for lymphocyte infiltration. Combining CGKRK-LIGHT with anti-vascular endothelial growth factor and checkpoint blockade amplified HEV frequency and T-cell accumulation in GBM, which is often sparsely infiltrated by immune effector cells, and reduced tumour burden. Furthermore, CGKRK and RGR peptides strongly bound to blood vessels in freshly resected human GBM, demonstrating shared peptide-binding activities in mouse and human primary brain tumour vessels. Thus, peptide-mediated LIGHT targeting is a highly translatable approach in primary brain cancer to reduce vascular leakiness and enhance immunotherapy. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Brain Neoplasms/drug therapy , Cell-Penetrating Peptides/metabolism , Drug Carriers , Glioblastoma/drug therapy , Neovascularization, Pathologic , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Vascular Remodeling/drug effects , Venules/drug effects , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/metabolism , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line , Drug Compounding , Female , Glioblastoma/blood , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Micelles , Middle Aged , Molecular Targeted Therapy , Pericytes/drug effects , Pericytes/metabolism , Pericytes/pathology , Phenotype , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Venules/metabolism , Venules/pathology
6.
Rheumatology (Oxford) ; 56(2): 294-302, 2017 02.
Article in English | MEDLINE | ID: mdl-27864565

ABSTRACT

OBJECTIVE: Angiogenesis is crucial in RA disease progression. Lymphotoxin ß receptor (LTßR)-induced activation of the non-canonical nuclear factor-κB (NF-κB) pathway via NF-κB-inducing kinase (NIK) has been implicated in this process. Consequently, inhibition of this pathway may hold therapeutic potential in RA. We describe a novel three-dimensional (3D) model of synovial angiogenesis incorporating endothelial cells (ECs), RA fibroblast-like synoviocytes (RAFLSs) and RA synovial fluid (RASF) to further investigate the contributions of NF-κB in this process. METHODS: Spheroids consisting of RAFLSs and ECs were stimulated with RASF, the LTßR ligands LTß and LIGHT, or growth factor bFGF and VEGF, followed by quantification of EC sprouting using confocal microscopy and digital image analysis. Next, the effects of anginex, NIK-targeting siRNA (siNIK), LTßR-Ig fusion protein (baminercept) and a novel pharmacological NIK inhibitor were investigated. RESULTS: RASF significantly promoted sprout formation, which was blocked by the established angiogenesis inhibitor anginex (P < 0.05). LTß and LIGHT induced significant sprouting (P < 0.05), as did bFGF/VEGF (P < 0.01). siNIK pre-treatment of ECs led to reductions in LTßR-induced vessel formation (P < 0.05). LTßR-Ig not only blocked LTß- or LIGHT-induced sprouting, but also RASF-induced sprouting (P < 0.05). The NIK inhibitor blocked angiogenesis induced by LTß, LIGHT, growth factors (P < 0.05) and RASF (P < 0.01). CONCLUSION: We present a novel 3D model of synovial angiogenesis incorporating RAFLSs, ECs and RASF that mimics the in vivo situation. Using this system, we demonstrate that non-canonical NF-κB signalling promotes neovascularization and show that this model is useful for dissecting relative contributions of signalling pathways in specific cell types to angiogenic responses and for testing pharmacological inhibitors of angiogenesis.


Subject(s)
Endothelial Cells/drug effects , NF-kappa B/metabolism , Neovascularization, Pathologic/metabolism , Synoviocytes/drug effects , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblast Growth Factors/pharmacology , Humans , Lymphotoxin beta Receptor , Lymphotoxin-beta/pharmacology , Microscopy, Confocal , Neovascularization, Pathologic/pathology , Peptides/pharmacology , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering , Recombinant Fusion Proteins/pharmacology , Signal Transduction , Synovial Fluid , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synoviocytes/metabolism , Synoviocytes/pathology , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , NF-kappaB-Inducing Kinase
7.
PLoS One ; 11(11): e0166589, 2016.
Article in English | MEDLINE | ID: mdl-27835685

