1
|
Hetta HF, Elsaghir A, Sijercic VC, Ahmed AK, Gad SA, Zeleke MS, Alanazi FE, Ramadan YN. Clinical Progress in Mesenchymal Stem Cell Therapy: A Focus on Rheumatic Diseases. Immun Inflamm Dis 2025; 13:e70189. [PMID: 40353645 PMCID: PMC12067559 DOI: 10.1002/iid3.70189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/10/2024] [Accepted: 03/21/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Rheumatic diseases are chronic immune-mediated disorders affecting multiple organ systems and significantly impairing patients' quality of life. Current treatments primarily provide symptomatic relief without offering a cure. Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic option due to their ability to differentiate into various cell types and their immunomodulatory, anti-inflammatory, and regenerative properties. This review aims to summarize the clinical progress of MSC therapy in rheumatic diseases, highlight key findings from preclinical and clinical studies, and discuss challenges and future directions. METHODOLOGY A comprehensive review of preclinical and clinical studies on MSC therapy in rheumatic diseases, including systemic lupus erythematosus, rheumatoid arthritis, ankylosing spondylitis, osteoarthritis, osteoporosis, Sjögren's syndrome, Crohn's disease, fibromyalgia, systemic sclerosis, dermatomyositis, and polymyositis, was conducted. Emerging strategies to enhance MSC efficacy and overcome current limitations were also analyzed. RESULTS AND DISCUSSION Evidence from preclinical and clinical studies suggests that MSC therapy can reduce inflammation, modulate immune responses, and promote tissue repair in various rheumatic diseases. Clinical trials have demonstrated potential benefits, including symptom relief and disease progression delay. However, challenges such as variability in treatment response, optimal cell source and dosing, long-term safety concerns, and regulatory hurdles remain significant barriers to clinical translation. Standardized protocols and further research are required to optimize MSC application. CONCLUSION MSC therapy holds promise for managing rheumatic diseases, offering potential disease-modifying effects beyond conventional treatments. However, large-scale, well-controlled clinical trials are essential to establish efficacy, safety, and long-term therapeutic potential. Addressing current limitations through optimized treatment protocols and regulatory frameworks will be key to its successful integration into clinical practice.
Collapse
Affiliation(s)
- Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of PharmacyUniversity of TabukTabukSaudi Arabia
| | - Alaa Elsaghir
- Department of Microbiology and Immunology, Faculty of PharmacyAssiut UniversityAssiutEgypt
| | | | - Abdulrahman K. Ahmed
- Emergency Medicine Unit, Department of Anaethesia and Intensive Care, Faculty of MedicineAssiut UniversityAssiutEgypt
| | - Sayed A. Gad
- Emergency Medicine Unit, Department of Anaethesia and Intensive Care, Faculty of MedicineAssiut UniversityAssiutEgypt
| | - Mahlet S. Zeleke
- Menelik II Medical and Health Science CollegeAddis AbabaEthiopia
| | - Fawaz E. Alanazi
- Department of Pharmacology and Toxicology, Faculty of PharmacyUniversity of TabukTabukSaudi Arabia
| | - Yasmin N. Ramadan
- Department of Microbiology and Immunology, Faculty of PharmacyAssiut UniversityAssiutEgypt
| |
Collapse
|
2
|
Wang X, Zhang Y, Xu C, Li Q, Ji P, Liu H, Zhang Y, Jin J, Yuan Z, Yuan M, Feng P, Wu Y, Liu W, Shen H, Wang P. MGP regulates the adipogenic differentiation of mesenchymal stem cells in osteoporosis via the Ca2+/CaMKII/RIP140/FABP3 axis. Cell Death Discov 2025; 11:166. [PMID: 40216750 PMCID: PMC11992250 DOI: 10.1038/s41420-025-02472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
The dysregulation of bone marrow mesenchymal stem cells (BM-MSCs) is crucial in the pathogenesis of osteoporosis, and adipogenic differentiation of BM-MSCs is considered an essential factor in this process. However, the mechanisms underlying the regulation of MSC adipogenic differentiation require further investigation. MGP (Matrix Gla Protein) was reported to impair the osteogenic differentiation. However, the mechanisms through which MGP regulates osteoporosis and bone-fat imbalance in MSCs are still unclear. In this study, we confirmed that the expression of MGP upregulated in osteoporosis and has a negative correlation with BMD (bone mineral density). Gain- and loss-of-function experiments were performed to ensure the role of MGP in MSC adipogenic differentiation. Mechanistically, MGP increased intracellular free Ca2+ levels and enhanced CaMKII phosphorylation, which in turn activated RIP140 protein degradation. This led to an increase in the transcription of FABP3, ultimately promoting adipogenic differentiation in MSCs. Furthermore, we demonstrated that using recombinant adeno-associated virus 9 (rAAV9) to silence MGP has the effect of alleviating bone loss and reversing the excessive bone marrow adipose tissue in mice with osteoporosis. In summary, our research has unveiled the regulatory role of MGP/Ca2+/CaMKII/RIP140/FABP3 axis in adipogenic differentiation in MSC and it might be a promising approach for osteoporosis treatment.
Collapse
Affiliation(s)
- Xinglang Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Yunhui Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Chenhao Xu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Quanfeng Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Pengfei Ji
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Huatao Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Yibin Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Jiahao Jin
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Zihao Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Miao Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Pei Feng
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China.
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China.
| |
Collapse
|
3
|
Liu WJ, Wang JX, Li QF, Zhang YH, Ji PF, Jin JH, Zhang YB, Yuan ZH, Feng P, Wu YF, Shen HY, Wang P. Fat mass and obesity-associated protein in mesenchymal stem cells inhibits osteoclastogenesis via lnc NORAD/miR-4284 axis in ankylosing spondylitis. World J Stem Cells 2025; 17:98911. [PMID: 40160686 PMCID: PMC11947893 DOI: 10.4252/wjsc.v17.i3.98911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/03/2025] [Accepted: 02/26/2025] [Indexed: 03/21/2025] Open
Abstract
BACKGROUND Ankylosing spondylitis (AS) is recognized as a long-term inflammatory disorder that leads to inflammation in the spine and joints, alongside abnormal bone growth. In previous studies, we reported that mesenchymal stem cells (MSCs) derived from individuals with AS demonstrated a remarkable inhibition in the formation of osteoclasts compared to those obtained from healthy donors. The mechanism through which MSCs from AS patients achieve this inhibition remains unclear. AIM To investigate the potential underlying mechanism by which MSCs from individuals with ankylosing spondylitis (AS-MSCs) inhibit osteoclastogenesis. METHODS We analysed fat mass and obesity-associated (FTO) protein levels in AS-MSCs and MSCs from healthy donors and investigated the effects and mechanism by which FTO in MSCs inhibits osteoclastogenesis by coculturing and measuring the levels of tartrate-resistant acid phosphatase, nuclear factor of activated T cells 1 and cathepsin K. RESULTS We found that FTO, an enzyme responsible for removing methyl groups from RNA, was more abundantly expressed in MSCs from AS patients than in those from healthy donors. Reducing FTO levels was shown to diminish the capacity of MSCs to inhibit osteoclast development. Further experimental results revealed that FTO affects the stability of the long non-coding RNA activated by DNA damage (NORAD) by altering its N6-methyladenosine methylation status. Deactivating NORAD in MSCs significantly increased osteoclast formation by affecting miR-4284, which could regulate the MSC-mediated inhibition of osteoclastogenesis reported in our previous research. CONCLUSION This study revealed elevated FTO levels in AS-MSCs and found that FTO regulated the ability of AS-MSCs to inhibit osteoclast formation through the long noncoding RNA NORAD/miR-4284 axis.
