1
|
Zheng L, Chen W, Yao K, Xie Y, Liao C, Zhou T. Clinical and preclinical studies of mesenchymal stem cells to alleviate peritoneal fibrosis. Stem Cell Res Ther 2024; 15:237. [PMID: 39080683 PMCID: PMC11290310 DOI: 10.1186/s13287-024-03849-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Peritoneal dialysis is an important part of end-stage kidney disease replacement therapy. However, prolonged peritoneal dialysis can result in peritoneal fibrosis and ultrafiltration failure, forcing patients to withdraw from peritoneal dialysis treatment. Therefore, there is an urgent need for some effective measures to alleviate the occurrence and progression of peritoneal fibrosis. Mesenchymal stem cells play a crucial role in immunomodulation and antifibrosis. Numerous studies have investigated the fact that mesenchymal stem cells can ameliorate peritoneal fibrosis mainly through the paracrine pathway. It has been discovered that mesenchymal stem cells participate in the improvement of peritoneal fibrosis involving the following signaling pathways: TGF-β/Smad signaling pathway, AKT/FOXO signaling pathway, Wnt/β-catenin signaling pathway, TLR/NF-κB signaling pathway. Additionally, in vitro experiments, mesenchymal stem cells have been shown to decrease mesothelial cell death and promote proliferation. In animal models, mesenchymal stem cells can enhance peritoneal function by reducing inflammation, neovascularization, and peritoneal thickness. Mesenchymal stem cell therapy has been demonstrated in clinical trials to improve peritoneal function and reduce peritoneal fibrosis, thus improving the life quality of peritoneal dialysis patients.
Collapse
Affiliation(s)
- Lingqian Zheng
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Wenmin Chen
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Kaijin Yao
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Yina Xie
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Chunling Liao
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China.
| |
Collapse
|
2
|
Verma S, Moreno IY, Prinholato da Silva C, Sun M, Cheng X, Gesteira TF, Coulson-Thomas VJ. Endogenous TSG-6 modulates corneal inflammation following chemical injury. Ocul Surf 2024; 32:26-38. [PMID: 38151073 PMCID: PMC11056311 DOI: 10.1016/j.jtos.2023.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/21/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
PURPOSE Tumor necrosis factor (TNF)-stimulated gene-6 (TSG-6) is upregulated in various pathophysiological contexts, where it has a diverse repertoire of immunoregulatory functions. Herein, we investigated the expression and function of TSG-6 during corneal homeostasis and after injury. METHODS Human corneas, eyeballs from BALB/c (TSG-6+/+), TSG-6+/- and TSG-6-/- mice, human immortalized corneal epithelial cells and murine corneal epithelial progenitor cells were prepared for immunostaining and real time PCR analysis of endogenous expression of TSG-6. Mice were subjected to unilateral corneal debridement or alkali burn (AB) injuries and wound healing assessed over time using fluorescein stain, in vivo confocal microscopy and histology. RESULTS TSG-6 is endogenously expressed in the human and mouse cornea and established corneal epithelial cell lines and is upregulated after injury. A loss of TSG-6 has no structural and functional effect in the cornea during homeostasis. No differences were noted in the rate of corneal epithelial wound closure between BALB/c, TSG-6+/- and TSG-6-/- mice. TSG-6-/- mice presented decreased inflammatory response within the first 24 h of injury and accelerated corneal wound healing following AB when compared to control mice. CONCLUSION TSG-6 is endogenously expressed in the cornea and upregulated after injury where it propagates the inflammatory response following chemical injury.
Collapse
Affiliation(s)
- Sudhir Verma
- College of Optometry, University of Houston, Houston, TX, United States; Department of Zoology, Deen Dayal Upadhyaya College, University of Delhi, Delhi, India
| | - Isabel Y Moreno
- College of Optometry, University of Houston, Houston, TX, United States
| | | | - Mingxia Sun
- College of Optometry, University of Houston, Houston, TX, United States
| | - Xuhong Cheng
- College of Optometry, University of Houston, Houston, TX, United States
| | - Tarsis F Gesteira
- College of Optometry, University of Houston, Houston, TX, United States
| | | |
Collapse
|
3
|
Drummond SP, Bartnik E, Kouvatsos N, Scott JL, Dyer DP, Thomson JM, Price AJ, Anand S, Biant LC, Leeuw T, Herrmann M, Milner CM, Day AJ. The recombinant Link module of human TSG-6 suppresses cartilage damage in models of osteoarthritis: A potential disease-modifying OA drug. Osteoarthritis Cartilage 2023; 31:1353-1364. [PMID: 37257556 DOI: 10.1016/j.joca.2023.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
OBJECTIVE To investigate the role of endogenous TSG-6 in human osteoarthritis (OA) and assess the disease-modifying potential of a TSG-6-based biological treatment in cell, explant and animal models of OA. DESIGN Knee articular cartilages from OA patients were analyzed for TSG-6 protein and mRNA expression using immunohistochemistry and RNAscope, respectively. The inhibitory activities of TSG-6 and its isolated Link module (Link_TSG6) on cytokine-induced degradation of OA cartilage explants were compared. Human mesenchymal stem/stromal cell-derived chondrocyte pellet cultures were used to determine the effects of Link_TSG6 and full-length TSG-6 on IL-1α-, IL-1β-, or TNF-stimulated ADAMTS4, ADAMTS5, and MMP13 mRNA expression. Link_TSG6 was administered i.a. to the rat ACLTpMMx model; cartilage damage and tactile allodynia were assessed. RESULTS TSG-6 is predominantly associated with chondrocytes in regions of cartilage damage where high TSG-6 expression aligns with low MMP13, the major collagenase implicated in OA progression. Link_TSG6 is more potent than full-length TSG-6 at inhibiting cytokine-mediated matrix breakdown in human OA cartilage explants;>50% of donor cartilages, from 59 tested, were responsive to Link_TSG6 treatment. Link_TSG6 also displayed more potent effects in 3D pellet cultures, suppressing ADAMTS4, ADAMTS5, and MMP13 gene expression, which was consistent with reduced aggrecanase and collagenase activities in explant cultures. Link_TSG6 treatment reduced touch-evoked pain behavior and dose-dependently inhibited cartilage damage in a rodent model of surgically-induced OA. CONCLUSIONS Link_TSG6 has enhanced chondroprotective activity compared to the full-length TSG-6 protein and shows potential as a disease modifying OA drug via its inhibition of aggrecanase and collagenase activity.
Collapse
Affiliation(s)
- Sheona P Drummond
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Eckart Bartnik
- Sanofi Aventis Deutschland GmbH, D-65926 Frankfurt, Germany
| | - Nikolaos Kouvatsos
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jenny L Scott
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Douglas P Dyer
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jennifer M Thomson
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Andrew J Price
- Nuffield Department of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Sanjay Anand
- Department of Orthopaedics, Stepping Hill Hospital, Stockport, UK
| | - Leela C Biant
- Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Manchester Orthopaedic Centre, Manchester University Hospitals Foundation Trust, Manchester, UK
| | - Thomas Leeuw
- Sanofi Aventis Deutschland GmbH, D-65926 Frankfurt, Germany
| | | | - Caroline M Milner
- Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| | - Anthony J Day
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
4
|
Albtoush N, Queisser KA, Zawerton A, Lauer ME, Beswick EJ, Petrey AC. TSG6 hyaluronan matrix remodeling dampens the inflammatory response during colitis. Matrix Biol 2023; 121:149-166. [PMID: 37391162 PMCID: PMC10530565 DOI: 10.1016/j.matbio.2023.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
In response to tissue injury, changes in the extracellular matrix (ECM) can directly affect the inflammatory response and contribute to disease progression or resolution. During inflammation, the glycosaminoglycan hyaluronan (HA) becomes modified by tumor necrosis factor stimulated gene-6 (TSG6). TSG6 covalently transfers heavy chain (HC) proteins from inter-α-trypsin inhibitor (IαI) to HA in a transesterification reaction and is to date is the only known HC-transferase. By modifying the HA matrix, TSG6 generates HC:HA complexes that are implicated in mediating both protective and pathological responses. Inflammatory bowel disease (IBD) is a lifelong chronic disorder with well-described remodeling of the ECM and increased mononuclear leukocyte influx into the intestinal mucosa. Deposition of HC:HA matrices is an early event in inflamed gut tissue that precedes and promotes leukocyte infiltration. However, the mechanisms by which TSG6 contributes to intestinal inflammation are not well understood. The aim of our study was to understand how the TSG6 and its enzymatic activity contributes to the inflammatory response in colitis. Our findings indicate that inflamed tissues of IBD patients show an elevated level of TSG6 and increased HC deposition and that levels of HA strongly associate with TSG6 levels in patient colon tissue specimens. Additionally, we observed that mice lacking TSG6 are more vulnerable to acute colitis and exhibit an aggravated macrophage-associated mucosal immune response characterized by elevated pro-inflammatory cytokines and chemokines and diminished anti-inflammatory mediators including IL-10. Surprisingly, along with significantly increased levels of inflammation in the absence of TSG6, tissue HA levels in mice were found to be significantly reduced and disorganized, absent of typical "HA-cable" structures. Inhibition of TSG6 HC-transferase activity leads to a loss of cell surface HA and leukocyte adhesion, indicating that the enzymatic functions of TSG6 are a major contributor to stability of the HA ECM during inflammation. Finally, using biochemically generated HC:HA matrices derived by TSG6, we show that HC:HA complexes can attenuate the inflammatory response of activated monocytes. In conclusion, our data suggests that TSG6 exerts a tissue-protective, anti-inflammatory effect via the generation of HC:HA complexes that become dysregulated in IBD.
Collapse
Affiliation(s)
- Nansy Albtoush
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112; Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kimberly A Queisser
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112; Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ash Zawerton
- Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mark E Lauer
- Lerner Research Institute, Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ellen J Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Aaron C Petrey
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112; Department of Pathology, Division of Microbiology & Immunology, University of Utah School of Medicine, Salt Lake City, Utah, 84132, USA; Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA; Lerner Research Institute, Department of Inflammation & Immunity, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
5
|
La Russa D, Di Santo C, Lizasoain I, Moraga A, Bagetta G, Amantea D. Tumor Necrosis Factor (TNF)-α-Stimulated Gene 6 (TSG-6): A Promising Immunomodulatory Target in Acute Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24021162. [PMID: 36674674 PMCID: PMC9865344 DOI: 10.3390/ijms24021162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/26/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Tumor necrosis factor (TNF)-α-stimulated gene 6 (TSG-6), the first soluble chemokine-binding protein to be identified in mammals, inhibits chemotaxis and transendothelial migration of neutrophils and attenuates the inflammatory response of dendritic cells, macrophages, monocytes, and T cells. This immunoregulatory protein is a pivotal mediator of the therapeutic efficacy of mesenchymal stem/stromal cells (MSC) in diverse pathological conditions, including neuroinflammation. However, TSG-6 is also constitutively expressed in some tissues, such as the brain and spinal cord, and is generally upregulated in response to inflammation in monocytes/macrophages, dendritic cells, astrocytes, vascular smooth muscle cells and fibroblasts. Due to its ability to modulate sterile inflammation, TSG-6 exerts protective effects in diverse degenerative and inflammatory diseases, including brain disorders. Emerging evidence provides insights into the potential use of TSG-6 as a peripheral diagnostic and/or prognostic biomarker, especially in the context of ischemic stroke, whereby the pathobiological relevance of this protein has also been demonstrated in patients. Thus, in this review, we will discuss the most recent data on the involvement of TSG-6 in neurodegenerative diseases, particularly focusing on relevant anti-inflammatory and immunomodulatory functions. Furthermore, we will examine evidence suggesting novel therapeutic opportunities that can be afforded by modulating TSG-6-related pathways in neuropathological contexts and, most notably, in stroke.