ABSTRACT

LIGHT (HVEM-L, TNFSF14, or CD258), an entity homologous to lymphotoxins, with inducible nature and the ability to compete with herpes simplex virus glycoprotein D for herpes virus entry mediator (HVEM)/tumor necrosis factor (TNF)-related 2, is a member of the TNF superfamily. It is expressed as a homotrimer on activated T cells and dendritic cells (DCs), and has three receptors: HVEM, LT-ß receptor (LTßR), and decoy receptor 3 (DcR3). So far, three receptors with distinct cellular expression patterns are known to interact with LIGHT. Follicular DCs and stromal cells bind LIGHT through LTßR. We monitored the effects of LIGHT on human bone marrow-derived mesenchymal stem cells (BM-MSCs). At first, we checked the negative and positive differentiation markers of BM-MSCs. And we confirmed the quality of MSCs by staining cells undergoing adipogenesis (Oil Red O staining), chondrogenesis (Alcian blue staining), and osteogenesis (Alizarin red staining). After rhLIGHT treatment, we monitored the count, viability, and proliferation of cells and cell cycle distribution. PDGF and TGFß production by rhLIGHT was examined by ELISA, and the underlying biological mechanisms were studied by immunoblotting by rhLIGHT treatment. LTßR was constitutively expressed on the surface of human BM-MSCs. Cell number and viability increased after rhLIGHT treatment. BM-MSC proliferation was induced by an increase in the S/G2/M phase. The expression of not only diverse cyclins such as cyclin B1, D1, D3, and E, but also CDK1 and CDK2, increased, while that of p27 decreased, after rhLIGHT treatment. RhLIGHT-induced PDGF and TGFß production mediated by STAT3 and Smad3 activation accelerated BM-MSC proliferation. Thus, LIGHT and LTßR interaction increases the survival and proliferation of human BM-MSCs, and therefore, LIGHT might play an important role in stem cell therapy.


Subject(s)
Bone Marrow Cells/drug effects , Cell Proliferation/drug effects , Lymphotoxin beta Receptor/genetics , Mesenchymal Stem Cells/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , CDC2 Protein Kinase , Cell Cycle , Cell Differentiation/drug effects , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , Gene Expression Regulation , Humans , Lymphotoxin beta Receptor/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microarray Analysis , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
8.
J Am Heart Assoc ; 5(7)2016 06 30.
Article in English | MEDLINE | ID: mdl-27364991

ABSTRACT

BACKGROUND: The pathogenesis of essential hypertension is multifactorial with different underlying mechanisms contributing to disease. We have recently shown that TNF superfamily member 14 LIGHT (an acronym for homologous to lymphotoxins, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, also known as TNFSF14) induces hypertension when injected into mice. Research reported here was undertaken to examine the role of transglutaminase (TGase) in LIGHT-induced hypertension. METHODS AND RESULTS: Initial experiments showed that plasma and kidney TGase activity was induced by LIGHT infusion (13.91±2.92 versus 6.75±1.92 mU/mL and 19.86±3.55 versus 12.00±0.97 mU/10 µg) and was accompanied with hypertension (169±7.16 versus 117.17±11.57 mm Hg at day 14) and renal impairment (proteinuria, 61.33±23.21 versus 20.38±9.01 µg/mg; osmolality, 879.57±93.02 versus 1407.2±308.04 mmol/kg). The increase in renal TGase activity corresponded to an increase in RNA for the tissue TGase isoform, termed TG2. Pharmacologically, we showed that LIGHT-induced hypertension and renal impairment did not occur in the presence of cystamine, a well-known competitive inhibitor of TGase activity. Genetically, we showed that LIGHT-mediated induction of TGase, along with hypertension and renal impairment, was dependent on interleukin-6 and endothelial hypoxia inducible factor-1α. We also demonstrated that interleukin-6, endothelial hypoxia inducible factor-1α, and TGase are required for LIGHT-induced production of angiotensin receptor agonistic autoantibodies. CONCLUSIONS: Thus, LIGHT-induced hypertension, renal impairment, and production of angiotensin receptor agonistic autoantibodies require TGase, most likely the TG2 isoform. Our findings establish TGase as a critical link between inflammation, hypertension, and autoimmunity.