Collapse
Affiliation(s)
- Wen-Jie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Jia-Xin Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Quan-Feng Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yun-Hui Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Peng-Fei Ji
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Jia-Hao Jin
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yi-Bin Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Zi-Hao Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Pei Feng
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yan-Feng Wu
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Hui-Yong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China.
| |
Collapse
|
4
|
Wei Y, Zhang S, Shao F, Sun Y. Ankylosing spondylitis: From pathogenesis to therapy. Int Immunopharmacol 2025; 145:113709. [PMID: 39644789 DOI: 10.1016/j.intimp.2024.113709] [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: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Ankylosing spondylitis (AS) is an autoimmune rheumatic disease that primarily affects the axial joints, with its etiology complex and still not fully understood. The unknown pathogenesis of AS limits the development of treatment strategies, so keeping up-to-date with the current research on AS can help in searching for potential therapeutic targets. In addition to the classic HLA-B27 genetic susceptibility and Th17-related inflammatory signals, increasing research is focusing on the influence of autoantigen-centered autoimmune responses and bone stromal cells on the onset of AS. Autoantigens derived from gut microbiota and preferential TCR both exacerbate the autoimmune response in patients with AS. Furthermore, dysregulated bone metabolism also promotes pathological new bone formation in AS. Current treatments approved for AS almost focus on the management of inflammation with inconsistent treatment results due to the heterogeneity of patients. In this review, we systematically summarized various pathogenesis and management of AS, meanwhile discussed the underlying risk factors and potential therapeutic targets.
Collapse
Affiliation(s)
- Yuxiao Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| | - Shuqiong Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| | - Fenli Shao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
5
|
Chen S, Pan Y, Guo Y, Sun X, Bai X, Liu M, Wang L, Xiao J, Chen C, Ma Y. Integrative bioinformatics and experimental analysis of curcumin's role in regulating ferroptosis to combat osteoporosis. Biochem Biophys Res Commun 2024; 739:150949. [PMID: 39541922 DOI: 10.1016/j.bbrc.2024.150949] [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: 08/14/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/17/2024]
Abstract
This study utilized bioinformatics and data mining techniques to explore the molecular mechanism of curcumin in treating osteoporosis (OP) through the lens of ferroptosis, thereby identifying novel therapeutic targets. The datasets GSE35958, GSE35956, GSE7429, and GSE7158 were obtained from the Gene Expression Omnibus (GEO) database. GSE35958 and GSE35956 were employed as training sets for data integration, while GSE7429 and GSE7158 served as independent validation sets. Through retrieval from the FerrDb database, iron death-related genes (FRGs) were identified, and the differentially expressed genes (DEGs) were intersected to obtain differentially expressed FRGs (DEFRGs). Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted, followed by the construction of a Protein-Protein Interaction (PPI) network to identify Hub genes. The DGIdb database was utilized to predict candidate drugs associated with the Hub genes, and molecular docking and in vitro experiments confirmed that curcumin targets the Hub genes EGFR and PTGS2.Through in vitro testing of curcumin on BMSC cells, researchers examined cell vitality, iron death, osteogenic differentiation, mineralization, and the impact on EGFR and PTGS2 levels. The analysis yielded 2212 DEGs, 484 FRGs, with 45 FDEGs at the intersection. GO analysis revealed involvement in regulating mitochondrial proteins, amino acid transport across plasma membranes, and protein ubiquitination. KEGG pathway analysis indicated associations with iron death, FoxO signaling, mTOR signaling, cell aging, osteoclast differentiation, and GSH metabolism. Utilizing the MCC algorithm, five Hub genes were identified: MAPK3, PTGS2, TGFB1, CYBB, and EGFR, showing diagnostic potential for iron death. Curcumin displayed affinity for EGFR and PTGS2, mitigating iron-induced effects on BMSCs such as increased reactive oxygen species, Fe3+ levels, and decreased mitochondrial membrane potential. Furthermore, curcumin reversed these effects, suggesting EGFR and PTGS2 as targets for curcumin to inhibit BMSC ferroptosis and potentially delay osteoporosis development. Maintaining iron homeostasis and targeting BMSC ferroptosis could offer therapeutic avenues for iron overload-induced osteoporosis.
Collapse
Affiliation(s)
- Shuangliu Chen
- School of Chinese Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Yalan Pan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Yang Guo
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xiaoxian Sun
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xue Bai
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Mengmin Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Lining Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Jiřimutu Xiao
- College of Mongolian Medicine, Inner Mongolia Medical University, China
| | - Cheng Chen
- Jiangyan Hospital Affiliated to Nanjing University of Chinese Medicine, China.
| | - Yong Ma
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, China; Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China; Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, China.
| |
Collapse
|
6
|
Jia Y, Zhang F, Meng X, Andreev D, Lyu P, Zhang W, Lai C, Schett G, Bozec A. Osteocytes support bone metastasis of melanoma cells by CXCL5. Cancer Lett 2024; 590:216866. [PMID: 38589005 DOI: 10.1016/j.canlet.2024.216866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/18/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Bone metastasis is a common complication of certain cancers such as melanoma. The spreading of cancer cells into the bone is supported by changes in the bone marrow environment. The specific role of osteocytes in this process is yet to be defined. By RNA-seq and chemokines screening we show that osteocytes release the chemokine CXCL5 when they are exposed to melanoma cells. Osteocytes-mediated CXCL5 secretion enhanced the migratory and invasive behaviour of melanoma cells. When the expression of the CXCL5 receptor, CXCR2, was down-regulated in melanoma cells in vitro, we observed a significant decrease in melanoma cell migration in response to osteocytes. Furthermore, melanoma cells with down-regulated CXCR2 expression showed less bone metastasis and less bone loss in the bone metastasis model in vivo. Furthermore, when simultaneously down-regulating CXCL5 in osteocytes and CXCR2 in melanoma cells, melanoma progression was abrogated in vivo. In summary, these data suggest a significant role of osteocytes in bone metastasis of melanoma, which is mediated through the CXCL5-CXCR2 pathway.