Collapse
Affiliation(s)
- Daniele La Russa
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
| | - Chiara Di Santo
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, and Instituto de Investigación Hospital 12 de Octubre (Imas12), 28040 Madrid, Spain
| | - Ana Moraga
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, and Instituto de Investigación Hospital 12 de Octubre (Imas12), 28040 Madrid, Spain
| | - Giacinto Bagetta
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
| | - Diana Amantea
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
- Correspondence:
| |
Collapse
|
6
|
Groeger SE, Hudel M, Zechel‐Gran S, Herrmann JM, Chakraborty T, Domann E, Meyle J. Recombinant
Porphyromonas gingivalis
W83 FimA alters immune response and metabolic gene expression in oral squamous carcinoma cells. Clin Exp Dent Res 2022; 8:976-987. [PMID: 35570325 PMCID: PMC9382057 DOI: 10.1002/cre2.588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives The Gram‐negative anaerobic rod Porphyromonas gingivalis (P. gingivalis) is regarded as a keystone pathogen in periodontitis and expresses a multitude of virulence factors iincluding fimbriae that are enabling adherence to and invasion in cells and tissues. The progression of periodontitis is a consequence of the interaction between the host immune response and periodontal pathogens. The aim of this study was to investigate the genome‐wide impact of recombinant fimbrial protein FimA from P. gingivalis W83 on the gene expression of oral squamous carcinoma cells by transcriptome analysis. Materials and Methods Human squamous cell carcinoma cells (SCC‐25) were stimulated for 4 and 24 h with recombinant FimA. RNA sequencing was performed and differential gene expression and enrichment were analyzed using gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and REACTOME. The results of transcriptome analysis were validated using quantitative real‐time polymerase chain reaction (PCR) with selected genes. Results Differential gene expression after 4 and 24 h revealed upregulation of 464 (4 h) and 179 genes (24 h) and downregulation of 69 (4 h) and 312 (24 h) genes. GO, KEGG, and REACTONE enrichment analysis identified a strong immunologic transcriptomic response signature after 4 h. After 24 h, mainly those genes were regulated, which belonged to cell metabolic pathways and replication. Real‐time PCR of selected genes belonging to immune response and signaling demonstrated strong upregulation of CCL20, TNFAIP6, CXCL8, TNFAIP3, and NFkBIA after both stimulation times. Conclusions These data shed light on the RNA transcriptome of human oral squamous carcinoma epithelial cells following stimulation with P. gingivalis FimA and identify a strong immunological gene expression response to this virulence factor. The data provide a base for future studies of molecular and cellular interactions between P. gingivalis and oral epithelium to elucidate basic mechanisms that may provide new prospects for periodontitis therapy and give new insights into the development and possible treatments of cancer.
Collapse
Affiliation(s)
- Sabine E. Groeger
- Department of Periodontology Justus‐Liebig University of Giessen Giessen Germany
| | - Martina Hudel
- Institute of Medical Microbiology Justus‐Liebig University of Giessen Giessen Germany
| | - Silke Zechel‐Gran
- Institute of Medical Microbiology Justus‐Liebig University of Giessen Giessen Germany
| | - Jens M. Herrmann
- Department of Periodontology Justus‐Liebig University of Giessen Giessen Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology Justus‐Liebig University of Giessen Giessen Germany
- German Center for Infection Research (DZIF) Partner Site Giessen‐Marburg‐Langen Giessen Germany
| | - Eugen Domann
- Institute of Medical Microbiology Justus‐Liebig University of Giessen Giessen Germany
- German Center for Infection Research (DZIF) Partner Site Giessen‐Marburg‐Langen Giessen Germany
- Institute of Hygiene and Environmental Medicine Justus‐Liebig University of Giessen Giessen Germany
| | - Joerg Meyle
- Department of Periodontology Justus‐Liebig University of Giessen Giessen Germany
| |
Collapse
|
7
|
Kang I, Hundhausen C, Evanko SP, Malapati P, Workman G, Chan CK, Rims C, Firestein GS, Boyle DL, MacDonald KM, Buckner JH, Wight TN. Crosstalk between CD4 T cells and synovial fibroblasts from human arthritic joints promotes hyaluronan-dependent leukocyte adhesion and inflammatory cytokine expression in vitro. Matrix Biol Plus 2022; 14:100110. [PMID: 35573706 PMCID: PMC9097711 DOI: 10.1016/j.mbplus.2022.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/06/2022] [Accepted: 04/21/2022] [Indexed: 11/27/2022] Open
|
8
|
Hu T, Liu Y, Li X, Li X, Liu Y, Wang Q, Huang J, Yu J, Wu Y, Chen S, Zeng T, Tan L. Tumor necrosis factor-alpha stimulated gene-6: A biomarker reflecting disease activity in rheumatoid arthritis. J Clin Lab Anal 2022; 36:e24395. [PMID: 35353944 PMCID: PMC9102767 DOI: 10.1002/jcla.24395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To explore the serum tumor necrosis factor-alpha stimulated gene-6 (TSG-6) level and its association with disease activity in rheumatoid arthritis (RA) patients. METHODS We recruited 176 RA patients, 178 non-RA patients (lupus erythematosus, osteoarthritis, ulcerative colitis, ankylosing spondylitis and psoriasis) and 71 healthy subjects. Serum TSG-6 levels were detected by enzyme-linked immunosorbent assay (ELISA). RA patients were divided into inactive RA and active RA groups by disease activity score of 28 joints based on C-reactive protein (DAS28-CRP). The receiver operating characteristic (ROC) curve and Spearman's rank correlation test analyzed the correlation between TSG-6 concentration and RA disease activity. RESULTS Tumor necrosis factor-alpha stimulated gene-6 levels in the RA group were increased (p < 0.01). TSG-6 concentrations indicated an upward tendency with increased disease activity; The area under the curve (AUC) of TSG-6 for diagnosing RA and assessing the severity of RA were 0.78 and 0.80, respectively; The combination of TSG-6 and anti-mutated citrullinated vimentin antibodies (anti-MCV) (sensitivity:98.4%)improved the diagnostic accuracy of RA. Binary logistic regression analysis showed that TSG-6 was an independent risk factor related to the severity of RA, and OR (95% CI) was 1.2 (1.003-1.453). CONCLUSION The TSG-6 levels in RA patients were elevated and related to disease activity. Therefore, TSG-6 may serve as a new potential biomarker for evaluating RA disease activity.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China.,School of Public Health of Nanchang University, Nanchang, China
| | - Yuhan Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China.,School of Public Health of Nanchang University, Nanchang, China
| | - Xu Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Xiaohang Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Yanzhao Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Qunxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Jiayi Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianlin Yu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Yang Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Simei Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Tingting Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liming Tan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| |
Collapse
|
9
|
The link module of human TSG-6 (Link_TSG6) promotes wound healing, suppresses inflammation and improves glandular function in mouse models of Dry Eye Disease. Ocul Surf 2021; 24:40-50. [PMID: 34968766 DOI: 10.1016/j.jtos.2021.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE To investigate the potential of the Link_TSG6 polypeptide comprising the Link module of human TSG-6 (TNF-stimulated gene/protein-6) as a novel treatment for dry eye disease (DED). METHODS We analyzed the therapeutic effects of topical application of Link_TSG6 in two murine models of DED, the NOD.B10.H2b mouse model and the desiccating stress model. The effects of Link_TSG6 on the ocular surface and DED were compared with those of full-length TSG-6 (FL_TSG6) and of 0.05% cyclosporine (Restasis®). Additionally, the direct effect of Link_TSG6 on wound healing of the corneal epithelium was evaluated in a mouse model of corneal epithelial debridement. RESULTS Topical Link_TSG6 administration dose-dependently reduced corneal epithelial defects in DED mice while increasing tear production and conjunctival goblet cell density. At the highest dose, no corneal lesions remained in ∼50% of eyes treated. Also, Link_TSG6 significantly suppressed the levels of inflammatory cytokines at the ocular surface and inhibited the infiltration of T cells in the lacrimal glands and draining lymph nodes. Link_TSG6 was more effective in decreasing corneal epithelial defects than an equimolar concentration of FL_TSG6. Link_TSG6 was significantly more potent than Restasis® at ameliorating clinical signs and reducing inflammation. Link_TSG6 markedly and rapidly facilitated epithelial healing in mice with corneal epithelial debridement wounds. CONCLUSION Link_TSG6 holds promise as a novel therapeutic agent for DED through its effects on the promotion of corneal epithelial healing and tear secretion, the preservation of conjunctival goblet cells and the suppression of inflammation.
Collapse
|
10
|
Systematic review of robust experimental models of rheumatoid arthritis for basic research. DIGITAL CHINESE MEDICINE 2021. [DOI: 10.1016/j.dcmed.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
11
|
Šínová R, Pavlík V, Ondrej M, Velebný V, Nešporová K. Hyaluronan: A key player or just a bystander in skin photoaging? Exp Dermatol 2021; 31:442-458. [PMID: 34726319 DOI: 10.1111/exd.14491] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/07/2021] [Accepted: 10/29/2021] [Indexed: 02/06/2023]
Abstract
Photoaged skin exhibits signs of inflammation, DNA damage and changes in morphology that are visible at the macroscopic and microscopic levels. Photoaging also affects the extracellular matrix (ECM) including hyaluronan (HA), the main polysaccharide component thereof. HA is a structurally simple but biologically complex molecule that serves as a water-retaining component and provides both a scaffold for a number of the proteins of the ECM and the ligand for cellular receptors. The study provides an overview of the literature concerning the changes in HA amount, size and metabolism, and the potential role of HA in photoaging. We also suggest novel HA contributions to photoaging based on our knowledge of the role of HA in other pathological processes, including the senescence and inflammation-triggered ECM reorganization. Moreover, we discuss potential direct or indirect intervention to mitigate photoaging that targets the hyaluronan metabolism, as well as supplementation.
Collapse
Affiliation(s)
- Romana Šínová
- Contipro a.s., Dolní Dobrouč, Czech Republic.,Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Vojtěch Pavlík
- Contipro a.s., Dolní Dobrouč, Czech Republic.,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Ondrej
- Contipro a.s., Dolní Dobrouč, Czech Republic.,Department of Radiobiology, Faculty of Military Health Sciences, University of Defense in Brno, Hradec Kralove, Czech Republic
| | | | | |
Collapse
|
12
|
Lipoxin A4-Mediated p38 MAPK Signaling Pathway Protects Mice Against Collagen-Induced Arthritis. Biochem Genet 2020; 59:346-365. [PMID: 33221976 DOI: 10.1007/s10528-020-10016-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/12/2020] [Indexed: 01/06/2023]
Abstract
The aim of the article was to study the mechanism of Lipoxin A4 (LXA4)-mediated p38 MAPK pathway protecting mice against collagen-induced arthritis (CIA). The impact of LXA4 (0, 5, 10, 15 nM) on synoviocytes proliferation of CIA mice was detected using 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. CIA mice were treated with LXA4, SB203580 (a p38 inhibitor), and/or anisomycin (a p38 agonist), and the arthritis severity score in each mouse was determined. The gene or protein expressions were detected with Western Blotting, ELISA, or qRT-PCR. LXA4 inhibited the synoviocytes proliferation of CIA mice with decreased levels of TNF-α, IL-6, IL-1β, and IFN-γ and reduced p-p38/total p38 expression in synoviocytes in a dose-dependent manner. LXA4 levels were decreased in synovial tissues and plasma of CIA mice, but p-p38/total p38 expression was increased in synovial tissues. LXA4 could downregulate p-p38/total p38 expression in synovial tissues of CIA mice. Both LXA4 and SB203580 reduced arthritis severity score of CIA mice with the reduction of synovial tissue hyperplasia and inflammatory cell infiltration. CIA mice treated with LXA4 and SB203580 had lower levels of TNF-α, IL-6, IL-1β, and IFN-γ, accompanying decreased MDA as well as increased SOD, CAT,and GPx. However, anisomycin could reverse the protect effects of LXA4 on CIA mice regarding the abovementioned inflammatory factors and oxidative stress indexes. LXA4 protected mice against collagen-induced arthritis via inhibiting p38 MAPK signaling pathway, which may be a potential new therapeutic target for rheumatoid arthritis.
Collapse
|
13
|
Zhu L, Donhou S, Burleigh A, Miotla Zarebska J, Curtinha M, Parisi I, Khan SN, Dell'Accio F, Chanalaris A, Vincent TL. TSG-6 Is Weakly Chondroprotective in Murine OA but Does not Account for FGF2-Mediated Joint Protection. ACR Open Rheumatol 2020; 2:605-615. [PMID: 33029956 PMCID: PMC7571392 DOI: 10.1002/acr2.11176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/11/2020] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Tumor necrosis factor α-stimulated gene 6 (TSG-6) is an anti-inflammatory protein highly expressed in osteoarthritis (OA), but its influence on the course of OA is unknown. METHODS Cartilage injury was assessed by murine hip avulsion or by recutting rested explants. Forty-two previously validated injury genes were quantified by real-time polymerase chain reaction in whole joints following destabilization of the medial meniscus (DMM) (6 hours and 7 days). Joint pathology was assessed at 8 and 12 weeks following DMM in 10-week-old male and female fibroblast growth factor 2 (FGF2)-/- , TSG-6-/- , TSG-6tg (overexpressing), FGF2-/- ;TSG-6tg (8 weeks only) mice, as well as strain-matched, wild-type controls. In vivo cartilage repair was assessed 8 weeks following focal cartilage injury in TSG-6tg and control mice. FGF2 release following cartilage injury was measured by enzyme-linked immunosorbent assay. RESULTS TSG-6 messenger RNA upregulation was strongly FGF2-dependent upon injury in vitro and in vivo. Fifteeen inflammatory genes were significantly increased in TSG-6-/- joints, including IL1α, Ccl2, and Adamts5 compared with wild type. Six genes were significantly suppressed in TSG-6-/- joints including Timp1, Inhibin βA, and podoplanin (known FGF2 target genes). FGF2 release upon cartilage injury was not influenced by levels of TSG-6. Cartilage degradation was significantly increased at 12 weeks post-DMM in male TSG-6-/- mice, with a nonsignificant 30% reduction in disease seen in TSG-6tg mice. No differences were observed in cartilage repair between genotypes. TSG-6 overexpression was unable to prevent accelerated OA in FGF2-/- mice. CONCLUSION TSG-6 influences early gene regulation in the destabilized joint and exerts a modest late chondroprotective effect. Although strongly FGF2 dependent, TSG-6 does not explain the strong chondroprotective effect of FGF2.