Subject(s)
Blood Pressure/drug effects , Hypertension/immunology , Inflammation/immunology , Transglutaminases/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Animals , Autoantibodies/immunology , Blood Pressure/immunology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Hypertension/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Kidney/immunology , Kidney/metabolism , Mice , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Proteinuria/immunology , Proteinuria/metabolism , Receptors, Angiotensin/immunology , Renal Insufficiency/immunology , Renal Insufficiency/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism
9.
J Cell Mol Med ; 20(10): 1861-71, 2016 10.
Article in English | MEDLINE | ID: mdl-27241100

ABSTRACT

LIGHT recruits and activates naive T cells in the islets at the onset of diabetes. IFN-γ secreted by activated T lymphocytes is involved in beta cell apoptosis. However, whether LIGHT sensitizes IFNγ-induced beta cells destruction remains unclear. In this study, we used the murine beta cell line MIN6 and primary islet cells as models for investigating the underlying cellular mechanisms involved in LIGHT/IFNγ - induced pancreatic beta cell destruction. LIGHT and IFN-γ synergistically reduced MIN6 and primary islet cells viability; decreased cell viability was due to apoptosis, as demonstrated by a significant increase in Annexin V(+) cell percentage, detected by flow cytometry. In addition to marked increases in cytochrome c release and NF-κB activation, the combination of LIGHT and IFN-γ caused an obvious decrease in expression of the anti-apoptotic proteins Bcl-2 and Bcl-xL, but an increase in expression of the pro-apoptotic proteins Bak and Bax in MIN6 cells. Accordingly, LIGHT deficiency led to a decrease in NF-κB activation and Bak expression, and peri-insulitis in non-obese diabetes mice. Inhibition of NF-κB activation with the specific NF-κB inhibitor, PDTC (pyrrolidine dithiocarbamate), reversed Bcl-xL down-regulation and Bax up-regulation, and led to a significant increase in LIGHT- and IFN-γ-treated cell viability. Moreover, cleaved caspase-9, -3, and PARP (poly (ADP-ribose) polymerase) were observed after LIGHT and IFN-γ treatment. Pretreatment with caspase inhibitors remarkably attenuated LIGHT- and IFNγ-induced cell apoptosis. Taken together, our results indicate that LIGHT signalling pathway combined with IFN-γ induces beta cells apoptosis via an NF-κB/Bcl2-dependent mitochondrial pathway.


Subject(s)
Apoptosis/drug effects , Interferon-gamma/pharmacology , Mitochondria/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cell Survival/drug effects , Cytochromes c/metabolism , Enzyme Activation/drug effects , Female , Insulin-Secreting Cells/metabolism , Mice, Inbred NOD , Mitochondria/drug effects , Models, Biological , Recombinant Fusion Proteins/metabolism , Stress, Physiological/drug effects
10.
J Immunol ; 195(5): 2429-41, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26209626