Collapse
Affiliation(s)
- Yewei Jia
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Fulin Zhang
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xianyi Meng
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Darja Andreev
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Pang Lyu
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wenshuo Zhang
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Chaobo Lai
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
7
|
Gao Y, Zou Y, Sokolowskei D, Xing X, Tower RJ, Lai Z, Shi J, Zhu L, Zheng Q, James AW, Xu J, Zhang Z. Nr4a1 enhances Wnt4 transcription to promote mesenchymal stem cell osteogenesis and alleviates inflammation-inhibited bone regeneration. Mol Ther 2024; 32:1479-1496. [PMID: 38429926 PMCID: PMC11081873 DOI: 10.1016/j.ymthe.2024.02.034] [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: 09/27/2023] [Revised: 01/15/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Intense inflammatory response impairs bone marrow mesenchymal stem cell (BMSC)-mediated bone regeneration, with transforming growth factor (TGF)-β1 being the most highly expressed cytokine. However, how to find effective and safe means to improve bone formation impaired by excessive TGF-β1 remains unclear. In this study, we found that the expression of orphan nuclear receptor Nr4a1, an endogenous repressor of TGF-β1, was suppressed directly by TGF-β1-induced Smad3 and indirectly by Hdac4, respectively. Importantly, Nr4a1 overexpression promoted BMSC osteogenesis and reversed TGF-β1-mediated osteogenic inhibition and pro-fibrotic effects. Transcriptomic and histologic analyses confirmed that upregulation of Nr4a1 increased the transcription of Wnt family member 4 (Wnt4) and activated Wnt pathway. Mechanistically, Nr4a1 bound to the promoter of Wnt4 and regulated its expression, thereby enhancing the osteogenic capacity of BMSCs. Moreover, treatment with Nr4a1 gene therapy or Nr4a1 agonist Csn-B could promote ectopic bone formation, defect repair, and fracture healing. Finally, we demonstrated the correlation of NR4A1 with osteogenesis and the activation of the WNT4/β-catenin pathway in human BMSCs and fracture samples. Taken together, these findings uncover the critical role of Nr4a1 in bone formation and alleviation of inflammation-induced bone regeneration disorders, and suggest that Nr4a1 has the potential to be a therapeutic target for accelerating bone healing.
Collapse
Affiliation(s)
- Yangshuai Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuming Zou
- Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Dimitri Sokolowskei
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert J Tower
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zejia Lai
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiawei Shi
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Liheng Zhu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Quan Zheng
- Department of Orthopedic Surgery, Luan Hospital Affiliated to Anhui Medical University, Luan, Anhui 237001, China
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Jiajia Xu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
8
|
Song C, Wang K, Qian B, Lu J, Qiao M, Qiu Y, Wang B, Yu Y. Nrf-2/ROS/NF-κB pathway is modulated by cynarin in human mesenchymal stem cells in vitro from ankylosing spondylitis. Clin Transl Sci 2024; 17:e13748. [PMID: 38450992 PMCID: PMC10918724 DOI: 10.1111/cts.13748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
Ankylosing spondylitis (AS) is an immune chronic inflammatory disease, resulting in back pain, stiffness, and thoracolumbar kyphotic deformity. Based on the reported anti-inflammatory and antioxidant capacities of cynarin (Cyn), this study explored its protective role and molecular mechanisms in mesenchymal stem cells (MSCs) from AS. The target pathways and genes were verified using Western blotting, quantitative real-time polymerase chain reaction, and immunofluorescent staining, while molecular docking analysis was conducted. In AS-MSCs, we found that the expression levels of p-NF-κB, IL-6, IL-1β, and TNF-α were higher and IκB-α, Nrf-2, and HO-1 were lower compared with healthy control (HC)-MSCs. With molecular docking analysis, the biding affinities between Cyn and Keap1-Nrf-2 and p65-IκB-α were predicted. The mRNA and protein expression of p-NF-κB, IL-6, IL-1β, and TNF-α and the reactive oxygen species (ROS) generation were downregulated following Cyn administration. Meanwhile, the expression level of IκB-α, Nrf-2, and HO-1 were significantly increased after Cyn pretreatment. The results suggested that the protective mechanisms of Cyn in AS-MSCs were based on enhancing the antioxidation and suppression of excessive inflammatory responses via Nrf-2/ROS/NF-κB axis. Our findings demonstrate that Cyn is a potential candidate for alleviating inflammation in AS.
Collapse
Affiliation(s)
- Chenyu Song
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Kaiyang Wang
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Bangping Qian
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Jingshun Lu
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Mu Qiao
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Bin Wang
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Yang Yu
- Division of Spine Surgery, Department of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| |
Collapse
|
9
|
Edderkaoui B. Chemokines in Cartilage Regeneration and Degradation: New Insights. Int J Mol Sci 2023; 25:381. [PMID: 38203552 PMCID: PMC10779035 DOI: 10.3390/ijms25010381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Cartilage plays a crucial role in the human body by forming long bones during development and growth to bear loads on joints and intervertebral discs. However, the increasing prevalence of cartilage degenerative disorders is a growing public health concern, especially due to the poor innate regenerative capacity of cartilage. Chondrocytes are a source of several inflammatory mediators that play vital roles in the pathogenesis of cartilage disorders. Among these mediators, chemokines have been explored as potential contributors to cartilage degeneration and regeneration. Our review focuses on the progress made during the last ten years in identifying the regulators and roles of chemokines and their receptors in different mechanisms related to chondrocytes and cartilage. Recent findings have demonstrated that chemokines influence cartilage both positively and negatively. Their induction and involvement in either process depends on the local molecular environment and is both site- and time-dependent. One of the challenges in defining the role of chemokines in cartilage pathology or regeneration is the apparent redundancy in the interaction of chemokines with their receptors. Hence, it is crucial to determine, for each situation, whether targeting specific chemokines or their receptors will help in developing effective therapeutic strategies for cartilage repair.
