Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in context

Background: Bone destruction is one of many severe complications that occurs in patients with rheumatoid arthritis (RA) and current therapies are unable to cure this manifestation. This study here aims to determine whether GMSC can directly inhibit osteoclast formation and eventually attenuate osteo...

Full description

Bibliographic Details
Main Authors: Yang Luo, Wenbin Wu, Jian Gu, Ximei Zhang, Junlong Dang, Julie Wang, Yongjiang Zheng, Feng Huang, Jia Yuan, Youqiu Xue, Qingling Fu, Umadevi Kandalam, Jacob Colello, Song Guo Zheng
Format: Article
Language:English
Published: Elsevier 2019-05-01
Series:EBioMedicine
Online Access:http://www.sciencedirect.com/science/article/pii/S235239641930297X
id doaj-13ea17b51a3343598d4f34eec8f850fd
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Yang Luo
Wenbin Wu
Jian Gu
Ximei Zhang
Junlong Dang
Julie Wang
Yongjiang Zheng
Feng Huang
Jia Yuan
Youqiu Xue
Qingling Fu
Umadevi Kandalam
Jacob Colello
Song Guo Zheng
spellingShingle Yang Luo
Wenbin Wu
Jian Gu
Ximei Zhang
Junlong Dang
Julie Wang
Yongjiang Zheng
Feng Huang
Jia Yuan
Youqiu Xue
Qingling Fu
Umadevi Kandalam
Jacob Colello
Song Guo Zheng
Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in context
EBioMedicine
author_facet Yang Luo
Wenbin Wu
Jian Gu
Ximei Zhang
Junlong Dang
Julie Wang
Yongjiang Zheng
Feng Huang
Jia Yuan
Youqiu Xue
Qingling Fu
Umadevi Kandalam
Jacob Colello
Song Guo Zheng
author_sort Yang Luo
title Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in context
title_short Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in context
title_full Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in context
title_fullStr Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in context
title_full_unstemmed Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in context
title_sort human gingival tissue-derived msc suppress osteoclastogenesis and bone erosion via cd39-adenosine signal pathway in autoimmune arthritisresearch in context
publisher Elsevier
series EBioMedicine
issn 2352-3964
publishDate 2019-05-01
description Background: Bone destruction is one of many severe complications that occurs in patients with rheumatoid arthritis (RA) and current therapies are unable to cure this manifestation. This study here aims to determine whether GMSC can directly inhibit osteoclast formation and eventually attenuate osteoclastogenesis and bone erosion in an inflammatory milieu. Method: GMSC were co-cultured with osteoclast precursors with or without CD39 inhibitor, CD73 inhibitor or adenosine receptors inhibitors pretreatment and osteoclast formation were evaluated in vitro. 2×10^6 GMSC per mouse were transferred to CIA mice and pathology scores, the frequency of osteoclasts, bone erosion in joints were assessed in vivo. Finding: GMSC but not control cells, markedly suppressed human or mice osteoclastogenesis in vitro. GMSC treatment also resulted in a dramatically decreased level of NF-κB p65/p50 in osteoclasts in vitro. Infusion of GMSC to CIA significantly attenuated the severity of arthritis, pathology scores, frequency of osteoclasts, particularly bone erosion, as well as a decreased expression of RANKL in synovial tissues in vivo. Blockade of CD39/CD73 or adenosine receptors has significantly abrogated the suppressive ability of GMSC in vitro and therapeutic effect of GMSC on bone erosion during CIA in vivo. Interpretation: GMSC inhibit osteoclast formation in vitro and in vivo partially via CD39-CD73-adenosine signals. Manipulation of GMSC may have a therapeutic implication on rheumatoid arthritis and other bone erosion related diseases. Fund: This study was supported by grants from the National Key R&D Program of China (2017YFA0105801 to F.H); the Zhujiang Innovative and Entrepreneurial Talent Team Award of Guangdong Province (2016 ZT 06S 252 to F·H) and National Institutes of Health (R01 AR059103, R61 AR073409 and NIH Star Award to S.G.Z). Keywords: Rheumatoid arthritis, Mesenchymal stem cells, CD39, Adenosine
url http://www.sciencedirect.com/science/article/pii/S235239641930297X
work_keys_str_mv AT yangluo humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT wenbinwu humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT jiangu humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT ximeizhang humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT junlongdang humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT juliewang humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT yongjiangzheng humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT fenghuang humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT jiayuan humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT youqiuxue humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT qinglingfu humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT umadevikandalam humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT jacobcolello humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