ABSTRACT

Lung epithelial cells are considered important sources of inflammatory molecules and extracellular matrix proteins that contribute to diseases such as asthma. Understanding the factors that stimulate epithelial cells may lead to new insights into controlling lung inflammation. This study sought to investigate the responsiveness of human lung epithelial cells to the TNF family molecules LIGHT and lymphotoxin αß (LTαß). Bronchial and alveolar epithelial cell lines, and primary human bronchial epithelial cells, were stimulated with LIGHT and LTαß, and expression of inflammatory cytokines and chemokines and markers of epithelial-mesenchymal transition and fibrosis/remodeling was measured. LTß receptor, the receptor shared by LIGHT and LTαß, was constitutively expressed on all epithelial cells. Correspondingly, LIGHT and LTαß strongly induced a limited but highly distinct set of inflammatory genes in all epithelial cells tested, namely the adhesion molecules ICAM-1 and VCAM-1; the chemokines CCL5, CCL20, CXCL1, CXCL3, CXCL5, and CXCL11; the cytokines IL-6, activin A and GM-CSF; and metalloproteinases matrix metalloproteinase-9 and a disintegrin and metalloproteinase domain-8. Importantly, induction of the majority of these inflammatory molecules was insensitive to the suppressive effects of the corticosteroid budesonide. LIGHT and LTαß also moderately downregulated E-cadherin, a protein associated with maintaining epithelial integrity, but did not significantly drive production of extracellular matrix proteins or α-smooth muscle actin. Thus, LIGHT and LTαß induce a distinct steroid-resistant inflammatory signature in airway epithelial cells via constitutively expressed LTß receptor. These findings support our prior murine studies that suggested the receptors for LIGHT and LTαß contribute to development of lung inflammation characteristic of asthma and idiopathic pulmonary fibrosis.


Subject(s)
Epithelial Cells/drug effects , Glucocorticoids/pharmacology , Inflammation Mediators/metabolism , Lymphotoxin alpha1, beta2 Heterotrimer/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Bronchi/cytology , Budesonide/pharmacology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Drug Resistance , Epithelial Cells/metabolism , Flow Cytometry , Gene Expression/drug effects , Humans , Immunoblotting , Lymphocytes/drug effects , Lymphocytes/metabolism , Monocytes/drug effects , Monocytes/metabolism , Receptors, Leukotriene B4/genetics , Receptors, Leukotriene B4/metabolism , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Mol Immunol ; 62(1): 96-103, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24980868

ABSTRACT

Signals mediated by members of the tumor necrosis factor receptor superfamily modulate a network of diverse processes including initiation of inflammatory responses and altering cell fate between pathways favoring survival and death. Although such pathways have been well-described for the TNF-α receptor, less is known about signaling induced by the TNF superfamily member LIGHT and how it is differentially altered by expression of its two receptors LTßR and HVEM in the same cell. We used cell lines with different relative expression of HVEM and LTßR to show that LIGHT-induced signals mediated by these receptors were associated with altered TRAF2 stability and RelA nuclear translocation. Production of the inflammatory chemokine CXCL10 was primarily mediated by LTßR. Higher expression of HVEM was associated with cell survival, while unopposed LTßR signaling favored pathways leading to apoptosis. Importantly, restoring HVEM expression in cells with low endogenous expression recapitulated the phenotype of cells with higher endogenous expression. Together, our data provide evidence that relative expression of HVEM and LTßR modulates canonical NF-κB and pro-apoptotic signals stimulated by LIGHT.


Subject(s)
Lymphocyte Activation , Lymphotoxin beta Receptor/physiology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Down-Regulation/drug effects , Down-Regulation/immunology , Gene Expression/physiology , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Interferon-gamma/pharmacology , Ligands , Lymphocyte Activation/drug effects , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , U937 Cells
12.
Sci Signal ; 7(311): ra13, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24497610

ABSTRACT

Precise regulation of nuclear factor κB (NF-κB) signaling is crucial for normal immune responses, and defective NF-κB activity underlies a range of immunodeficiencies. NF-κB is activated through two signaling cascades: the classical and noncanonical pathways. The classical pathway requires inhibitor of κB kinase ß (IKKß) and NF-κB essential modulator (NEMO), and hypomorphic mutations in the gene encoding NEMO (ikbkg) lead to inherited immunodeficiencies, collectively termed NEMO-ID. Noncanonical NF-κB activation requires NF-κB-inducing kinase (NIK) and IKKα, but not NEMO. We found that noncanonical NF-κB was basally active in peripheral blood mononuclear cells from NEMO-ID patients and that noncanonical NF-κB signaling was similarly enhanced in cell lines lacking functional NEMO. NIK, which normally undergoes constitutive degradation, was aberrantly present in resting NEMO-deficient cells, and regulation of its abundance was rescued by reconstitution with full-length NEMO, but not a mutant NEMO protein unable to physically associate with IKKα or IKKß. Binding of NEMO to IKKα was not required for ligand-dependent stabilization of NIK or noncanonical NF-κB signaling. Rather, an intact and functional IKK complex was essential to suppress basal NIK activity in unstimulated cells. Despite interacting with IKKα and IKKß to form an IKK complex, NEMO mutants associated with immunodeficiency failed to rescue classical NF-κB signaling or reverse the accumulation of NIK. Together, these findings identify a crucial role for classical NF-κB activity in the suppression of basal noncanonical NF-κB signaling.