Collapse
Affiliation(s)
- Bouchra Edderkaoui
- Musculoskeletal Disease Center, Research Service, VA Loma Linda Healthcare Systems, Loma Linda, CA 92357, USA;
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
10
|
Jing Z, Li Y, Zhang H, Chen T, Yu J, Xu X, Zou Y, Wang X, Xiang K, Gong X, He P, Fu Y, Ren M, Ji P, Yang S. Tobacco toxins induce osteoporosis through ferroptosis. Redox Biol 2023; 67:102922. [PMID: 37826866 PMCID: PMC10571034 DOI: 10.1016/j.redox.2023.102922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Clinical epidemiological studies have confirmed that tobacco smoking disrupts bone homeostasis and is an independent risk factor for the development of osteoporosis. The low viability and inferior osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) are important etiologies of osteoporosis. However, few basic studies have elucidated the specific mechanisms that tobacco toxins devastated BMSCs and consequently induced or exacerbated osteoporosis. Herein, our clinical data showed the bone mineral density (BMD) values of femoral neck in smokers were significantly lower than non-smokers, meanwhile cigarette smoke extract (CSE) exposure led to a significant decrease of BMD in rats and dysfunction of rat BMSCs (rBMSCs). Transcriptomic analysis and phenotype experiments suggested that the ferroptosis pathway was significantly activated in CSE-treated rBMSCs. Accumulated intracellular reactive oxygen species activated AMPK signaling, furtherly promoted NCOA4-mediated ferritin-selective autophagic processes, increased labial iron pool and lipid peroxidation deposition, and ultimately led to ferroptosis in rBMSCs. Importantly, in vivo utilization of ferroptosis and ferritinophagy inhibitors significantly alleviated BMD loss in CSE-exposed rats. Our study innovatively reveals the key mechanism of smoking-related osteoporosis, and provides a possible route targeting on the perspective of BMSC ferroptosis for future prevention and treatment of smoking-related bone homeostasis imbalance.
Collapse
Affiliation(s)
- Zheng Jing
- Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuzhou Li
- Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - He Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tao Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Jinrui Yu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xinxin Xu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yulong Zou
- Department of Orthopedics, Second Affiliated Hospital of Chongqing Medical University, China
| | - Xu Wang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Xiang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xuerui Gong
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ping He
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yiru Fu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Mingxing Ren
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Sheng Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
11
|
Skubica P, Husakova M, Dankova P. In vitro osteoclastogenesis in autoimmune diseases - Strengths and pitfalls of a tool for studying pathological bone resorption and other disease characteristics. Heliyon 2023; 9:e21925. [PMID: 38034780 PMCID: PMC10682642 DOI: 10.1016/j.heliyon.2023.e21925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Osteoclasts play a critical role in bone pathology frequently associated with autoimmune diseases. Studying the etiopathogenesis of these diseases and their clinical manifestations can involve in vitro osteoclastogenesis, an experimental technique that utilizes osteoclast precursors that are relatively easily accessible from peripheral blood or synovial fluid. However, the increasing number of methodical options to study osteoclastogenesis in vitro poses challenges in translating findings to clinical research and practice. This review compares and critically evaluates previous research work based on in vitro differentiation of human osteoclast precursors originating from patients, which aimed to explain autoimmune pathology in rheumatic and enteropathic diseases. The discussion focuses primarily on methodical differences between the studies, including the origin of osteoclast precursors, culture conditions, and methods for identifying osteoclasts and assessing their activity. Additionally, the review examines the clinical significance of the three most commonly used in vitro approaches: induced osteoclastogenesis, spontaneous osteoclastogenesis, and cell co-culture. By analyzing and integrating the gathered information, this review proposes general connections between different studies, even in cases where their results are seemingly contradictory. The derived conclusions and future directions aim to enhance our understanding of a potential and limitations of in vitro osteoclastogenesis and provide a foundation for discussing novel methods (such as osteoclastogenesis dynamic) and standardized approaches (such as spontaneous osteoclastogenesis) for future use in autoimmune disease research.
Collapse
Affiliation(s)
- Patrik Skubica
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Marketa Husakova
- First Faculty of Medicine, Charles University, Prague and Institute of Rheumatology, Prague, Czech Republic
| | - Pavlina Dankova
- Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
12
|
Feng X, Qiao J, Xu W. Impact of immune regulation and differentiation dysfunction of mesenchymal stem cells on the disease process in ankylosing spondylitis and prospective analysis of stem cell transplantation therapy. Postgrad Med J 2023; 99:1138-1147. [PMID: 37689998 DOI: 10.1093/postmj/qgad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/19/2023] [Accepted: 08/11/2023] [Indexed: 09/11/2023]
Abstract
Ankylosing spondylitis (AS) is a rheumatic bone and joint disease caused by inflammation, erosion, and pathological bone formation. The pathological features of chronic inflammation, bone destruction, and pathological ossification occur due to the disruption of the body's immune regulation and altered bone remodeling balance. Mesenchymal stem cells (MSCs) have multidirectional differentiation potential and immunomodulatory functions and play an important role in immune regulation and bone formation. The immune regulation and osteogenic capacity of MSCs in AS are altered by factors such as genetic background, internal environment, infection, and mechanical forces that drive disease development. This review further evaluates the role of MSCs dysfunction in inflammation and pathological bone formation by analyzing the effects of the above-mentioned factors on MSCs function and also looks forward to the prospects of MSCs in treating AS, providing some ideas for an in-depth study of inflammation and ectopic ossification. KEY MESSAGES
Collapse
Affiliation(s)
- Xinzhe Feng
- Department of Joint Bone Disease Surgery, Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Junjie Qiao
- Department of Joint Bone Disease Surgery, Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Weidong Xu
- Department of Joint Bone Disease Surgery, Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
13
|
Mauro D, Gandolfo S, Tirri E, Schett G, Maksymowych WP, Ciccia F. The bone marrow side of axial spondyloarthritis. Nat Rev Rheumatol 2023:10.1038/s41584-023-00986-6. [PMID: 37407716 DOI: 10.1038/s41584-023-00986-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 07/07/2023]
Abstract
Spondyloarthritis (SpA) is characterized by the infiltration of innate and adaptive immune cells into entheses and bone marrow. Molecular, cellular and imaging evidence demonstrates the presence of bone marrow inflammation, a hallmark of SpA. In the spine and the peripheral joints, bone marrow is critically involved in the pathogenesis of SpA. Evidence suggests that bone marrow inflammation is associated with enthesitis and that there are roles for mechano-inflammation and intestinal inflammation in bone marrow involvement in SpA. Specific cell types (including mesenchymal stem cells, innate lymphoid cells and γδ T cells) and mediators (Toll-like receptors and cytokines such as TNF, IL-17A, IL-22, IL-23, GM-CSF and TGFβ) are involved in these processes. Using this evidence to demonstrate a bone marrow rather than an entheseal origin for SpA could change our understanding of the disease pathogenesis and the relevant therapeutic approach.