Collapse
Affiliation(s)
- Linyi Zhu
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Shannah Donhou
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Annika Burleigh
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Jadwiga Miotla Zarebska
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Marcia Curtinha
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Ida Parisi
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Sumayya Nafisa Khan
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | | | - Anastasios Chanalaris
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| | - Tonia L Vincent
- Kennedy Institute of Rheumatology, Arthritis Research UK Centre for OA Pathogenesis, University of Oxford, UK
| |
Collapse
|
14
|
Shakya S, Mack JA, Alipour M, Maytin EV. Cutaneous Wounds in Mice Lacking TSG-6 Exhibit Delayed Closure and an Abnormal Inflammatory Response. J Invest Dermatol 2020; 140:2505-2514. [PMID: 32422216 DOI: 10.1016/j.jid.2020.04.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 04/12/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
We investigated how loss of TSG-6 affects wound closure and skin inflammation. TSG-6 has several known biological functions, including the enzymatic transfer of heavy-chain proteins from inter-α-trypsin inhibitor to hyaluronan to form heavy-chain protein-hyaluronan complexes. TSG-6 and heavy-chain protein-hyaluronan are constitutively expressed in normal skin and increase post-wounding but are completely absent in TSG-6-null mice. Wound closure rates are significantly delayed in TSG-6-null mice relative to wildtype mice. Neutrophil recruitment is delayed in early wounds (12 hours and day 1), whereas late wounds (day 7) show elevated neutrophil accumulation. In addition, granulation phase resolution is delayed, with persistent blood vessels and reduced dermal collagen at 10 days. The proinflammatory cytokine TNFα is elevated >3-fold in unwounded TSG-6-null skin and increases further after wounding (from 12 hours to 7 days) before returning to baseline by day 10. Other cytokines examined, such as IL-6, IL-10, and monocyte chemotactic protein-1, showed no consistent differences. Reintroduction of TSG-6 into TSG-6-null wounds rescues both the delay in wound closure and the aberrant neutrophil phenotype. In summary, our study indicates that TSG-6 plays an important role in regulating wound closure and inflammation during cutaneous wound repair.
Collapse
Affiliation(s)
- Sajina Shakya
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio, USA; Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Judith A Mack
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; Department of Dermatology, Dermatology & Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Minou Alipour
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio, USA; Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Edward V Maytin
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio, USA; Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; Department of Dermatology, Dermatology & Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
15
|
Broeren MGA, Di Ceglie I, Bennink MB, van Lent PLEM, van den Berg WB, Koenders MI, Blaney Davidson EN, van der Kraan PM, van de Loo FAJ. Treatment of collagenase-induced osteoarthritis with a viral vector encoding TSG-6 results in ectopic bone formation. PeerJ 2018; 6:e4771. [PMID: 29868252 PMCID: PMC5984587 DOI: 10.7717/peerj.4771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 04/24/2018] [Indexed: 12/22/2022] Open
Abstract
Objective Tumor necrosis factor-inducible gene 6 (TSG-6) has anti-inflammatory and chondroprotective effects in mouse models of inflammatory arthritis. Because cartilage damage and inflammation are also observed in osteoarthritis (OA), we determined the effect of viral overexpression of TSG-6 in experimental osteoarthritis. Methods Bone marrow-derived cells were differentiated to multinucleated osteoclasts in the presence of recombinant TSG-6 or after transduction with a lentiviral TSG-6 expression vector. Multi-nucleated osteoclasts were analyzed after tartrate resistant acid phosphatase staining and resorption activity was determined on dentin slices. Collagenase-induced osteoarthritis (CIOA) was induced in C57BL/6 mice after intra-articular injection of an adenoviral TSG-6 or control luciferase expression vector. Inflammation-related protease activity was measured using bioluminescent Prosense probes. After a second adenovirus injection, cartilage damage was assessed in histological sections stained with Safranin-O. Ectopic bone formation was scored in X-ray images of the affected knees. Results TSG-6 did not inhibit the formation of multi-nucleated osteoclasts, but caused a significant reduction in the resorption activity on dentin slices. Adenoviral TSG-6 gene therapy in CIOA could not reduce the cartilage damage compared to the luciferase control virus and no significant difference in inflammation-related protease activity was noted between the TSG-6 and control treated group. Instead, X-ray analysis and histological analysis revealed the presence of ectopic bone formation in the TSG-6 treated group. Conclusion Gene therapy based on the expression of TSG-6 could not provide cartilage protection in experimental osteoarthritis, but instead resulted in increased ectopic bone formation.
Collapse
Affiliation(s)
- Mathijs G A Broeren
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Irene Di Ceglie
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Miranda B Bennink
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wim B van den Berg
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije I Koenders
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fons A J van de Loo
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Watanabe R, Sato Y, Ozawa N, Takahashi Y, Koba S, Watanabe T. Emerging Roles of Tumor Necrosis Factor-Stimulated Gene-6 in the Pathophysiology and Treatment of Atherosclerosis. Int J Mol Sci 2018; 19:E465. [PMID: 29401724 PMCID: PMC5855687 DOI: 10.3390/ijms19020465] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/22/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor-stimulated gene-6 (TSG-6) is a 35-kDa glycoprotein that has been shown to exert anti-inflammatory effects in experimental models of arthritis, acute myocardial infarction, and acute cerebral infarction. Several lines of evidence have shed light on the pathophysiological roles of TSG-6 in atherosclerosis. TSG-6 suppresses inflammatory responses of endothelial cells, neutrophils, and macrophages as well as macrophage foam cell formation and vascular smooth muscle cell (VSMC) migration and proliferation. Exogenous TSG-6 infusion and endogenous TSG-6 attenuation with a neutralizing antibody for four weeks retards and accelerates, respectively, the development of aortic atherosclerotic lesions in ApoE-deficient mice. TSG-6 also decreases the macrophage/VSMC ratio (a marker of plaque instability) and promotes collagen fibers in atheromatous plaques. In patients with coronary artery disease (CAD), plasma TSG-6 levels are increased and TSG-6 is abundantly expressed in the fibrous cap within coronary atheromatous plaques, indicating that TSG-6 increases to counteract the progression of atherosclerosis and stabilize the plaque. These findings indicate that endogenous TSG-6 enhancement and exogenous TSG-6 replacement treatments are expected to emerge as new lines of therapy against atherosclerosis and related CAD. Therefore, this review provides support for the clinical utility of TSG-6 in the diagnosis and treatment of atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Yuki Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Nana Ozawa
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Yui Takahashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan.
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0392, Japan.
| |
Collapse
|
17
|
TSG-6 - a double-edged sword for osteoarthritis (OA). Osteoarthritis Cartilage 2018; 26:245-254. [PMID: 29129649 PMCID: PMC5807166 DOI: 10.1016/j.joca.2017.10.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/20/2017] [Accepted: 10/31/2017] [Indexed: 02/02/2023]
Abstract
PURPOSE To explore mechanisms underlying the association of TSG-6 with osteoarthritis (OA) progression. METHODS TSG-6-mediated heavy chain (HC) transfer (TSG-6 activity) and its association with inflammatory mediators were quantified in knee OA (n=25) synovial fluids (SFs). Paired intact and damaged cartilages from the same individuals (20 tibial and 12 meniscal) were analyzed by qRT-PCR and immunohistochemistry (IHC) for gene and protein expression of TSG-6 and components of Inter-alpha-Inhibitor (IαI) and TSG-6 activity ± spiked in IαI. Primary chondrocyte cultures (n=5) ± IL1β or TNFα were evaluated for gene expression. The effects of TSG-6 activity on cartilage extracellular matrix (ECM) assembly were explored using quantitative hyaluronan (HA)-aggrecan binding assays. RESULTS TSG-6 activity was significantly associated (R > 0.683, P < 0.0002) with inflammatory mediators including TIMP-1, A2M, MMP3, VEGF, VCAM-1, ICAM-1 and IL-6. Although TSG-6 protein and mRNA were highly expressed in damaged articular and meniscal cartilage and cytokine-treated chondrocytes, there was little or no cartilage expression of components of the IαI complex (containing HC1). By IHC, TSG-6 was present throughout lesioned cartilage but HC1 only at lesioned surfaces. TSG-6 impaired HA-aggrecan assembly, but TSG-6 mediated HA-HC formation reduced this negative effect. CONCLUSIONS TSG-6 activity is a global inflammatory biomarker in knee OA SF. IαI, supplied from outside cartilage, only penetrates the cartilage surface, restricting TSG-6 activity (HC transfer) to this region. Therefore, unopposed TSG-6 in intermediate and deep regions of OA cartilage could possibly block matrix assembly, leading to futile synthesis and account for increased risk of OA progression.
Collapse
|
18
|
Day AJ, Milner CM. TSG-6: A multifunctional protein with anti-inflammatory and tissue-protective properties. Matrix Biol 2018; 78-79:60-83. [PMID: 29362135 DOI: 10.1016/j.matbio.2018.01.011] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023]
Abstract
Tumor necrosis factor- (TNF) stimulated gene-6 (TSG-6) is an inflammation-associated secreted protein that has been implicated as having important and diverse tissue protective and anti-inflammatory properties, e.g. mediating many of the immunomodulatory and beneficial activities of mesenchymal stem/stromal cells. TSG-6 is constitutively expressed in some tissues, which are either highly metabolically active or subject to challenges from the environment, perhaps providing protection in these contexts. The diversity of its functions are dependent on the binding of TSG-6 to numerous ligands, including matrix molecules such as glycosaminoglycans, as well as immune regulators and growth factors that themselves interact with these linear polysaccharides. It is becoming apparent that TSG-6 can directly affect matrix structure and modulate the way extracellular signalling molecules interact with matrix. In this review, we focus mainly on the literature for TSG-6 over the last 10 years, summarizing its expression, structure, ligand-binding properties, biological functions and highlighting TSG-6's potential as a therapeutic for a broad range of disease indications.