Subject(s)
Immunologic Deficiency Syndromes/metabolism , Leukocytes, Mononuclear/metabolism , NF-kappa B/metabolism , Signal Transduction , Animals , Cells, Cultured , Gene Expression/drug effects , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Immunoblotting , Immunologic Deficiency Syndromes/genetics , Jurkat Cells , Leukocytes, Mononuclear/drug effects , Mice , Mutation , NF-kappa B p52 Subunit/metabolism , NIH 3T3 Cells , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , NF-kappaB-Inducing Kinase
13.
Gastric Cancer ; 16(2): 155-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22850801

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have the ability to migrate into tumors and therefore are potential vehicles for the therapy of malignant diseases. In this study, we investigated the use of umbilical cord blood mesenchymal stem cells (UCB-MSCs) as carriers for a constant source of transgenic LIGHT (TNFSF14) to target tumor cells in vivo. METHODS: Lentiviral vectors carrying LIGHT genes were constructed, producing viral particles with a titer of 2 × 10(8) TU/L. Fourteen days after UCB-MSCs transfected by LIGHT gene packaged lentivirus had been injected into mouse gastric cancer models, the expression levels of LIGHT mRNA and protein were detected by reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). Then the tumors' approximate volumes were measured. RESULTS: The treatment with MSC-LIGHT demonstrated a strong suppressive effect on tumor growth compared to treatment with MSC and NaCl (p < 0.001). Examination of pathological sections of the tumor tissues showed that the areas of tumor necrocis in the MSC-LIGHT group were larger than those in the MSC group. Moreover, we found that MSCs with LIGHT were able to significantly induce apoptosis of tumor cells. The expression levels of LIGHT mRNA and protein were significantly higher in the UCB-MSCs with the LIGHT gene than the levels in UCB-MSCs (p < 0.001). CONCLUSION: These results suggest that UCB-MSCs carrying the LIGHT gene have the potential to be used as effective delivery vehicles in the treatment of gastric cancers.


Subject(s)
Fetal Blood/cytology , Genetic Therapy/methods , Mesenchymal Stem Cells/metabolism , Stomach Neoplasms/therapy , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Base Sequence , Disease Models, Animal , Gene Expression Regulation , Humans , Lentivirus/genetics , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/virology , Mice , Mice, Nude , Molecular Sequence Data , Stomach Neoplasms/pathology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
14.
Biochem Biophys Res Commun ; 428(4): 451-7, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23131560