Collapse
Affiliation(s)
- Daniele Mauro
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Saviana Gandolfo
- Unit of Rheumatology, San Giovanni Bosco Hospital, Naples, Italy
| | - Enrico Tirri
- Unit of Rheumatology, San Giovanni Bosco Hospital, Naples, Italy
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander University (FAU) Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Francesco Ciccia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
14
|
Lin YC, Zheng G, Liu HT, Wang P, Yuan WQ, Zhang YH, Peng XS, Li GJ, Wu YF, Shen HY. USP7 promotes the osteoclast differentiation of CD14+ human peripheral blood monocytes in osteoporosis via HMGB1 deubiquitination. J Orthop Translat 2023; 40:80-91. [PMID: 37333461 PMCID: PMC10275958 DOI: 10.1016/j.jot.2023.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/20/2023] Open
Abstract
Background Abnormal osteoclast and osteoblast differentiation is an essential pathological process in osteoporosis. As an important deubiquitinase enzyme, ubiquitin-specific peptidase 7 (USP7) participates in various disease processes through posttranslational modification. However, the mechanism by which USP7 regulates osteoporosis remains unknown. Herein, we aimed to investigate whether USP7 regulates abnormal osteoclast differentiation in osteoporosis. Methods The gene expression profiles of blood monocytes were preprocessed to analyze the differential expression of USP genes. CD14+ peripheral blood mononuclear cells (PBMCs) were isolated from whole blood collected from osteoporosis patients (OPs) and healthy donors (HDs), and the expression pattern of USP7 during the differentiation of CD14+ PBMCs into osteoclasts was detected by western blotting. The role of USP7 in the osteoclast differentiation of PBMCs treated with USP7 siRNA or exogenous rUSP7 was further investigated by the F-actin assay, TRAP staining and western blotting. Moreover, the interaction between high-mobility group protein 1 (HMGB1) and USP7 was investigated by coimmunoprecipitation, and the regulation of the USP7-HMGB1 axis in osteoclast differentiation was further verified. Osteoporosis in ovariectomized (OVX) mice was then studied using the USP7-specific inhibitor P5091 to identify the role of USP7 in osteoporosis. Results The bioinformatic analyses and CD14+ PBMCs from osteoporosis patients confirmed that the upregulation of USP7 was associated with osteoporosis. USP7 positively regulates the osteoclast differentiation of CD14+ PBMCs in vitro. Mechanistically, USP7 promoted osteoclast formation by binding to and deubiquitination of HMGB1. In vivo, P5091 effectively attenuates bone loss in OVX mice. Conclusion We demonstrate that USP7 promotes the differentiation of CD14+ PBMCs into osteoclasts via HMGB1 deubiquitination and that inhibition of USP7 effectively attenuates bone loss in osteoporosis in vivo.The translational potential of this article:The study reveals novel insights into the role of USP7 in the progression of osteoporosis and provides a new therapeutic target for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Yu-Cong Lin
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Hua-Tao Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Wei-Quan Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Yun-Hui Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Xiao-Shuai Peng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Guo-Jian Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Yan-Feng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| | - Hui-Yong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, PR China
| |
Collapse
|
15
|
Zeng Y, Deng JJ, Jiang QL, Wang CL, Zhang L, Li T, Jiang J. Thyrotropin inhibits osteogenic differentiation of human periodontal ligament stem cells. J Periodontal Res 2023; 58:668-678. [PMID: 36807238 DOI: 10.1111/jre.13109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/07/2023] [Accepted: 02/07/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND OBJECTIVE Periodontal ligament stem cells (PDLSCs) are derived from the periodontal ligament and have the characteristics of pluripotent differentiation, including osteogenesis, and are one of the important seed cells in oral tissue engineering. Thyrotropin (TSH) has been shown to regulate bone metabolism independently of thyroid hormone, including the fate of osteoblasts and osteoclasts, but whether it affects osteogenic differentiation of PDLSCs is unknown. MATERIALS AND METHODS PDLSCs were isolated and cultured from human periodontal ligament and grown in osteogenic medium (containing sodium β-glycerophosphate, ascorbic acid, and dexamethasone). Recombinant human TSH was added to the culture medium. Osteogenic differentiation of PDLSCs was assessed after 14 days by staining with alkaline phosphatase and alizarin red and by detection of osteogenic differentiation genes. Differentially expressed genes (DEGs) in PDLSCs under TSH were detected by high-throughput sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyzed the biological functions and signaling pathways involved in DEGs. RESULTS We found that osteogenic differentiation of PDLSCs was significantly inhibited in the presence of TSH: including decreased calcium nodule formation, decreased alkaline phosphatase levels, and decreased collagen synthesis. Using high-throughput sequencing, we found changes in the expression of some osteogenesis-related genes, which may be the reason that TSH inhibits osteogenic differentiation of PDLSCs. CONCLUSION Unless TSH is ≥10 mU/L, patients with subclinical hypothyroidism usually do not undergo thyroxine supplementation therapy. However, in this work, we found that elevated TSH inhibited the osteogenic differentiation of PDLSCs. Therefore, correction of TSH levels in patients with subclinical hypothyroidism may be beneficial to improve orthodontic, implant, and periodontitis outcomes in these patients.
Collapse
Affiliation(s)
- Yang Zeng
- Department of Orthodontics, Affiliated Stomatology Hospital of Southwest Medical University, Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, Sichuan Province, China
| | - Ji-Jun Deng
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qi-Lan Jiang
- Department of Clinical Nutrition, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chun-Lian Wang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Li Zhang
- Department of Orthodontics, Affiliated Stomatology Hospital of Southwest Medical University, Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, Sichuan Province, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
16
|
Fatica M, D'Antonio A, Novelli L, Triggianese P, Conigliaro P, Greco E, Bergamini A, Perricone C, Chimenti MS. How Has Molecular Biology Enhanced Our Undertaking of axSpA and Its Management. Curr Rheumatol Rep 2023; 25:12-33. [PMID: 36308677 PMCID: PMC9825525 DOI: 10.1007/s11926-022-01092-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE This review aims at investigating pathophysiological mechanisms in spondyloarthritis (SpA). Analysis of genetic factors, immunological pathways, and abnormalities of bone metabolism lay the foundations for a better understanding of development of the axial clinical manifestations in patients, allowing physician to choose the most appropriate therapeutic strategy in a more targeted manner. RECENT FINDINGS In addition to the contribution of MHC system, findings emerged about the role of non-HLA genes (as ERAP1 and 2, whose inhibition could represent a new therapeutic approach) and of epigenetic mechanisms that regulate the expression of genes involved in SpA pathogenesis. Increasing evidence of bone metabolism abnormalities secondary to the activation of immunological pathways suggests the development of various bone anomalies that are present in axSpA patients. SpA are a group of inflammatory diseases with a multifactorial origin, whose pathogenesis is linked to the genetic predisposition, the action of environmental risk factors, and the activation of immune response. It is now well known how bone metabolism leads to long-term structural damage via increased bone turnover, bone loss and osteoporosis, osteitis, erosions, osteosclerosis, and osteoproliferation. These effects can exist in the same patient over time or even simultaneously. Evidence suggests a cross relationship among innate immunity, autoimmunity, and bone remodeling in SpA, making treatment approach a challenge for rheumatologists. Specifically, treatment targets are consistently increasing as new drugs are upcoming. Both biological and targeted synthetic drugs are promising in terms of their efficacy and safety profile in patients affected by SpA.