Collapse
Affiliation(s)
- Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| | - Caroline M Milner
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
19
|
Zhang C, Zhang B, Wang H, Tao Q, Ge S, Zhai Z. Tumor necrosis factor alpha-stimulated gene-6 (TSG-6) inhibits the inflammatory response by inhibiting the activation of P38 and JNK signaling pathway and decreases the restenosis of vein grafts in rats. Heart Vessels 2017; 32:1536-1545. [DOI: 10.1007/s00380-017-1059-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022]
|
20
|
Liu HM, Liu YT, Zhang J, Ma LJ. Bone marrow mesenchymal stem cells ameliorate lung injury through anti-inflammatory and antibacterial effect in COPD mice. ACTA ACUST UNITED AC 2017; 37:496-504. [PMID: 28786060 DOI: 10.1007/s11596-017-1763-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 06/01/2017] [Indexed: 01/08/2023]
Abstract
The anti-inflammatory and antibacterial mechanisms of bone marrow mesenchymal stem cells (MSCs) ameliorating lung injury in chronic obstructive pulmonary disease (COPD) mice induced by cigarette smoke and Haemophilus Parainfluenza (HPi) were studied. The experiment was divided into four groups in vivo: control group, COPD group, COPD+HPi group, and COPD+HPi+MSCs group. The indexes of emphysematous changes, inflammatory reaction and lung injury score, and antibacterial effects were evaluated in all groups. As compared with control group, emphysematous changes were significantly aggravated in COPD group, COPD+HPi group and COPD+HPi+MSCs group (P<0.01), the expression of necrosis factor-kappaB (NF-κB) signal pathway and proinflammatory cytokines in bronchoalveolar lavage fluid (BALF) were increased (P<0.01), and the phagocytic activity of alveolar macrophages was downregulated (P<0.01). As compared with COPD group, lung injury score, inflammatory cells and proinflammatory cytokines were significantly increased in the BALF of COPD+HPi group and COPD+HPi+MSCs group (P<0.01). As compared with COPD+HPi group, the expression of tumor necrosis factor-α stimulated protein/gene 6 (TSG-6) was increased, the NF-κB signal pathway was depressed, proinflammatory cytokine was significantly reduced, the anti-inflammatory cytokine IL-10 was increased, and lung injury score was significantly reduced in COPD+HPi+MSCs group. Meanwhile, the phagocytic activity of alveolar macrophages was significantly enhanced and bacterial counts in the lung were decreased. The results indicated cigarette smoke caused emphysematous changes in mice and the phagocytic activity of alveolar macrophages was decreased. The lung injury of acute exacerbation of COPD mice induced by cigarette smoke and HPi was alleviated through MSCs transplantation, which may be attributed to the fact that MSCs could promote macrophages into anti-inflammatory phenotype through secreting TSG-6, inhibit NF-кB signaling pathway, and reduce inflammatory response through reducing proinflammatory cytokines and promoting the expression of the anti-inflammatory cytokine. Simultaneously, MSCs could enhance phagocytic activity of macrophages and bacterial clearance. Meanwhile, we detected anti-inflammatory and antibacterial activity of macrophages regulated by MSCs in vitro. As compared with RAW264.7+HPi+CSE group, the expression of NF-кB p65, IL-1β, IL-6 and TNF-α was significantly reduced, and the phagocytic activity of macrophages was significantly increased in RAW264.7+HPi+CSE+MSCs group (P<0.01). The result indicated the macrophages co-cultured with MSCs may inhibit NF-кB signaling pathway and promote phagocytosis by paracrine mechanism.
Collapse
Affiliation(s)
- Hong-Mei Liu
- Department of Respiratory Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, China.
| | - Yi-Tong Liu
- Department of Respiratory Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - Jing Zhang
- Department of Respiratory Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - Li-Jun Ma
- Department of Respiratory Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| |
Collapse
|
21
|
TNFα-stimulated gene-6 (TSG6) activates macrophage phenotype transition to prevent inflammatory lung injury. Proc Natl Acad Sci U S A 2016; 113:E8151-E8158. [PMID: 27911817 DOI: 10.1073/pnas.1614935113] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
TNFα-stimulated gene-6 (TSG6), a 30-kDa protein generated by activated macrophages, modulates inflammation; however, its mechanism of action and role in the activation of macrophages are not fully understood. Here we observed markedly augmented LPS-induced inflammatory lung injury and mortality in TSG6-/- mice compared with WT (TSG6+/+) mice. Treatment of mice with intratracheal instillation of TSG6 prevented LPS-induced lung injury and neutrophil sequestration, and increased survival in mice. We found that TSG6 inhibited the association of TLR4 with MyD88, thereby suppressing NF-κB activation. TSG6 also prevented the expression of proinflammatory proteins (iNOS, IL-6, TNFα, IL-1β, and CXCL1) while increasing the expression of anti-inflammatory proteins (CD206, Chi3l3, IL-4, and IL-10) in macrophages. This shift was associated with suppressed activation of proinflammatory transcription factors STAT1 and STAT3. In addition, we observed that LPS itself up-regulated the expression of TSG6 in TSG6+/+ mice, suggesting an autocrine role for TSG6 in transitioning macrophages. Thus, TSG6 functions by converting macrophages from a proinflammatory to an anti-inflammatory phenotype secondary to suppression of TLR4/NF-κB signaling and STAT1 and STAT3 activation.
Collapse
|
22
|
Atheroprotective Effects of Tumor Necrosis Factor-Stimulated Gene-6. JACC Basic Transl Sci 2016; 1:494-509. [PMID: 30167534 PMCID: PMC6113406 DOI: 10.1016/j.jacbts.2016.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/13/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor-stimulated gene-6 (TSG-6), an anti-inflammatory protein, was shown to be localized in the neointima of injury-induced rat arteries. However, the modulatory effect of TSG-6 on atherogenesis has not yet been reported. We aimed to evaluate the atheroprotective effects of TSG-6 on human endothelial cells (HECs), human monocyte-derived macrophages (HMDMs), human aortic smooth muscle cells (HASMCs) in vitro, and aortic lesions in apolipoprotein E-deficient mice, along with expression levels of TSG-6 in coronary lesions and plasma from patients with coronary artery disease (CAD). TSG-6 was abundantly expressed in HECs, HMDMs, and HASMCs in vitro. TSG-6 significantly suppressed cell proliferation and lipopolysaccharide-induced up-regulation of monocyte chemotactic protein-1, intercellular adhesion molecule-1, and vascular adhesion molecule-1 in HECs. TSG-6 significantly suppressed inflammatory M1 phenotype and suppressed oxidized low-density lipoprotein-induced foam cell formation associated with down-regulation of CD36 and acyl-CoA:cholesterol acyltransferase-1 in HMDMs. In HASMCs, TSG-6 significantly suppressed migration and proliferation, but increased collagen-1 and -3 expressions. Four-week infusion of TSG-6 into apolipoprotein E-deficient mice significantly retarded the development of aortic atherosclerotic lesions with decreased vascular inflammation, monocyte/macrophage, and SMC contents and increased collagen fibers. In addition, it decreased peritoneal M1 macrophages with down-regulation of inflammatory molecules and lowered plasma total cholesterol levels. In patients with CAD, plasma TSG-6 levels were significantly increased, and TSG-6 was highly expressed in the fibrous cap within coronary atherosclerotic plaques. These results suggest that TSG-6 contributes to the prevention and stability of atherosclerotic plaques. Thus, TSG-6 may serve as a novel therapeutic target for CAD.
Collapse
Key Words
- ABCA1, ATP-binding cassette transporter A1
- ACAT1, acyl-CoA:cholesterol acyltransferase-1
- AngII, angiotensin II
- ApoE−/−, apolipoprotein E deficient
- CAD, coronary artery disease
- ECM, extracellular matrix
- HASMC, human aortic smooth muscle cell
- HMDM, human monocyte-derived macrophage
- HUVEC, human umbilical vein endothelial cell
- MMP, matrix metalloproteinase
- TIMP, tissue inhibitor of metalloproteinase
- TSG, tumor necrosis factor stimulated gene
- TSG-6
- VSMC, vascular smooth muscle cell
- atherosclerosis
- coronary artery disease
- endothelial cell
- macrophage
- oxLDL, oxidized low-density lipoprotein
- vascular smooth muscle cell
Collapse
|
23
|
Liu L, Song H, Duan H, Chai J, Yang J, Li X, Yu Y, Zhang X, Hu X, Xiao M, Feng R, Yin H, Hu Q, Yang L, Du J, Li T. TSG-6 secreted by human umbilical cord-MSCs attenuates severe burn-induced excessive inflammation via inhibiting activations of P38 and JNK signaling. Sci Rep 2016; 6:30121. [PMID: 27444207 PMCID: PMC4957124 DOI: 10.1038/srep30121] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/29/2016] [Indexed: 02/06/2023] Open
Abstract
The hMSCs have become a promising approach for inflammation treatment in acute phase. Our previous study has demonstrated that human umbilical cord-MSCs could alleviate the inflammatory reaction of severely burned wound. In this study, we further investigated the potential role and mechanism of the MSCs on severe burn-induced excessive inflammation. Wistar rats were randomly divided into following groups: Sham, Burn, Burn+MSCs, Burn+MAPKs inhibitors, and Burn, Burn+MSCs, Burn+Vehicle, Burn+siTSG-6, Burn+rhTSG-6 in the both experiments. It was found that MSCs could only down-regulate P38 and JNK signaling, but had no effect on ERK in peritoneal macrophages of severe burn rats. Furthermore, suppression of P38 and JNK activations significantly reduced the excessive inflammation induced by severe burn. TSG-6 was secreted by MSCs using different inflammatory mediators. TSG-6 from MSCs and recombinant human (rh)TSG-6 all significantly reduced activations of P38 and JNK signaling induced by severe burn and then attenuated excessive inflammations. On the contrary, knockdown TSG-6 in the cells significantly increased phosphorylation of P38 and JNK signaling and reduced therapeutic effect of the MSCs on excessive inflammation. Taken together, this study suggested TSG-6 from MSCs attenuated severe burn-induced excessive inflammation via inhibiting activation of P38 and JNK signaling.
Collapse
Affiliation(s)
- Lingying Liu
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Huifeng Song
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Hongjie Duan
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Jiake Chai
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Jing Yang
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Xiao Li
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Yonghui Yu
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Xulong Zhang
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Xiaohong Hu
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Mengjing Xiao
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Rui Feng
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Huinan Yin
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Quan Hu
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Longlong Yang
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Jundong Du
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| | - Tianran Li
- Department of Burn &Plastic Surgery, the First Affiliated Hospital to PLA General Hospital
| |
Collapse
|
24
|
Jiang D, Muschhammer J, Qi Y, Kügler A, de Vries JC, Saffarzadeh M, Sindrilaru A, Beken SV, Wlaschek M, Kluth MA, Ganss C, Frank NY, Frank MH, Preissner KT, Scharffetter-Kochanek K. Suppression of Neutrophil-Mediated Tissue Damage-A Novel Skill of Mesenchymal Stem Cells. Stem Cells 2016; 34:2393-406. [PMID: 27299700 PMCID: PMC5572139 DOI: 10.1002/stem.2417] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/18/2016] [Accepted: 04/29/2016] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are crucial for tissue homeostasis and regeneration. Though of prime interest, their potentially protective role on neutrophil-induced tissue damage, associated with high morbidity and mortality, has not been explored in sufficient detail. Here we report the therapeutic skill of MSCs to suppress unrestrained neutrophil activation and to attenuate severe tissue damage in a murine immune-complex mediated vasculitis model of unbalanced neutrophil activation. MSC-mediated neutrophil suppression was due to intercellular adhesion molecule 1-dependent engulfment of neutrophils by MSCs, decreasing overall neutrophil numbers. Similar to MSCs in their endogenous niche of murine and human vasculitis, therapeutically injected MSCs via upregulation of the extracellular superoxide dismutase (SOD3), reduced super-oxide anion concentrations and consequently prevented neutrophil death, neutrophil extracellular trap formation and spillage of matrix degrading neutrophil elastase, gelatinase and myeloperoxidase. SOD3-silenced MSCs did not exert tissue protective effects. Thus, MSCs hold substantial therapeutic promise to counteract tissue damage in conditions with unrestrained neutrophil activation.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Jana Muschhammer
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Yu Qi
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Andrea Kügler
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Juliane C de Vries
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Mona Saffarzadeh
- Department of Biochemistry, School of Medicine, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Anca Sindrilaru
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Seppe Vander Beken
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | | | | | - Natasha Y Frank
- Department of Medicine, Boston VA Healthcare System, West Roxbury, Massachusetts, USA.,Division of Genetics, Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Markus H Frank
- Division of Genetics, Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Transplant Research Program, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.,School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Klaus T Preissner
- Department of Biochemistry, School of Medicine, Justus-Liebig-University of Giessen, Giessen, Germany
| | | |
Collapse
|
25
|
Dyer DP, Salanga CL, Johns SC, Valdambrini E, Fuster MM, Milner CM, Day AJ, Handel TM. The Anti-inflammatory Protein TSG-6 Regulates Chemokine Function by Inhibiting Chemokine/Glycosaminoglycan Interactions. J Biol Chem 2016; 291:12627-12640. [PMID: 27044744 PMCID: PMC4933465 DOI: 10.1074/jbc.m116.720953] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Indexed: 12/14/2022] Open
Abstract
TNF-stimulated gene-6 (TSG-6) is a multifunctional protein secreted in response to pro-inflammatory stimuli by a wide range of cells, including neutrophils, monocytes, and endothelial cells. It has been shown to mediate anti-inflammatory and protective effects when administered in disease models, in part, by reducing neutrophil infiltration. Human TSG-6 inhibits neutrophil migration by binding CXCL8 through its Link module (Link_TSG6) and interfering with the presentation of CXCL8 on cell-surface glycosaminoglycans (GAGs), an interaction that is vital for the function of many chemokines. TSG-6 was also found to interact with chemokines CXCL11 and CCL5, suggesting the possibility that it may function as a broad specificity chemokine-binding protein, functionally similar to those encoded by viruses. This study was therefore undertaken to explore the ability of TSG-6 to regulate the function of other chemokines. Herein, we demonstrate that Link_TSG6 binds chemokines from both the CXC and CC families, including CXCL4, CXCL12, CCL2, CCL5, CCL7, CCL19, CCL21, and CCL27. We also show that the Link_TSG6-binding sites on chemokines overlap with chemokine GAG-binding sites, and that the affinities of Link_TSG6 for these chemokines (KD values 1–85 nm) broadly correlate with chemokine-GAG affinities. Link_TSG6 also inhibits chemokine presentation on endothelial cells not only through a direct interaction with chemokines but also by binding and therefore masking the availability of GAGs. Along with previous work, these findings suggest that TSG-6 functions as a pluripotent regulator of chemokines by modulating chemokine/GAG interactions, which may be a major mechanism by which TSG-6 produces its anti-inflammatory effects in vivo.