ABSTRACT

Fibrosis is an abnormal response to organ injury, characterized by accumulation of activated fibroblasts at the sites of injury. Fibroblasts arise from several sources, including resident fibroblasts and circulating fibrocytes that infiltrate organ tissue. Recently, epithelial-mesenchymal transition (EMT) has been recognized as a source of mesenchymal cells. EMT is induced by various growth factors, such as transforming growth factor (TGF)-ß1, and enhanced by inflammatory cytokines. Recently the tumor necrosis factor superfamily member LIGHT has been implicated in the pathogenesis of inflammatory disease and airway remodeling in severe asthma. We hypothesized that LIGHT might contribute to the pathogenesis of airway fibrosis via enhancement of EMT. Therefore, we investigated LIGHT's ability to induce EMT. A549 cells were stimulated with LIGHT, TGF-ß1 or both for 48h. To estimate EMT, we evaluated the expression of epithelial and mesenchymal markers using immunocytochemistry, Western blotting and quantitative RT-PCR. Signaling pathways for EMT were characterized by Western analysis to detect phosphorylation of Erk1/2 and smad2. LIGHT enhanced TGF-ß1-induced EMT both morphologically, by suppressing E-cadherin and enhancing vimentin, and functionally, by enhancing cell contractility. Additionally, LIGHT induced EMT without TGF-ß1. Evaluation of the mechanism showed that LIGHT did not induce TGF-ß1 production or affect the smad-snai1 pathway. Inhibition of Erk1/2 phosphorylation reduced LIGHT-induced EMT, indicating the Erk1/2 pathway to be a key pathway in LIGHT-induced EMT. In summary, LIGHT enhanced TGF-ß1-induced EMT but also induced EMT via the Erk1/2 pathway by itself, without TGF-ß1 signaling. LIGHT may contribute to the pathogenesis of airway fibrosis through enhancement of EMT.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Pulmonary Alveoli/physiology , Respiratory Mucosa/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Fibrosis , Humans , MAP Kinase Signaling System , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Respiratory Mucosa/drug effects , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
15.
Clin Exp Immunol ; 168(3): 318-24, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22519595

ABSTRACT

LIGHT [the name of which is derived from 'homologous to lymphotoxins, exhibits inducible expression, competes with herpes simplex virus glycoprotein D for herpes simplex virus entry mediator (HVEM), and expressed by T lymphocytes'], is a member of the tumour necrosis factor superfamily that is involved in various inflammatory diseases. We aimed to estimate the relevance of plasma LIGHT levels as a biomarker for atopic dermatitis (AD). In order to understand the putative role of LIGHT in AD pathogenesis, we also investigate the effects of LIGHT on a monocytic cell line, human acute monocytic leukaemia cell line (THP-1). We examined plasma LIGHT levels, total serum IgE, serum value of CCL17 and peripheral blood eosinophil counts in patients with AD and healthy subjects. The effects of LIGHT on activation and apoptosis in THP-1 cells were also investigated. The plasma concentrations of LIGHT in AD patients were significantly higher than those in healthy individuals and the concentrations decreased as the symptoms were improved by treatment. The LIGHT plasma concentrations correlated with IgE levels and the Severity Scoring of AD (SCORAD) index. In addition, LIGHT stimulation increased expression of CD86 and induced production of interleukin-1ß in THP-1 cells. Apoptosis was inhibited, the Bcl-2 level increased and the caspase-3 level decreased in THP-1 cells stimulated with LIGHT, compared to unstimulated control cells. These results suggest that plasma LIGHT levels may be one of the promising biomarkers for AD.


Subject(s)
Dermatitis, Atopic/blood , Dermatitis, Atopic/immunology , Eosinophils/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/blood , Adult , Apoptosis/drug effects , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Biomarkers/blood , Cell Line, Tumor , Chemokine CCL17/blood , Disease Progression , Eosinophils/pathology , Female , Humans , Immunoglobulin E/blood , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Middle Aged , Monocytes/drug effects , Monocytes/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Up-Regulation , Young Adult
16.
Cytokine ; 57(2): 294-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22172512

ABSTRACT

Rheumatoid arthritis (RA) is associated with pathological bone destruction mediated by osteoclasts. Although RANKL has been reported as a crucial factor for osteoclastogenesis, several other factors increased in RA support osteoclast formation and resorption in the absence of RANKL such as TNF-alpha and LIGHT. To date, in vitro bone resorption experiments are reported as the mean area of bone resorption per cortical or dentine slices and do not provide any information about depth and volume of resorption. The aims of this study were to assess these parameters by light microscopy and vertical scanning profilometry (VSP). Peripheral blood mononuclear cells were used as a source of osteoclast precursors and were cultured for up to 21 days in the presence of RANKL, TNF-alpha/IL-1 or LIGHT. Mean area, depth and volume of resorption were assessed by light microscopy and vertical scanning profilometry. As expected, RANKL induced large resorption pits (10,876 ± 2190µm(2)) whereas TNF-alpha/IL-1 and LIGHT generated smaller pits (respectively 1328 ± 210 and 1267 ± 173µm(2)) with no noticeable differences between these two cytokines. Depth and volume of resorption measured by VSP showed that RANKL promoted deep resorption pits resulting in large volume of resorption. Interestingly, although mean area of resorption was similar between TNF-alpha/IL-1 and LIGHT, the depth and volume of resorption of these lacunae were significantly increased by 2-fold with TNF-alpha/IL-1. These results provide evidence that although LIGHT appeared elevated in the synovial fluid of RA patients, its role in bone resorption is less than TNF-alpha/IL-1 or RANKL.