Collapse
Affiliation(s)
- Mauro Fatica
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Arianna D'Antonio
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Lucia Novelli
- UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Elisabetta Greco
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alberto Bergamini
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Perricone
- Rheumatology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
17
|
Lai G, Zhao R, Zhuang W, Hou Z, Yang Z, He P, Wu J, Sang H. BMSC-derived exosomal miR-27a-3p and miR-196b-5p regulate bone remodeling in ovariectomized rats. PeerJ 2022; 10:e13744. [PMID: 36168439 PMCID: PMC9509671 DOI: 10.7717/peerj.13744] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/27/2022] [Indexed: 01/22/2023] Open
Abstract
Background In the bone marrow microenvironment of postmenopausal osteoporosis (PMOP), bone marrow mesenchymal stem cell (BMSC)-derived exosomal miRNAs play an important role in bone formation and bone resorption, although the pathogenesis has yet to be clarified. Methods BMSC-derived exosomes from ovariectomized rats (OVX-Exo) and sham-operated rats (Sham-Exo) were co-cultured with bone marrow-derived macrophages to study their effects on osteoclast differentiation. Next-generation sequencing was utilized to identify the differentially expressed miRNAs (DE-miRNAs) between OVX-Exo and Sham-Exo, while target genes were analyzed using bioinformatics. The regulatory effects of miR-27a-3p and miR-196b-5p on osteogenic differentiation of BMSCs and osteoclast differentiation were verified by gain-of-function and loss-of-function analyses. Results Osteoclast differentiation was significantly enhanced in the OVX-Exo treatment group compared to the Sham-Exo group. Twenty DE-miRNAs were identified between OVX-Exo and Sham-Exo, among which miR-27a-3p and miR-196b-5p promoted the expressions of osteogenic differentiation markers in BMSCs. In contrast, knockdown of miR-27a-3p and miR-196b-5p increased the expressions of osteoclastic markers in osteoclast. These 20 DE-miRNAs were found to target 11435 mRNAs. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that these target genes were involved in several biological processes and osteoporosis-related signaling pathways. Conclusion BMSC-derived exosomal miR-27a-3p and miR-196b-5p may play a positive regulatory role in bone remodeling.
Collapse
Affiliation(s)
- Guohua Lai
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Renli Zhao
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weida Zhuang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zuoxu Hou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zefeng Yang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Peipei He
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiachang Wu
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongxun Sang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Zhou C, Liang T, Jiang J, Zhang Z, Chen J, Chen T, Chen L, Sun X, Huang S, Zhu J, Wu S, Zhan X, Liu C. Immune cell infiltration-related clinical diagnostic model for Ankylosing Spondylitis. Front Genet 2022; 13:949882. [PMID: 36263434 PMCID: PMC9575679 DOI: 10.3389/fgene.2022.949882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Background: The pathogenesis and diagnosis of Ankylosing Spondylitis (AS) has remained uncertain due to several reasons, including the lack of studies on the local and systemic immune response in AS. To construct a clinical diagnostic model, this study identified the micro RNA-messenger RNA (miRNA-mRNA) interaction network and immune cell infiltration-related hub genes associated with AS. Materials and Methods: Total RNA was extracted and purified from the interspinous ligament tissue samples of three patients with AS and three patients without AS; miRNA and mRNA microarrays were constructed using the extracted RNA. Bioinformatic tools were used to construct an miRNA-mRNA network, identify hub genes, and analyze immune infiltration associated with AS. Next, we collected the blood samples and clinical characteristics of 359 patients (197 with AS and 162 without AS). On the basis of the clinical characteristics and results of the routine blood tests, we selected immune-related cells whose numbers were significantly different in patients with AS and patients without AS. Univariate and multivariate logistic regression analysis was performed to construct a nomogram. Immunohistochemistry staining analysis was utilized to verify the differentially expression of LYN in AS and controls. Results: A total of 225 differentially expressed miRNAs (DE miRNAs) and 406 differentially expressed mRNAs (DE mRNAs) were identified from the microarray. We selected 15 DE miRNAs and 38 DE mRNAs to construct a miRNA-mRNA network. The expression of LYN, an immune-related gene, correlated with the counts of monocytes, neutrophils, and dendritic cells. Based on the independent predictive factors of sex, age, and counts of monocytes, neutrophils, and white blood cells, a nomogram was established. Receiver operating characteristic (ROC) analysis was performed to evaluate the nomogram, with a C-index of 0.835 and AUC of 0.855. Conclusion:LYN, an immune-related hub gene, correlated with immune cell infiltration in patients with AS. In addition, the counts of monocytes and neutrophils were the independent diagnostic factors for AS. If verified in future studies, a diagnostic model based on these findings may be used to predict AS effectively.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xinli Zhan
- *Correspondence: Xinli Zhan, ; Chong Liu,
| | - Chong Liu
- *Correspondence: Xinli Zhan, ; Chong Liu,
| |
Collapse
|
19
|
Liu C, Liang T, Zhang Z, Chen J, Xue J, Zhan X, Ren L. Transfer of microRNA-22-3p by M2 macrophage-derived extracellular vesicles facilitates the development of ankylosing spondylitis through the PER2-mediated Wnt/β-catenin axis. Cell Death Dis 2022; 8:269. [PMID: 35606376 PMCID: PMC9126881 DOI: 10.1038/s41420-022-00900-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/07/2022] [Accepted: 02/09/2022] [Indexed: 11/25/2022]
Abstract
Pathological osteogenesis and inflammation possess critical significance in ankylosing spondylitis (AS). The current study aimed to elucidate the mechanisms regarding extracellular vesicle (EV)-packaged microRNA-22-3p (miR-22-3p) from M2 macrophages in the osteogenic differentiation of mesenchymal stem cells (MSCs) in AS. EVs were initially isolated from M2 macrophages, which had been treated with either restored or depleted miR-22-3p. AS-BMSCs were subsequently treated with M2 macrophage-derived EVs to detect osteogenic differentiation in BMSCs using gain- or loss-of-function experiments. The binding affinity among miR-22-3p, period circadian protein 2 (PER2), and Wnt7b was identified. Finally, AS mouse models were established for testing the effects of M2-EV-miR-22-3p on the bone metastatic microenvironment in vivo. miR-22-3p from M2 macrophages could be transferred into BMSCs via EVs, which promoted the osteogenic differentiation of AS-BMSCs. miR-22-3p inhibited PER2, while PER2 blocked the Wnt/β-catenin signaling pathway via Wnt7b inhibition. M2-EV-shuttled miR-22-3p facilitated alkaline phosphatase activity and extracellular matrix mineralization via PER2-regulated Wnt/β-catenin axis, stimulating the BMSC osteogenic differentiation. Taken together, these findings demonstrate that miR-22-3p in M2 macrophage-released EVs downregulates PER2 to facilitate the osteogenesis of MSCs via Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Chong Liu
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China
| | - Tuo Liang
- Guangxi Medical University, Nanning, 530021, P. R. China
| | - Zide Zhang
- Guangxi Medical University, Nanning, 530021, P. R. China
| | - Jiarui Chen
- Guangxi Medical University, Nanning, 530021, P. R. China
| | - Jang Xue
- Guangxi Medical University, Nanning, 530021, P. R. China
| | - Xinli Zhan
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P. R. China.