Collapse
Affiliation(s)
- Douglas P Dyer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0684; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, Scotland, United Kingdom
| | - Catherina L Salanga
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0684
| | - Scott C Johns
- Medical and Research Sections, Veterans Affairs San Diego Healthcare System, La Jolla, California 92093; Department of Medicine, Division of Pulmonary and Critical Care, University of California, San Diego, La Jolla, California 92093
| | - Elena Valdambrini
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom; Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Mark M Fuster
- Medical and Research Sections, Veterans Affairs San Diego Healthcare System, La Jolla, California 92093; Department of Medicine, Division of Pulmonary and Critical Care, University of California, San Diego, La Jolla, California 92093
| | - Caroline M Milner
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom.
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom; Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom.
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093-0684.
| |
Collapse
|
26
|
An X, Li L, Chen Y, Luo A, Ni Z, Liu J, Yuan Y, Shi M, Chen B, Long D, Cheng J, Lu Y. Mesenchymal Stem Cells Ameliorated Glucolipotoxicity in HUVECs through TSG-6. Int J Mol Sci 2016; 17:483. [PMID: 27043548 PMCID: PMC4848939 DOI: 10.3390/ijms17040483] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/22/2016] [Accepted: 03/25/2016] [Indexed: 02/05/2023] Open
Abstract
Glucolipotoxicity is one of the critical causal factors of diabetic complications. Whether mesenchymal stem cells (MSCs) have effects on glucolipotoxicity in human umbilical vein endothelial cells (HUVECs) and mechanisms involved are unclear. Thirty mM glucose plus 100 μM palmitic acid was used to induce glucolipotoxicity in HUVECs. MSCs and HUVECs were co-cultured at the ratio of 1:5 via Transwell system. The mRNA expressions of inflammatory factors were detected by RT-qPCR. The productions of reactive oxygen species (ROS), cell cycle and apoptosis were analyzed by flow cytometry. The tumor necrosis factor-α stimulated protein 6 (TSG-6) was knockdown in MSCs by RNA interference. High glucose and palmitic acid remarkably impaired cell viability and tube formation capacity, as well as increased the mRNA expression of inflammatory factors, ROS levels, and cell apoptosis in HUVECs. MSC co-cultivation ameliorated these detrimental effects in HUVECs, but no effect on ROS production. Moreover, TSG-6 was dramatically up-regulated by high glucose and fatty acid stimulation in both MSCs and HUVECs. TSG-6 knockdown partially abolished the protection mediated by MSCs. MSCs had protective effects on high glucose and palmitic acid induced glucolipotoxicity in HUVECs, and TSG-6 secreted by MSCs was likely to play an important role in this process.
Collapse
Affiliation(s)
- Xingxing An
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
- School of Biomedical Sciences, CHIRI Biosciences, Curtin University, GPO Box U1987, Perth, WA 6845, Australia.
| | - Ai Luo
- Sichuan Neo-Life Stem Cell Biotech Inc. Chengdu 610041, China.
| | - Zuyao Ni
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Meimei Shi
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bo Chen
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health; West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
27
|
Comparison of Topical Application of TSG-6, Cyclosporine, and Prednisolone for Treating Dry Eye. Cornea 2016; 35:536-42. [DOI: 10.1097/ico.0000000000000756] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
28
|
Beltran SR, Svoboda KKH, Kerns DG, Sheth A, Prockop DJ. Anti-inflammatory protein tumor necrosis factor-α-stimulated protein 6 (TSG-6) promotes early gingival wound healing: an in vivo study. J Periodontol 2016; 86:62-71. [PMID: 25269522 DOI: 10.1902/jop.2014.140187] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Human multipotent mesenchymal stromal cells (hMSCs) produce tumor necrosis factor (TNF)-α-stimulated protein 6 (TSG-6). TSG-6 modulates proinflammatory cytokine cascades and enhances tissue repair. This study tests the effects of recombinant human TSG-6 (rhTSG-6) on gingival wound healing within the first 2 days post-surgery. METHODS After gingival resection in 120 Sprague-Dawley rats, 2 µg rhTSG-6 in 5-µL phosphate-buffered saline (PBS) or the same volume of only PBS solution was injected into gingival tissue approximating the surgical wound. Control animals did not receive injections. Tissue biopsies and blood were collected at 1 to 2, 6 to 8, 24, and 48 hours post-surgery (n = 10 per group). Specimens were analyzed via histologic analysis and enzyme-linked immunosorbent assay (ELISA) for quantification and comparison of inflammatory markers interleukin (IL)-1β, IL-6, TNF-α, and myeloperoxidase (MPO). Wound photographs were taken for a double-masked clinical assessment at each time period. Weights were recorded for all animals pre- and post-surgery. RESULTS Animals injected with rhTSG-6 had significantly less severe clinical inflammation at 6 to 8 (P = 0.01228), 24 (P = 0.01675), and 48 (P = 0.0186) hours. Sham and control animals had more weight loss at 24 and 48 hours. Sham and control animals had more pronounced cellular infiltrate. rhTSG-6-treated animals had significantly less MPO (P = 0.027) at 24 hours and IL-1β (P = 0.027) at 24 and 48 hours. IL-6 showed a marginal significant difference at 6 to 8 hours, but there was no significant difference for TNF-α. CONCLUSION rhTSG-6 reduced postoperative gingival inflammation by reducing levels of proinflammatory cytokines and cellular infiltrate and may offer significant promise as an anti-inflammatory agent for gingival surgery.
Collapse
Affiliation(s)
- Stacy R Beltran
- Department of Periodontics, Texas A&M University Baylor College of Dentistry, Dallas, TX
| | | | | | | | | |
Collapse
|
29
|
Carceller MC, Guillén MI, Ferrándiz ML, Alcaraz MJ. Paracrine in vivo inhibitory effects of adipose tissue-derived mesenchymal stromal cells in the early stages of the acute inflammatory response. Cytotherapy 2016; 17:1230-9. [PMID: 26276006 DOI: 10.1016/j.jcyt.2015.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/13/2015] [Accepted: 06/01/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS Excessive or unresolved inflammation leads to tissue lesions. Adipose tissue-derived mesenchymal stromal cells (AMSCs) have shown protective effects that may be dependent on the modulation of inflammation by secreted factors. METHODS We used the zymosan-induced mouse air pouch model at two time points (4 h and 18 h) to evaluate the in vivo effects of AMSCs and their conditioned medium (CM) on key steps of the early inflammatory response. We assessed the effects of AMSCs and CM on leukocyte migration and myeloperoxidase activity. The levels of chemokines, cytokines and eicosanoids in exudates were measured by use of enzyme-linked immunoassay or radio-immunoassay. In addition, the expression of cyclooxygenase-2 and microsomal prostaglandin E synthase-1 (mPGES-1) was studied by use of Western blotting and the phosphorylation of p65 nuclear factor-κB (NF-κB) by immunofluorescence. RESULTS All inflammatory parameters were significantly reduced by CM and AMSCs to a similar extent at 4 h after zymosan injection with lower effects at 18 h. The observed inhibition of leukocyte migration was associated with reduced levels of chemokines and leukotriene B4. Interleukin-1β, interleukin-6, tumor necrosis factor-α and tumor necrosis factor-stimulated gene 6 levels were significantly decreased. The downregulation of mPGES-1 was associated with inhibition of prostaglandin E2 production. Our results suggest that these anti-inflammatory effects are related, in part, to the inhibition of NF-κB activation. CONCLUSIONS AMSCs dampen the early process of inflammation in the zymosan-induced mouse air pouch model through paracrine mechanisms. These results support the potential utility of these cells as a source of novel treatment approaches for inflammatory pathologies.
Collapse
Affiliation(s)
| | - María Isabel Guillén
- Department of Pharmacology and IDM, University of Valencia, Valencia, Spain; Department of Chemistry, Biochemistry and Molecular Biology, Cardenal Herrera-CEU University, Valencia, Spain
| | | | - María José Alcaraz
- Department of Pharmacology and IDM, University of Valencia, Valencia, Spain.
| |
Collapse
|
30
|
Liang J, Jiang D, Noble PW. Hyaluronan as a therapeutic target in human diseases. Adv Drug Deliv Rev 2016; 97:186-203. [PMID: 26541745 PMCID: PMC4753080 DOI: 10.1016/j.addr.2015.10.017] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/07/2023]
Abstract
Accumulation and turnover of extracellular matrix is a hallmark of tissue injury, repair and remodeling in human diseases. Hyaluronan is a major component of the extracellular matrix and plays an important role in regulating tissue injury and repair, and controlling disease outcomes. The function of hyaluronan depends on its size, location, and interactions with binding partners. While fragmented hyaluronan stimulates the expression of an array of genes by a variety of cell types regulating inflammatory responses and tissue repair, cell surface hyaluronan provides protection against tissue damage from the environment and promotes regeneration and repair. The interactions of hyaluronan and its binding proteins participate in the pathogenesis of many human diseases. Thus, targeting hyaluronan and its interactions with cells and proteins may provide new approaches to developing therapeutics for inflammatory and fibrosing diseases. This review focuses on the role of hyaluronan in biological and pathological processes, and as a potential therapeutic target in human diseases.
Collapse
Affiliation(s)
- Jiurong Liang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W Noble
- Department of Medicine and Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
31
|
Kim DK, Choi H, Nishida H, Oh JY, Gregory C, Lee RH, Yu JM, Watanabe J, An SY, Bartosh TJ, Prockop DJ. Scalable Production of a Multifunctional Protein (TSG-6) That Aggregates with Itself and the CHO Cells That Synthesize It. PLoS One 2016; 11:e0147553. [PMID: 26793973 PMCID: PMC4721919 DOI: 10.1371/journal.pone.0147553] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/05/2016] [Indexed: 01/11/2023] Open
Abstract
TNF-α stimulated gene/protein 6 (TNFAIP6/TSG-6) is a multifunctional protein that has a number of potential therapeutic applications. Experiments and clinical trials with TSG-6, however, have been limited by the technical difficulties of producing the recombinant protein. We prepared stable clones of CHO cells that expressed recombinant human TSG-6 (rhTSG-6) as a secreted glycoprotein. Paradoxically, both cell number and protein production decreased dramatically when the clones were expanded. The decreases occurred because the protein aggregated the synthesizing CHO cells by binding to the brush border of hyaluronan that is found around many cultured cells. In addition, the rhTSG-6 readily self-aggregated. To address these problems, we added to the medium an inhibitor of hyaluronan synthesis and heparin to compete with the binding of TSG-6 to hyaluronan. Also, we optimized the composition of the culture medium, and transferred the CHO cells from a spinner culture system to a bioreactor that controlled pH and thereby decreased pH-dependent binding properties of the protein. With these and other improvements in the culture conditions, we obtained 57.0 mg ± 9.16 S.D. of rhTSG-6 in 5 or 6 liter of medium. The rhTSG-6 accounted for 18.0% ± 3.76 S.D. of the total protein in the medium. We then purified the protein with a Ni-chelate column that bound the His tag engineered into the C-terminus of the protein followed by an anion exchange column. The yield of the purified monomeric rhTSG-6 was 4.1 mg to 5.6 mg per liter of culture medium. After intravenous injection into mice, the protein had a longer plasma half-life than commercially available rhTSG-6 isolated from a mammalian cell lysate, apparently because it was recovered as a secreted glycoprotein. The bioactivity of the rhTSG-6 in suppressing inflammation was demonstrated in a murine model.