AT songguozheng humangingivaltissuederivedmscsuppressosteoclastogenesisandboneerosionviacd39adenosinesignalpathwayinautoimmunearthritisresearchincontext
_version_ 1724976699452424192
spelling doaj-13ea17b51a3343598d4f34eec8f850fd2020-11-25T01:57:03ZengElsevierEBioMedicine2352-39642019-05-0143620631Human gingival tissue-derived MSC suppress osteoclastogenesis and bone erosion via CD39-adenosine signal pathway in autoimmune arthritisResearch in contextYang Luo0Wenbin Wu1Jian Gu2Ximei Zhang3Junlong Dang4Julie Wang5Yongjiang Zheng6Feng Huang7Jia Yuan8Youqiu Xue9Qingling Fu10Umadevi Kandalam11Jacob Colello12Song Guo Zheng13Department of Clinical Immunology, Sun Yat-sen University Third Affiliated Hospital, Guangzhou 510000, PR China; Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USA; Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR ChinaDepartment of Clinical Immunology, Sun Yat-sen University Third Affiliated Hospital, Guangzhou 510000, PR ChinaDivision of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USA; Division of Rheumatology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USADepartment of Clinical Immunology, Sun Yat-sen University Third Affiliated Hospital, Guangzhou 510000, PR China; Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USA; Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA 43201, USADepartment of Clinical Immunology, Sun Yat-sen University Third Affiliated Hospital, Guangzhou 510000, PR China; Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USADivision of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USA; Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA 43201, USADivision of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USADepartment of Clinical Immunology, Sun Yat-sen University Third Affiliated Hospital, Guangzhou 510000, PR ChinaDepartment of Clinical Immunology, Sun Yat-sen University Third Affiliated Hospital, Guangzhou 510000, PR ChinaDepartment of Clinical Immunology, Sun Yat-sen University Third Affiliated Hospital, Guangzhou 510000, PR China; Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USA; Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA 43201, USAOtorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong 510080, PR ChinaDepartment of Pediatric Dentistry, College of Dental Medicine, Nova Southeastern University, Davie, FL 33328, USADivision of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey 17033, USADivision of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA 43201, USA; Corresponding author at: Ohio State University College of Medicine and Wexner Medical Center, 480 Medical Center Dr, Columbus, OH 43201, USA.Background: Bone destruction is one of many severe complications that occurs in patients with rheumatoid arthritis (RA) and current therapies are unable to cure this manifestation. This study here aims to determine whether GMSC can directly inhibit osteoclast formation and eventually attenuate osteoclastogenesis and bone erosion in an inflammatory milieu. Method: GMSC were co-cultured with osteoclast precursors with or without CD39 inhibitor, CD73 inhibitor or adenosine receptors inhibitors pretreatment and osteoclast formation were evaluated in vitro. 2×10^6 GMSC per mouse were transferred to CIA mice and pathology scores, the frequency of osteoclasts, bone erosion in joints were assessed in vivo. Finding: GMSC but not control cells, markedly suppressed human or mice osteoclastogenesis in vitro. GMSC treatment also resulted in a dramatically decreased level of NF-κB p65/p50 in osteoclasts in vitro. Infusion of GMSC to CIA significantly attenuated the severity of arthritis, pathology scores, frequency of osteoclasts, particularly bone erosion, as well as a decreased expression of RANKL in synovial tissues in vivo. Blockade of CD39/CD73 or adenosine receptors has significantly abrogated the suppressive ability of GMSC in vitro and therapeutic effect of GMSC on bone erosion during CIA in vivo. Interpretation: GMSC inhibit osteoclast formation in vitro and in vivo partially via CD39-CD73-adenosine signals. Manipulation of GMSC may have a therapeutic implication on rheumatoid arthritis and other bone erosion related diseases. Fund: This study was supported by grants from the National Key R&D Program of China (2017YFA0105801 to F.H); the Zhujiang Innovative and Entrepreneurial Talent Team Award of Guangdong Province (2016 ZT 06S 252 to F·H) and National Institutes of Health (R01 AR059103, R61 AR073409 and NIH Star Award to S.G.Z). Keywords: Rheumatoid arthritis, Mesenchymal stem cells, CD39, Adenosinehttp://www.sciencedirect.com/science/article/pii/S235239641930297X