| | - Liang Ren
- Reproductive Medicine Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, P.R. China.
| |
Collapse
|
20
|
Ji L, Li X, He S, Chen S. Regulation of osteoclast-mediated bone resorption by microRNA. Cell Mol Life Sci 2022; 79:287. [PMID: 35536437 PMCID: PMC11071904 DOI: 10.1007/s00018-022-04298-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/15/2022] [Accepted: 04/08/2022] [Indexed: 02/08/2023]
Abstract
Osteoclast-mediated bone resorption is responsible for bone metabolic diseases, negatively impacting people's health and life. It has been demonstrated that microRNA influences the differentiation of osteoclasts by regulating the signaling pathways during osteoclast-mediated bone resorption. So far, the involved mechanisms have not been fully elucidated. This review introduced the pathways involved in osteoclastogenesis and summarized the related microRNAs binding to their specific targets to mediate the downstream pathways in osteoclast-mediated bone resorption. We also discuss the clinical potential of targeting microRNAs to treat osteoclast-mediated bone resorption as well as the challenges of avoiding potential side effects and producing efficient delivery methods.
Collapse
Affiliation(s)
- Ling Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shushu He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
21
|
[Hyperactivation of PI3K/AKT/mTOR signal pathway impairs TNF-α-induced autophagy in mesenchymal stem cells from patients with ankylosing spondylitis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:272-277. [PMID: 35365453 PMCID: PMC8983359 DOI: 10.12122/j.issn.1673-4254.2022.02.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To investigate the changes in autophagy of mesenchymal stem cells (MSCs) from patients with ankylosing spondylitis and explore the mechanism for decreased autophagy in ASMSCs. METHODS MSCs collected from 14 patients with AS (ASMSCs) and from 15 healthy donors (HDMSCs) were cultured in the absence or presence of 25 ng/mL TNF-α for 6 h. Autophagy of the cells was determined by immunofluorescence staining of GFP-LC3B, and the results were confirmed by detecting the protein expressions of autophagy markers LC3 II/LC3 I and P62. The mRNA expressions of the related genes were detected using qRT-PCR, and the protein expressions of the autophagy markers and signaling pathway-related molecules were determined with Western blotting. TG100713 was used to block the PI3K/AKT/mTOR signal pathway, and its effect on autophagy of ASMSCs was evaluated. RESULTS ASMSCs showed significantly weaker GFP-LC3B puncta staining and lower protein expression levels of LC3 II/LC3 I but higher levels of P62 protein (P < 0.05), indicating a decreased autophagy capacity as compared with HDMSCs. TNF-α-induced ASMSCs showed significantly higher protein expressions of p-PI3K/ PI3K, p-AKT/AKT and p-mTOR/mTOR than HDMSCs (P < 0.05), suggesting hyperactivation of the PI3K/AKT/mTOR signaling pathway in ASMSCs. Blocking PI3K/AKT/mTOR signaling with TG100713 eliminated the difference in TNF-α-induced autophagy between HDMSCs and ASMSCs. CONCLUSION In patients with AS, hyperactivation of the PI3K/AKT/mTOR signaling pathway results in decreased autophagy of the MSCs and potentially contributes to chronic inflammation.
Collapse
|
22
|
Liao HT, Tsai CY, Lai CC, Hsieh SC, Sun YS, Li KJ, Shen CY, Wu CH, Lu CH, Kuo YM, Li TH, Chou CT, Yu CL. The Potential Role of Genetics, Environmental Factors, and Gut Dysbiosis in the Aberrant Non-Coding RNA Expression to Mediate Inflammation and Osteoclastogenic/Osteogenic Differentiation in Ankylosing Spondylitis. Front Cell Dev Biol 2022; 9:748063. [PMID: 35127698 PMCID: PMC8811359 DOI: 10.3389/fcell.2021.748063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/09/2021] [Indexed: 12/17/2022] Open
Abstract
Ankylosing spondylitis (AS) or radiographic axial spondyloarthritis is a chronic immune-mediated rheumatic disorder characterized by the inflammation in the axial skeleton, peripheral joints, and soft tissues (enthesis, fascia, and ligament). In addition, the extra-skeletal complications including anterior uveitis, interstitial lung diseases and aortitis are found. The pathogenesis of AS implicates an intricate interaction among HLA (HLA-B27) and non-HLA loci [endoplasmic reticulum aminopeptidase 1 (ERAP1), and interleukin-23 receptor (IL23R), gut dysbiosis, immune plasticity, and numerous environmental factors (infections, heavy metals, stress, cigarette smoking, etc.) The latter multiple non-genetic factors may exert a powerful stress on epigenetic regulations. These epigenetic regulations of gene expression contain DNA methylation/demethylation, histone modifications and aberrant non-coding RNAs (ncRNAs) expression, leading to inflammation and immune dysfunctions. In the present review, we shall discuss these contributory factors that are involved in AS pathogenesis, especially the aberrant ncRNA expression and its effects on the proinflammatory cytokine productions (TNF-α, IL-17 and IL-23), T cell skewing to Th1/Th17, and osteoclastogenic/osteogenic differentiation. Finally, some potential investigatory approaches are raised for solving the puzzles in AS pathogenesis.