Collapse
Affiliation(s)
- Dong-Ki Kim
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Hosoon Choi
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Hidetaka Nishida
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Joo Youn Oh
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Carl Gregory
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Ji Min Yu
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Jun Watanabe
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Su Yeon An
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Thomas J. Bartosh
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
| | - Darwin J. Prockop
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott and White, Temple, Texas, United States of America
- * E-mail:
| |
Collapse
|
32
|
Wang H, Chen Z, Li XJ, Ma L, Tang YL. Anti-inflammatory cytokine TSG-6 inhibits hypertrophic scar formation in a rabbit ear model. Eur J Pharmacol 2015; 751:42-9. [PMID: 25661977 DOI: 10.1016/j.ejphar.2015.01.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 01/07/2015] [Accepted: 01/14/2015] [Indexed: 01/09/2023]
Abstract
Hypertrophic scars are characterized by excessive fibrosis and extracellular matrix (ECM) deposition and can be functionally and cosmetically problematic; however, there are few satisfactory treatments for controlling hypertrophic scars. The inflammatory cells and cytokines involved in excessive inflammation during wound healing facilitate fibroblast proliferation and collagen deposition, leading to pathologic scar formation. TSG-6 exhibits anti-inflammatory activity. This study examined the effect of recombinant TSG-6 on inflammation in hypertrophic scars using a rabbit ear model. Six 7-mm, full-thickness, circular wounds were made on the ears of 12 rabbits. TSG-6 and PBS were intradermally injected into the right and left ear wounds, respectively. The methods of TEM and TUNEL were used to detect fibroblast apoptosis. The expressions of inflammatory factors: IL-1β, IL-6 and TNF-α, were detected by immunohistochemistry and real time polymerase chain reaction. Collagen I and III expression detected by immunohistochemistry and Masson׳s trichrome staining and SEI (scar elevation index) was used to evaluate the extent of scarring. TSG-6 injection mitigated the formation of a hypertrophic scar in the rabbit ear. TSG-6-treated wounds exhibited decreased inflammation compared with the control group, as evidenced by the lower levels of IL-1β, IL-6, TNF-α and MPO. The SEI and the synthesis of collagens I and III were significantly decreased in the TSG-6-treated scars compared with control scars. The apoptosis rate was higher in the TSG-6-treated scars. TSG-6 exhibited anti-inflammatory effects during the wound healing process and cicatrization and significantly diminished hypertrophic scar formation in a rabbit ear model.
Collapse
Affiliation(s)
- Hui Wang
- Department of Plastic Surgery First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Zhao Chen
- Department of Plastic Surgery First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xiao-Jing Li
- Department of Plastic Surgery First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Li Ma
- Aesthetic Medicine Research Office, College of Nursing, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Yue-Ling Tang
- Department of Plastic Surgery First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
33
|
Abd-Allah SH, Shalaby SM, Abd-Elbary E, Saleh AA, El-Magd MA. Human peripheral blood CD34+ cells attenuate oleic acid-induced acute lung injury in rats. Cytotherapy 2014; 17:443-53. [PMID: 25536864 DOI: 10.1016/j.jcyt.2014.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 10/24/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND AIMS Adult stem cell-based therapy is a promising novel approach for treatment of acute lung injury (ALI). In this study, we evaluated the therapeutic effect of isolated human peripheral blood CD34+ progenitor cells in an ALI rat model, induced by oleic acid (OA) injection. METHODS Seventy-five adult female rats were used in this study. Group A, control without treatment, and group B, control injected with phosphate-buffered saline, comprised 15 rats each; the remaining 45 rats were injected with OA to induce ALI and were further subdivided into 3 groups: group C (ALI group, 15 rats), group D (ALI and fibroblast group, 15 rats) and group E (ALI and CD34+ cell group, 15 rats). RESULTS CD34+ cells transplantation in rats with OA-induced lung injury improves the arterial PaO(2) and wet/dry ratio, reduces infiltration of inflammatory cells and decreases lung vascular permeability as determined by reduced intra-alveolar and interstitial patchy congestion and hemorrhage as well as decreased interstitial edema. Additionally, lung inflammation determined by expression of the pro-inflammatory mediators intercellular adhesion molecule 1 and tumor necrosis factor-α was attenuated in CD34+ cell-treated rats at 6, 24 and 48 h post-OA challenge compared with non-treated rats. Moreover, the expression of anti-inflammatory molecule interleukin-10 was up-regulated in the lung of OA-induced ALI rats after administration of CD34+ cells. The important finding was that human TNF-α-induced protein 6 (TSG-6) gene expression was significantly up-regulated in rats treated with CD34+ cells. CONCLUSIONS The freshly isolated human peripheral blood-derived CD34+ cells may be used as an important source of stem cells that improve ALI. The anti-inflammatory properties of CD34+ cells in the lung are explained, at least in part, by activation of CD34+ cells to express TSG-6.
Collapse
Affiliation(s)
- Somia H Abd-Allah
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Sally M Shalaby
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman Abd-Elbary
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ayman A Saleh
- Department of Animal Wealth Development, Genetics and Genetic Engineering, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Mohammed Abu El-Magd
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt.
| |
Collapse
|
34
|
Lauer ME, Hascall VC, Green DE, DeAngelis PL, Calabro A. Irreversible heavy chain transfer to chondroitin. J Biol Chem 2014; 289:29171-9. [PMID: 25135638 DOI: 10.1074/jbc.m114.600809] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have recently demonstrated that the transfer of heavy chains (HCs) from inter-α-inhibitor, via the enzyme TSG-6 (tumor necrosis factor-stimulated gene 6), to hyaluronan (HA) oligosaccharides is an irreversible event in which subsequent swapping of HCs between HA molecules does not occur. We now describe our results of HC transfer experiments to chondroitin sulfate A, chemically desulfated chondroitin, chemoenzymatically synthesized chondroitin, unsulfated heparosan, heparan sulfate, and alginate. Of these potential HC acceptors, only chemically desulfated chondroitin and chemoenzymatically synthesized chondroitin were HC acceptors. The kinetics of HC transfer to chondroitin was similar to HA. At earlier time points, HCs were more widely distributed among the different sizes of chondroitin chains. As time progressed, the HCs migrated to lower molecular weight chains of chondroitin. Our interpretation is that TSG-6 swaps the HCs from the larger, reversible sites on chondroitin chains, which function as HC acceptors, onto smaller chondroitin chains, which function as irreversible HC acceptors. HCs transferred to smaller chondroitin chains were unable to be swapped off the smaller chondroitin chains and transferred to HA. HCs transferred to high molecular weight HA were unable to be swapped onto chondroitin. We also present data that although chondroitin was a HC acceptor, HA was the preferred acceptor when chondroitin and HA were in the same reaction mixture.
Collapse
Affiliation(s)
- Mark E Lauer
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195 and
| | - Vincent C Hascall
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195 and
| | - Dixy E Green
- the Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126
| | - Paul L DeAngelis
- the Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126
| | - Anthony Calabro
- From the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195 and
| |
Collapse
|
35
|
Prockop DJ. Concise review: two negative feedback loops place mesenchymal stem/stromal cells at the center of early regulators of inflammation. Stem Cells 2014; 31:2042-6. [PMID: 23681848 DOI: 10.1002/stem.1400] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 08/29/2012] [Indexed: 12/29/2022]
Abstract
Recent data demonstrated that MSCs can be activated by proinflammatory signals to introduce two negative feedback loops into the generic pathway of inflammation. In one loop, the activated MSCs secrete PGE2 that drives resident macrophages with an M1 proinflammatory phenotype toward an M2 anti-inflammatory phenotype. In the second loop, the activated MSCs secrete TSG-6 that interacts with CD44 on resident macrophages to decrease TLR2/NFκ-B signaling and thereby decrease the secretion of proinflammatory mediators of inflammation. The PGE2 and TSG-6 negative feedback loops allow MSCs to serve as regulators of the very early phases of inflammation. These and many related observations suggest that the MSC-like cells found in most tissues may be part of the pantheon of cells that protect us from foreign invaders, tissue injury, and aging.
Collapse
Affiliation(s)
- Darwin J Prockop
- Texas A&M Health Science Center College of Medicine Institute for Regenerative Medicine, Scott & White, Temple, Texas, USA
| |
Collapse
|
36
|
Higman VA, Briggs DC, Mahoney DJ, Blundell CD, Sattelle BM, Dyer DP, Green DE, DeAngelis PL, Almond A, Milner CM, Day AJ. A refined model for the TSG-6 link module in complex with hyaluronan: use of defined oligosaccharides to probe structure and function. J Biol Chem 2014; 289:5619-34. [PMID: 24403066 PMCID: PMC3937638 DOI: 10.1074/jbc.m113.542357] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Indexed: 11/25/2022] Open
Abstract
Tumor necrosis factor-stimulated gene-6 (TSG-6) is an inflammation-associated hyaluronan (HA)-binding protein that contributes to remodeling of HA-rich extracellular matrices during inflammatory processes and ovulation. The HA-binding domain of TSG-6 consists solely of a Link module, making it a prototypical member of the superfamily of proteins that interacts with this high molecular weight polysaccharide composed of repeating disaccharides of D-glucuronic acid and N-acetyl-D-glucosamine (GlcNAc). Previously we modeled a complex of the TSG-6 Link module in association with an HA octasaccharide based on the structure of the domain in its HA-bound conformation. Here we have generated a refined model for a HA/Link module complex using novel restraints identified from NMR spectroscopy of the protein in the presence of 10 distinct HA oligosaccharides (from 4- to 8-mers); the model was then tested using unique sugar reagents, i.e. chondroitin/HA hybrid oligomers and an octasaccharide in which a single sugar ring was (13)C-labeled. The HA chain was found to make more extensive contacts with the TSG-6 surface than thought previously, such that a D-glucuronic acid ring makes stacking and ionic interactions with a histidine and lysine, respectively. Importantly, this causes the HA to bend around two faces of the Link module (resembling the way that HA binds to CD44), potentially providing a mechanism for how TSG-6 can reorganize HA during inflammation. However, the HA-binding site defined here may not play a role in TSG-6-mediated transfer of heavy chains from inter-α-inhibitor onto HA, a process known to be essential for ovulation.
Collapse
Affiliation(s)
- Victoria A. Higman
- From the Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - David C. Briggs
- Wellcome Trust Centre for Cell Matrix Research
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT United Kingdom, and
| | - David J. Mahoney
- From the Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Charles D. Blundell
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT United Kingdom, and
| | - Benedict M. Sattelle
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT United Kingdom, and
| | - Douglas P. Dyer
- Wellcome Trust Centre for Cell Matrix Research
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT United Kingdom, and
| | - Dixy E. Green
- the Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Paul L. DeAngelis
- the Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Andrew Almond
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT United Kingdom, and
| | - Caroline M. Milner
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT United Kingdom, and
| | - Anthony J. Day
- Wellcome Trust Centre for Cell Matrix Research
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT United Kingdom, and
| |
Collapse
|
37
|
Dyer DP, Thomson JM, Hermant A, Jowitt TA, Handel TM, Proudfoot AEI, Day AJ, Milner CM. TSG-6 inhibits neutrophil migration via direct interaction with the chemokine CXCL8. THE JOURNAL OF IMMUNOLOGY 2014; 192:2177-85. [PMID: 24501198 DOI: 10.4049/jimmunol.1300194] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
TNF-stimulated gene/protein-6 (TSG-6) is expressed by many different cell types in response to proinflammatory cytokines and plays an important role in the protection of tissues from the damaging consequences of acute inflammation. Recently, TSG-6 was identified as being largely responsible for the beneficial effects of multipotent mesenchymal stem cells, for example in the treatment of animal models of myocardial infarction and corneal injury/allogenic transplant. The protective effect of TSG-6 is due in part to its inhibition of neutrophil migration, but the mechanisms underlying this activity remain unknown. In this study, we have shown that TSG-6 inhibits chemokine-stimulated transendothelial migration of neutrophils via a direct interaction (KD, ∼ 25 nM) between TSG-6 and the glycosaminoglycan binding site of CXCL8, which antagonizes the association of CXCL8 with heparin. Furthermore, we found that TSG-6 impairs the binding of CXCL8 to cell surface glycosaminoglycans and the transport of CXCL8 across an endothelial cell monolayer. In vivo this could limit the formation of haptotactic gradients on endothelial heparan sulfate proteoglycans and, hence, integrin-mediated tight adhesion and migration. We further observed that TSG-6 suppresses CXCL8-mediated chemotaxis of neutrophils; this lower potency effect might be important at sites where there is high local expression of TSG-6. Thus, we have identified TSG-6 as a CXCL8-binding protein, making it, to our knowledge, the first soluble mammalian chemokine-binding protein to be described to date. We have also revealed a potential mechanism whereby TSG-6 mediates its anti-inflammatory and protective effects. This could inform the development of new treatments for inflammation in the context of disease or following transplantation.