Subject(s)
Bone Resorption/pathology , Interleukin-1/pharmacology , Osteoclasts/drug effects , Osteoclasts/pathology , RANK Ligand/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus Size/drug effects , Humans , Models, Biological
17.
Protein Expr Purif ; 80(2): 239-45, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21745574

ABSTRACT

LIGHT is a membrane-bound protein that belongs to the tumor necrosis factor (TNF) superfamily ligands. In this study, we established an effective strategy for producing a bioactive soluble form of LIGHT (sLIGHT), an extracellular region (Ile84-Val²4°) of human LIGHT. Because sLIGHT was expressed as inclusion bodies in Escherichia coli, we investigated reagents that enhance the renaturation of sLIGHT from the inclusion bodies. Interestingly, L-cysteine in the denaturation buffer containing 3.5 M guanidine hydrochloride significantly improved the renaturation efficiency of sLIGHT. The effect of L-cysteine was synergistically enhanced by L-arginine in the refolding buffer. The optimal concentrations of L-cysteine and L-arginine in the denaturation and refolding buffers were 8 mM and 0.8 M, respectively. With these buffers, approximately 90 mg of sLIGHT was purified from 200 g of frozen E. coli cells. sLIGHT thus obtained significantly induced apoptosis in the WiDr human colon adenocarcinoma cell line at nanomolar concentrations, the same amount of sLIGHT that was produced by Sf9 insect cells. These results suggest that L-cysteine in the denaturation buffer enhances the renaturation of recombinant proteins from inclusion bodies in E. coli.


Subject(s)
Cysteine/pharmacology , Escherichia coli/metabolism , Inclusion Bodies/metabolism , Protein Renaturation , Tumor Necrosis Factor Ligand Superfamily Member 14/isolation & purification , Alanine/pharmacology , Apoptosis , Biological Assay , Buffers , Cell Line, Tumor , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Guanidine/pharmacology , Humans , Plasmids/genetics , Plasmids/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
18.
Vascul Pharmacol ; 53(1-2): 28-37, 2010.
Article in English | MEDLINE | ID: mdl-20298810

ABSTRACT

LIGHT is known to act as a novel mediator for atherogenesis. Furthermore, it has been reported that emodin, an active component extracted from rhubarb, can stop the growth of cancer cells. However, it is not known if emodin exerts anti-atherogenic effects in the human monocyte, THP-1, following treatment with LIGHT. In this study, we evaluated the inhibitory effect of emodin and rhein on LIGHT-induced migration in THP-1. Emodin and rhein decreased the level of LIGHT-induced generation of ROS, as well as the expression of CCR1, CCR2 and ICAM-1 and the production of IL-8, MCP-1, TNF-alpha, and IL-6. Emodin and rhein also decreased the phosphorylation of the p38 MAPK and IkB-alpha. Furthermore, the NADPH oxidase assembly inhibitor, AEBSF, and the blocker of NADPH oxidase, p47(phox) small interference RNA (siRNA), also efficiently blocked LIGHT-induced migration, CCR1, CCR2, ICAM-1, and HVEM expression, and p38 MAPK and NF-kB activation. These findings indicate that the inhibitory effects of emodin and rhein on LIGHT-induced migration occur via decreasing ROS production and NADPH oxidase p47(phox) activation. Taken together, these results indicate that emodin and rhein have the potential for use as an anti-atherosclerosis agent.