Collapse
Affiliation(s)
- Douglas P Dyer
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wisniewski HG, Colón E, Liublinska V, Karia RJ, Stabler TV, Attur M, Abramson SB, Band PA, Kraus VB. TSG-6 activity as a novel biomarker of progression in knee osteoarthritis. Osteoarthritis Cartilage 2014; 22:235-41. [PMID: 24333293 PMCID: PMC3939799 DOI: 10.1016/j.joca.2013.12.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/22/2013] [Accepted: 12/02/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To establish whether there is an association between TSG-6 activity and osteoarthritis progression. DESIGN TSG-6 activity was determined in 132 synovial fluids from patients with OA of the knee, using a novel quantitative TSG-6 activity assay. The association between TSG-6 activities at baseline and four distinct disease progression states, determined at 3-year follow-up, was analyzed using logistic regression. RESULTS There was a statistically significant relationship between TSG-6 activity at baseline and all OA progression states over a 3-year period. Patient knees with TSG-6 activities in the top tenth percentile, compared to the median activity, had an odds ratio (OR) of at least 7.86 (confidence interval (CI) [3.2, 20.5]) for total knee arthroplasty (TKA) within 3 years, and of at least 5.20 (CI [1.8, 13.9]) after adjustment for confounding factors. Receiver operating characteristic (ROC) analysis for knee arthroplasty yielded a cut-off point of 13.3 TSG-6 activity units/ml with the following parameters: area under the curve 0.90 (CI [0.804, 0.996]), sensitivity 0.91 (CI [0.59, 0.99]), specificity 0.82 (CI [0.74, 0.88]) and a negative predictive value (NPV) of 0.99 (CI [0.934, 0.994]). CONCLUSION The TSG-6 activity is a promising independent biomarker for OA progression. Given the high NPV, this assay may be particularly suitable for identifying patients at low risk of rapid disease progression and to assist in the timing of arthroplasty.
Collapse
Affiliation(s)
- Hans-Georg Wisniewski
- Department of Microbiology, New York University, School of Medicine, New York, NY 10016
| | - Elisa Colón
- Department of Microbiology, New York University, School of Medicine, New York, NY 10016
| | | | - Raj J. Karia
- Department of Orthopedic Surgery, New York University, School of Medicine, and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Thomas V. Stabler
- Department of Medicine, Duke University, School of Medicine, Durham, NC 27710
| | - Mukundan Attur
- Department of Medicine, New York University, School of Medicine, and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Steven B. Abramson
- Department of Medicine, New York University, School of Medicine, and NYU Hospital for Joint Diseases, New York, NY 10003
| | - Philip A. Band
- Department of Orthopedic Surgery, New York University, School of Medicine, and NYU Hospital for Joint Diseases, New York, NY 10003, and Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016
| | - Virginia B. Kraus
- Department of Medicine, Duke University, School of Medicine, Durham, NC 27710
| |
Collapse
|
39
|
Chong KW, Chanalaris A, Burleigh A, Jin H, Watt FE, Saklatvala J, Vincent TL. Fibroblast growth factor 2 drives changes in gene expression following injury to murine cartilage in vitro and in vivo. ACTA ACUST UNITED AC 2013; 65:2346-55. [PMID: 23740825 PMCID: PMC3992838 DOI: 10.1002/art.38039] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 05/23/2013] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The articular cartilage is known to be highly mechanosensitive, and a number of mechanosensing mechanisms have been proposed as mediators of the cellular responses to altered mechanical load. These pathways are likely to be important in tissue homeostasis as well as in the pathogenesis of osteoarthritis. One important injury-activated pathway involves the release of pericellular fibroblast growth factor 2 (FGF-2) from the articular cartilage. Using a novel model of murine cartilage injury and surgically destabilized joints in mice, we examined the extent to which FGF-2 contributes to the cellular gene response to injury. METHODS Femoral epiphyses from 5-week-old wild-type mice were avulsed and cultured in serum-free medium. Explant lysates were Western blotted for phospho-JNK, phospho-p38, and phospho-ERK or were fixed for immunohistochemical analysis of the nuclear translocation of p65 (indicative of NF-κB activation). RNA was extracted from injured explants, rested explants that had been stimulated with recombinant FGF-2 or FGF-18, or whole joints from either wild-type mice or FGF-2(-/-) mice. Reverse transcription-polymerase chain reaction was performed to examine a number of inflammatory response genes that had previously been identified in a microarray analysis. RESULTS Murine cartilage avulsion injury resulted in rapid activation of the 3 MAP kinase pathways as well as NF-κB. Almost all genes identified in murine joints following surgical destabilization were also regulated in cartilage explants upon injury. Many of these genes, including those for activin A (Inhba), tumor necrosis factor-stimulated gene 6 (Tnfaip6), matrix metalloproteinase 19 (Mmp19), tissue inhibitor of metalloproteinases 1 (Timp1), and podoplanin (Pdpn), were significantly FGF-2 dependent following injury to cartilage in vitro and to joint tissues in vivo. CONCLUSION FGF-2-dependent gene expression occurs in vitro and in vivo in response to cartilage/joint injury in mice.
Collapse
Affiliation(s)
- Ka-Wing Chong
- Kennedy Institute of Rheumatology and University of Oxford, London, UK
| | | | | | | | | | | | | |
Collapse
|
40
|
Capp E, Milner CM, Williams J, Hauck L, Jauckus J, Strowitzki T, Germeyer A. Modulation of tumor necrosis factor-stimulated gene-6 (TSG-6) expression in human endometrium. Arch Gynecol Obstet 2013; 289:893-901. [DOI: 10.1007/s00404-013-3080-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 10/29/2013] [Indexed: 01/09/2023]
|
41
|
Watanabe J, Shetty AK, Hattiangady B, Kim DK, Foraker JE, Nishida H, Prockop DJ. Administration of TSG-6 improves memory after traumatic brain injury in mice. Neurobiol Dis 2013; 59:86-99. [PMID: 23851308 DOI: 10.1016/j.nbd.2013.06.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/18/2013] [Accepted: 06/25/2013] [Indexed: 01/03/2023] Open
Abstract
Traumatic brain injury (TBI) causes multiple long-term defects including a loss of working memory that is frequently incapacitating. Administrations of mesenchymal stem/stromal cells (MSCs) previously produced beneficial effects in models of TBI as well as other disease models. In several models, the beneficial effects were explained by the MSCs being activated to express TSG-6, a multifunctional protein that modulates inflammation. In a mouse model of TBI, we found the initial mild phase of the inflammatory response persisted for at least 24h and was followed by secondary severe response that peaked at 3days. Intravenous human MSCs or TSG-6 during initial mild phase decreased neutrophil extravasation, expression of matrix metalloproteinase 9 by endothelial cells and neutrophils, and the subsequent blood brain barrier leakage in secondary phase. Administration of TSG-6 also decreased the lesion size at 2weeks. Importantly, the acute administration of TSG-6 within 24h of TBI was followed 6 to 10weeks later by improvements in memory, depressive-like behavior and the number of newly born-neurons. The data suggested that acute administration of TSG-6 may be an effective therapy for decreasing some of the long-term consequences of TBI.
Collapse
Affiliation(s)
- Jun Watanabe
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, TX 76502, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Winkler CW, Foster SC, Itakura A, Matsumoto SG, Asari A, McCarty OJT, Sherman LS. Hyaluronan oligosaccharides perturb lymphocyte slow rolling on brain vascular endothelial cells: implications for inflammatory demyelinating disease. Matrix Biol 2013; 32:160-8. [PMID: 23333375 DOI: 10.1016/j.matbio.2013.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/31/2012] [Accepted: 01/02/2013] [Indexed: 12/21/2022]
Abstract
Inflammatory demyelinating diseases like multiple sclerosis are characterized by mononuclear cell infiltration into the central nervous system. The glycosaminoglycan hyaluronan and its receptor, CD44, are implicated in the initiation and progression of a mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Digestion of hyaluronan tethered to brain vascular endothelial cells by a hyaluronidase blocks the slow rolling of lymphocytes along activated brain vascular endothelial cells and delays the onset of EAE. These effects could be due to the elimination of hyaluronan or the generation of hyaluronan digestion products that influence lymphocytes or endothelial cells. Here, we found that hyaluronan dodecasaccharides impaired activated lymphocyte slow rolling on brain vascular endothelial cells when applied to lymphocytes but not to the endothelial cells. The effects of hyaluronan dodecasaccharides on lymphocyte rolling were independent of CD44 and a receptor for degraded hyaluronan, Toll-like receptor-4. Subcutaneous injection of hyaluronan dodecasaccharides or tetrasaccharides delayed the onset of EAE in a manner similar to subcutaneous injection of hyaluronidase. Hyaluronan oligosaccharides can therefore act directly on lymphocytes to modulate the onset of inflammatory demyelinating disease.
Collapse
Affiliation(s)
- Clayton W Winkler
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Ave., Beaverton, OR 97006, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Wang N, Shao Y, Mei Y, Zhang L, Li Q, Li D, Shi S, Hong Q, Lin H, Chen X. Novel mechanism for mesenchymal stem cells in attenuating peritoneal adhesion: accumulating in the lung and secreting tumor necrosis factor α-stimulating gene-6. Stem Cell Res Ther 2012; 3:51. [PMID: 23217986 PMCID: PMC3580481 DOI: 10.1186/scrt142] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 12/04/2012] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION We previously found that mesenchymal stem cells (MSCs) injected intravenously could attenuate peritoneal adhesion by secreting tumor necrosis alpha-stimulating gene (TSG)-6, while MSCs injected intraperitoneally could not. However, the underlying mechanism remains unclear. This study was designed to investigate the means by which MSCs exert their effects. METHODS Rat bone marrow-derived MSCs/red fluorescent protein (RFP) were injected either intraperitoneally or intravenously into Sprague-Dawley (SD) rats at different time points after peritoneal scraping. Peritoneal adhesions were evaluated macroscopically at day 14 after scraping. The distribution of MSCs injected intraperitoneally or intravenously was traced by two-photon fluorescence confocal imaging and immunofluorescence microscopy. The co-localization of MSCs and macrophages in the lung and the spleen, and the expression of TSG-6 in MSCs trapped in the lung or the spleen were evaluated by immunofluorescence microscopy. The concentration of TSG-6 in serum was evaluated by ELISA. After intravenous injection of TSG-6- small interfering (si) RNA-MSCs, the expression of TSG-6 in MSCs and the concentration of TSG-6 in serum were reevaluated, and peritoneal adhesions were evaluated macroscopically and histologically. RESULTS MSCs injected intraperitoneally failed to reduce peritoneal adhesion, and MSCs injected intravenously markedly improved peritoneal adhesion. Two-photon fluorescence confocal imaging showed that MSCs injected intravenously accumulated mainly in the lung, where they remained for seven days, and immunofluorescence microscopy showed few MSCs phagocytosed by macrophages. In contrast, large numbers of MSCs accumulated in the spleen with obvious phagocytosis by macrophages even at 4 hours after intraperitoneal injection. Immunofluorescence microscopy showed that MSCs that accumulated in the lung after intravenous injection could express TSG-6 within 12 hours, but TSG-6-siRNA-MSCs or MSCs accumulated in the spleen after intraperitoneal injection did not. ELISA showed that the concentration of TSG-6 in serum was increased at 4 hours after intravenous injection of MSCs, while there was no increase after injection of TSG-6-siRNA-MSCs or after intraperitoneal injection of MSCs. Moreover, intravenous injection of TSG-6-siRNA-MSCs failed to attenuate peritoneal adhesion. CONCLUSIONS Our findings suggest that intravenously injected MSCs accumulated in the lung and attenuated peritoneal adhesion by secreting TSG-6, but intraperitoneally injected MSCs were phagocytosed by macrophages in the spleen and failed to attenuate peritoneal adhesion.
Collapse
|
44
|
Swaidani S, Cheng G, Lauer ME, Sharma M, Mikecz K, Hascall VC, Aronica MA. TSG-6 protein is crucial for the development of pulmonary hyaluronan deposition, eosinophilia, and airway hyperresponsiveness in a murine model of asthma. J Biol Chem 2012; 288:412-22. [PMID: 23118230 DOI: 10.1074/jbc.m112.389874] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hyaluronan (HA) deposition is often correlated with mucosal inflammatory responses, where HA mediates both protective and pathological responses. By modifying the HA matrix, Tnfip6 (TNF-α-induced protein-6; also known as TSG-6 (TNF-stimulated gene-6)) is thought to potentiate anti-inflammatory and anti-plasmin effects that are inhibitory to leukocyte extravasation. In this study, we examined the role of endogenous TSG-6 in the pathophysiological responses associated with acute allergic pulmonary inflammation. Compared with wild-type littermate controls, TSG-6(-/-) mice exhibited attenuated inflammation marked by a significant decrease in pulmonary HA concentrations measured in the bronchoalveolar lavage and lung tissue. Interestingly, despite the equivalent induction of both humoral and cellular Th2 immunity and the comparable levels of cytokines and chemokines typically associated with eosinophilic pulmonary inflammation, airway eosinophilia was significantly decreased in TSG-6(-/-) mice. Most importantly, contrary to their counterpart wild-type littermates, TSG-6(-/-) mice were resistant to the induction of airway hyperresponsiveness and manifested improved lung mechanics in response to methacholine challenge. Our study demonstrates that endogenous TSG-6 is dispensable for the induction of Th2 immunity but is essential for the robust increase in pulmonary HA deposition, propagation of acute eosinophilic pulmonary inflammation, and development of airway hyperresponsiveness. Thus, TSG-6 is implicated in the experimental murine model of allergic pulmonary inflammation and is likely to contribute to the pathogenesis of asthma.
Collapse
Affiliation(s)
- Shadi Swaidani
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Wang N, Li Q, Zhang L, Lin H, Hu J, Li D, Shi S, Cui S, Zhou J, Ji J, Wan J, Cai G, Chen X. Mesenchymal stem cells attenuate peritoneal injury through secretion of TSG-6. PLoS One 2012; 7:e43768. [PMID: 22912904 PMCID: PMC3422344 DOI: 10.1371/journal.pone.0043768] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 07/24/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mesothelial cell injury plays an important role in peritoneal fibrosis. Present clinical therapies aimed at alleviating peritoneal fibrosis have been largely inadequate. Mesenchymal stem cells (MSCs) are efficient for repairing injuries and reducing fibrosis. This study was designed to investigate the effects of MSCs on injured mesothelial cells and peritoneal fibrosis. METHODOLOGY/PRINCIPAL FINDINGS Rat bone marrow-derived MSCs (5 × 10(6)) were injected into Sprague-Dawley (SD) rats via tail vein 24 h after peritoneal scraping. Distinct reductions in adhesion formation; infiltration of neutrophils, macrophage cells; number of fibroblasts; and level of transforming growth factor (TGF)-β1 were found in MSCs-treated rats. The proliferation and repair of peritoneal mesothelial cells in MSCs-treated rats were stimulated. Mechanically injured mesothelial cells co-cultured with MSCs in transwells showed distinct increases in migration and proliferation. In vivo imaging showed that MSCs injected intravenously mainly accumulated in the lungs which persisted for at least seven days. No apparent MSCs were observed in the injured peritoneum even when MSCs were injected intraperitoneally. The injection of serum-starved MSCs-conditioned medium (CM) intravenously reduced adhesions similar to MSCs. Antibody based protein array of MSCs-CM showed that the releasing of TNFα-stimulating gene (TSG)-6 increased most dramatically. Promotion of mesothelial cell repair and reduction of peritoneal adhesion were produced by the administration of recombinant mouse (rm) TSG-6, and were weakened by TSG-6-RNA interfering. CONCLUSIONS/SIGNIFICANCE Collectively, these results indicate that MSCs may attenuate peritoneal injury by repairing mesothelial cells, reducing inflammation and fibrosis. Rather than the engraftment, the secretion of TSG-6 by MSCs makes a major contribution to the therapeutic benefits of MSCs.
Collapse
Affiliation(s)
- Nan Wang
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
- Medical College, NanKai University, Tianjin, China
| | - Qinggang Li
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Li Zhang
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Hongli Lin
- Department of Nephrology, the First Affiliated Hospital of Dalian Medical University, Liaoning, China
| | - Jie Hu
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Diangeng Li
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Suozhu Shi
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Shaoyuan Cui
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Jianhui Zhou
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Jiayao Ji
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
- Medical College, NanKai University, Tianjin, China
| | - Jiajia Wan
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
- Medical College, NanKai University, Tianjin, China
| | - Guangyan Cai
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
- * E-mail: (XC); (GC)
| | - Xiangmei Chen
- State Key Laboratory of Kidney Diseases, Department of Nephrology, PLA General Hospital and Military Medical Postgraduate College, Beijing, China
- * E-mail: (XC); (GC)
| |
Collapse
|
46
|
Hull RL, Johnson PY, Braun KR, Day AJ, Wight TN. Hyaluronan and hyaluronan binding proteins are normal components of mouse pancreatic islets and are differentially expressed by islet endocrine cell types. J Histochem Cytochem 2012; 60:749-60. [PMID: 22821669 PMCID: PMC3524560 DOI: 10.1369/0022155412457048] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The pancreatic islet comprises endocrine, vascular, and neuronal cells. Signaling among these cell types is critical for islet function. The extracellular matrix (ECM) is a key regulator of cell-cell signals, and while some islet ECM components have been identified, much remains unknown regarding its composition. We investigated whether hyaluronan, a common ECM component that may mediate inflammatory events, and molecules that bind hyaluronan such as versican, tumor necrosis factor-stimulated gene 6 (TSG-6), and components of inter-α-trypsin inhibitor (IαI), heavy chains 1 and 2 (ITIH1/ITIH2), and bikunin, are normally produced in the pancreatic islet. Mouse pancreata and isolated islets were obtained for microscopy (with both paraformaldehyde and Carnoy's fixation) and mRNA. Hyaluronan was present predominantly in the peri-islet ECM, and hyaluronan synthase isoforms 1 and 3 were also expressed in islets. Versican was produced in α cells; TSG-6 in α and β cells; bikunin in α, β, and δ cells; and ITIH1/ITIH2 predominantly in β cells. Our findings demonstrate that hyaluronan, versican, TSG-6, and IαI are normal islet components and that different islet endocrine cell types contribute these ECM components. Thus, dysfunction of either α or β cells likely alters islet ECM composition and could thereby further disrupt islet function.
Collapse
Affiliation(s)
- Rebecca L Hull
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, USA.
| | | | | | | | | |
Collapse
|
47
|
Burleigh A, Chanalaris A, Gardiner MD, Driscoll C, Boruc O, Saklatvala J, Vincent TL. Joint immobilization prevents murine osteoarthritis and reveals the highly mechanosensitive nature of protease expression in vivo. ACTA ACUST UNITED AC 2012; 64:2278-88. [DOI: 10.1002/art.34420] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
Chang MY, Chan CK, Braun KR, Green PS, O'Brien KD, Chait A, Day AJ, Wight TN. Monocyte-to-macrophage differentiation: synthesis and secretion of a complex extracellular matrix. J Biol Chem 2012; 287:14122-35. [PMID: 22351750 PMCID: PMC3340194 DOI: 10.1074/jbc.m111.324988] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Although monocyte- and macrophage-derived molecules are known to promote extracellular matrix (ECM) disruption and destabilization, it is less appreciated that they also synthesize molecules contributing to ECM formation, stabilization, and function. We have identified and characterized the synthesis of proteoglycans and related proteins, some not previously known to be associated with macrophages. Proteoglycan extracts of [35S]sulfate- and 35S-trans amino acid-radiolabeled culture media from THP-1 monocytes induced to differentiate by treatment with phorbol myristate acetate revealed three major proteins of ∼25, 90, and 100 kDa following chondroitin ABC lyase digestion. The 25-kDa protein was predominant for monocytes, whereas the 90- and 100-kDa proteins were predominant for macrophages. Tandem mass spectrometry identified (i) the 25-kDa core protein as serglycin, (ii) the 90-kDa core protein as inter-α-inhibitor heavy chain 2 (IαIHC2), and (iii) the 100-kDa core as amyloid precursor-like protein 2 (APLP2). Differentiation was also associated with (i) a >500-fold increase in mRNA for TNF-stimulated gene-6, an essential cofactor for heavy chain-mediated matrix stabilization; (ii) a >800-fold increase in mRNA for HAS2, which is responsible for hyaluronan synthesis; and (iii) a 3-fold increase in mRNA for versican, which interacts with hyaluronan. Biochemical evidence is also presented for an IαIHC2-APLP2 complex, and immunohistochemical staining of human atherosclerotic lesions demonstrates similar staining patterns for APLP2 and IαIHC2 with macrophages, whereas serglycin localizes to the underlying glycosaminoglycan-rich region. These findings indicate that macrophages synthesize many of the molecules participating in ECM formation and function, suggesting a novel role for these molecules in the differentiation of macrophages in the development of atherosclerosis.
Collapse
Affiliation(s)
- Mary Y Chang
- Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Roddy GW, Oh JY, Lee RH, Bartosh TJ, Ylostalo J, Coble K, Rosa RH, Prockop DJ. Action at a distance: systemically administered adult stem/progenitor cells (MSCs) reduce inflammatory damage to the cornea without engraftment and primarily by secretion of TNF-α stimulated gene/protein 6. Stem Cells 2012; 29:1572-9. [PMID: 21837654 DOI: 10.1002/stem.708] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous reports demonstrated that the deleterious effects of chemical injury to the cornea were ameliorated by local or systemic administration of adult stem/progenitor cells from bone marrow referred to as mesenchymal stem or stromal cells (MSCs). However, the mechanisms for the beneficial effects of MSCs on the injured cornea were not clarified. Herein, we demonstrated that human MSCs (hMSCs) were effective in reducing corneal opacity and inflammation without engraftment after either intraperitoneal (i.p.) or intravenous (i.v.) administration following chemical injury to the rat cornea. A quantitative assay for human mRNA for glyceraldehyde 3-phosphate dehydrogenase (GAPDH) demonstrated that less than 10 hMSCs were present in the corneas of rats 1-day and 3 days after i.v. or i.p. administration of 1 × 10(7) hMSCs. In vitro experiments using a transwell coculture system demonstrated that chemical injury to corneal epithelial cells activated hMSCs to secrete the multipotent anti-inflammatory protein TNF-α stimulated gene/protein 6 (TSG-6). In vivo, the effects of i.v. injection of hMSCs were largely abrogated by knockdown of TSG-6. Also, the effects of hMSCs were essentially duplicated by either i.v. or topical administration of TSG-6. Therefore, the results demonstrated that systemically administered hMSCs reduce inflammatory damage to the cornea without engraftment and primarily by secretion of the anti-inflammatory protein TSG-6 in response to injury signals from the cornea.
Collapse
Affiliation(s)
- Gavin W Roddy
- Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine at Scott & White, Temple, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Nagyeri G, Radacs M, Ghassemi-Nejad S, Tryniszewska B, Olasz K, Hutas G, Gyorfy Z, Hascall VC, Glant TT, Mikecz K. TSG-6 protein, a negative regulator of inflammatory arthritis, forms a ternary complex with murine mast cell tryptases and heparin. J Biol Chem 2011; 286:23559-69. [PMID: 21566135 DOI: 10.1074/jbc.m111.222026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TSG-6 (TNF-α-stimulated gene/protein 6), a hyaluronan (HA)-binding protein, has been implicated in the negative regulation of inflammatory tissue destruction. However, little is known about the tissue/cell-specific expression of TSG-6 in inflammatory processes, due to the lack of appropriate reagents for the detection of this protein in vivo. Here, we report on the development of a highly sensitive detection system and its use in cartilage proteoglycan (aggrecan)-induced arthritis, an autoimmune murine model of rheumatoid arthritis. We found significant correlation between serum concentrations of TSG-6 and arthritis severity throughout the disease process, making TSG-6 a better biomarker of inflammation than any of the other arthritis-related cytokines measured in this study. TSG-6 was present in arthritic joint tissue extracts together with the heavy chains of inter-α-inhibitor (IαI). Whereas TSG-6 was broadly detectable in arthritic synovial tissue, the highest level of TSG-6 was co-localized with tryptases in the heparin-containing secretory granules of mast cells. In vitro, TSG-6 formed complexes with the tryptases murine mast cell protease-6 and -7 via either heparin or HA. In vivo TSG-6-tryptase association could also be detected in arthritic joint extracts by co-immunoprecipitation. TSG-6 has been reported to suppress inflammatory tissue destruction by enhancing the serine protease-inhibitory activity of IαI against plasmin. TSG-6 achieves this by transferring heavy chains from IαI to HA, thus liberating the active bikunin subunit of IαI. Because bikunin is also present in mast cell granules, we propose that TSG-6 can promote inhibition of tryptase activity via a mechanism similar to inhibition of plasmin.
Collapse
Affiliation(s)
- Gyorgy Nagyeri
- Section of Molecular Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|