Subject(s)
Anthraquinones/pharmacology , Emodin/pharmacology , Reactive Oxygen Species/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/physiopathology , Cell Line , Cell Movement , Enzyme Inhibitors/pharmacology , Humans , Monocytes/drug effects , Monocytes/metabolism , NADPH Oxidases/drug effects , NADPH Oxidases/metabolism , Recombinant Proteins , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
19.
Mol Immunol ; 47(4): 666-70, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19939453

ABSTRACT

TNFSF14 is involved in the pathogenesis of some inflammatory diseases such as arthritis. CXCL10 and CXCL11 recruit Th1 cells, and the productions of these chemokines are related to the exacerbation of some inflammatory diseases including arthritis and periodontal disease. We examined in vitro effects of TNFSF14 on IFN-gamma-induced CXCL10 and CXCL11 production in human gingival fibroblasts (HGFs). HGFs constitutively expressed TNFSF14 receptors, LTbetaR and HVEM. TNFSF14 enhanced IFN-gamma-induced secretion of CXCL10 and CXCL11 from HGFs. IFN-gamma treatment increased HVEM expression on HGFs. TNFSF14 in combination with IFN-gamma resulted in increased activation of p38 MAPK, ERK and IkappaB-alpha compared with TNFSF14 or IFN-gamma alone. Moreover, inhibitors of p38 MAPK, ERK and NF-kappaB abolished the CXCL10 and CXCL11 productions from TNFSF14 with IFN-gamma-stimulated HGFs. These effects of TNFSF14 may promote the infiltration of Th1 cells into lesions with inflammatory diseases. TNFSF14 might act as a proinflammatory cytokine in some inflammatory diseases thus is a candidate therapeutic target.


Subject(s)
Chemokine CXCL10/biosynthesis , Chemokine CXCL11/biosynthesis , Fibroblasts/drug effects , Fibroblasts/metabolism , Gingiva/cytology , Interferon-gamma/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/enzymology , Humans , I-kappa B Proteins/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
20.
Int J Obes (Lond) ; 34(1): 146-56, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19786966

ABSTRACT

BACKGROUND: LIGHT (lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells) is a member of the tumor necrosis factor (TNF) family, primarily expressed in lymphocytes, which was associated with the induction of pro-inflammatory cytokines and alterations of lipid homeostasis in animal models. We aimed to analyze whether LIGHT has a role in the human obesity-associated inflammatory status. METHODS: The association between circulating LIGHT concentrations and clinical variables was studied in 190 subjects with different degrees of obesity and glucose tolerance. The expression and release of 21 different cytokines, and the expression of genes involved in lipid metabolism were also evaluated after stimulation with LIGHT in cultured human differentiated adipocytes. RESULTS: Serum LIGHT concentrations positively associated with body mass index (BMI), fat mass, glycated hemoglobin and fasting triglycerides, and negatively with high-density lipoprotein cholesterol. Circulating LIGHT concentrations were significantly increased in morbidly obese subjects and in patients with type 2 diabetes. LIGHT induced the secretion of several cytokines and upregulated the expression and secretion of interleukin-6 (IL-6), IL-8, Growth Regulated Oncogene (GRO) and monocyte chemotactic protein-1 (MCP-1). These observations were concomitant with the activation of nuclear factor (NF)-kappaB signalling in human differentiated adipocytes. LIGHT also upregulated the expression and synthesis of its own receptor (herpesvirus entry mediator (HVEM)) and decreased the expression of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) and fatty acid synthase. CONCLUSION: These data suggest that LIGHT may have a role in mediating chronic inflammation and alterations of lipid metabolism in obese subjects.


Subject(s)
Adipocytes/metabolism , Cytokines/metabolism , Hypertriglyceridemia/metabolism , Obesity/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Body Mass Index , Cells, Cultured , Humans , Hypertriglyceridemia/genetics , Male , Obesity/genetics , Up-Regulation/genetics , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...