Collapse
Affiliation(s)
- Hsien-Tzung Liao
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Chang-Youh Tsai
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- *Correspondence: Chang-Youh Tsai, ; Chia-Li Yu,
| | - Chien-Chih Lai
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Song-Chou Hsieh
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Syuan Sun
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Ko-Jen Li
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chieh-Yu Shen
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Han Wu
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Hsun Lu
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Min Kuo
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tzu-Hao Li
- Division of Allergy, Immunology and Rheumatology, Taipei, Taiwan
- Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chung-Tei Chou
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Chia-Li Yu
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- *Correspondence: Chang-Youh Tsai, ; Chia-Li Yu,
| |
Collapse
|
23
|
Klosterhoff BS, Vantucci CE, Kaiser J, Ong KG, Wood LB, Weiss JA, Guldberg RE, Willett NJ. Effects of osteogenic ambulatory mechanical stimulation on early stages of BMP-2 mediated bone repair. Connect Tissue Res 2022; 63:16-27. [PMID: 33820456 PMCID: PMC8490484 DOI: 10.1080/03008207.2021.1897582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Mechanical loading of bone defects through rehabilitation is a promising approach to stimulate repair and reduce nonunion risk; however, little is known about how therapeutic mechanical stimuli modulate early-stage repair before mineralized bone formation. The objective of this study was to investigate the early effects of osteogenic loading on cytokine expression and angiogenesis during the first 3 weeks of BMP-2 mediated segmental bone defect repair.Materials and Methods: A rat model of BMP-2 mediated bone defect repair was subjected to an osteogenic mechanical loading protocol using ambulatory rehabilitation and a compliant, load-sharing fixator with an integrated implantable strain sensor. The effect of fixator load-sharing on local tissue strain, angiogenesis, and cytokine expression was evaluated.Results: Using sensor readings for local measurements of boundary conditions, finite element simulations showed strain became amplified in remaining soft tissue regions between 1 and 3 weeks (Week 3: load-sharing: -1.89 ± 0.35% and load-shielded: -1.38 ± 0.35% vs. Week 1: load-sharing: -1.54 ± 0.17%; load-shielded: -0.76 ± 0.06%). Multivariate analysis of cytokine arrays revealed that load-sharing significantly altered expression profiles in the defect tissue at 2 weeks compared to load-shielded defects. Specifically, loading reduced VEGF (p = 0.052) and increased CXCL5 (LIX) levels. Subsequently, vascular volume in loaded defects was reduced relative to load-shielded defects but similar to intact bone at 3 weeks. Endochondral bone repair was also observed histologically in loaded defects at 3 weeks.Conclusions: Together, these results demonstrate that moderate ambulatory strains previously shown to stimulate bone regeneration significantly alter early angiogenic and cytokine signaling and may promote endochondral ossification.
Collapse
Affiliation(s)
- Brett S. Klosterhoff
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - Casey E. Vantucci
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - Jarred Kaiser
- Research Service, Atlanta VA Medical Center, Decatur, GA,Department of Orthopaedics, Emory University, Atlanta, GA
| | | | - Levi B. Wood
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - Jeffrey A. Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT,Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT,Department of Orthopedics, University of Utah, Salt Lake City, UT
| | | | - Nick J. Willett
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA,Research Service, Atlanta VA Medical Center, Decatur, GA,Department of Orthopaedics, Emory University, Atlanta, GA
| |
Collapse
|
24
|
Yang W, Li HY, Wu YF, Mi RJ, Liu WZ, Shen X, Lu YX, Jiang YH, Ma MJ, Shen HY. ac4C acetylation of RUNX2 catalyzed by NAT10 spurs osteogenesis of BMSCs and prevents ovariectomy-induced bone loss. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 26:135-147. [PMID: 34513300 PMCID: PMC8413676 DOI: 10.1016/j.omtn.2021.06.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 06/25/2021] [Indexed: 12/16/2022]
Abstract
N-acetyltransferase 10 (NAT10) is the key enzyme for N4-acetylcytidine (ac4C) modification of mRNA, which participates in various cellular processes and is related to many diseases. Here, we explore the relationships among osteoblast differentiation, NAT10, and ac4C, and we found that NAT0 expression and the ac4C level of total RNA were decreased in the bone tissues of bilateral ovariectomized (OVX) mice and osteoporosis patients. Adenoviruses overexpressing NAT10 reversed bone loss, and Remodelin, an NAT10 inhibitor, enhanced the loss of bone mass in OVX mice. Moreover, bone marrow-derived mesenchymal stem cells (BMSCs) with low-level ac4C modification formed fewer calcium nodules in vitro with NAT10 silencing, whereas BMSCs with high-level ac4C modification formed more calcium nodules with NAT10 overexpression. Moreover, we demonstrated that the ac4C level of runt-related transcription factor 2 (RUNX2) mRNA was increased after BMSCs were cultured in osteogenic medium (OM) and decreased after NAT10 silencing. The RUNX2 mRNA half-life and protein expression decreased after silencing NAT10 in BMSCs. Therefore, NAT10-based ac4C modification promotes the osteogenic differentiation of BMSCs by regulating the RUNX2 ac4C level. Because abnormal levels of NAT10 are probably one of the mechanisms responsible for osteoporosis, NAT10 is a new potential therapeutic target for this disease.
Collapse
Affiliation(s)
- W Yang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - H Y Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - Y F Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, People's Republic of China
| | - R J Mi
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, People's Republic of China
| | - W Z Liu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - X Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - Y X Lu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, People's Republic of China
| | - Y H Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - M J Ma
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China
| | - H Y Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, Guangdong 518033, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| |
Collapse
|
25
|
Wang B, Gong S, Shao W, Han L, Li Z, Zhang Z, Zheng Y, Ouyang F, Ma Y, Xu W, Feng Y. Comprehensive analysis of pivotal biomarkers, immune cell infiltration and therapeutic drugs for steroid-induced osteonecrosis of the femoral head. Bioengineered 2021; 12:5971-5984. [PMID: 34488536 PMCID: PMC8815624 DOI: 10.1080/21655979.2021.1972081] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is a progressive disease that leads to an increased disability rate. This study aimed to ascertain biomarkers, infiltrating immune cells, and therapeutic drugs for SONFH. The gene expression profile of the GSE123568 dataset was downloaded from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were identified using the NetworkAnalyst platform. Functional enrichment, protein-protein interaction network (PPI), and module analyses were performed using Metascape tools. An immune cell abundance identifier was used to explore immune cell infiltration. Furthermore, hub genes were identified based on maximal clique centrality (MCC) evaluation using cytoHubba application and confirmed by qRT-PCR using clinical samples. Finally, the L1000 platform was used to determine potential drugs for SONFH treatment. The SONFH mouse model was used to determine the therapeutic effects of aspirin. In total, 429 DEGs were identified in SONFH samples. Functional enrichment analysis showed that these DEGs were enriched in myeloid leukocyte activation and osteoclast differentiation processes. A set of nine immune cell types was confirmed to be markedly different between the SONFH and control samples. All 10 hub genes were significantly highly expressed in the serum of SONFH patients, as shown by qRT-PCR. Finally, the therapeutic effect of aspirin on SONFH was examined in animal experiments. Taken together, our data revealed the hub genes and infiltrating immune cells in SONFH, and we also screened potential drugs for use in SONFH treatment.
Collapse
Affiliation(s)
- Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Song Gong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Lizhi Han
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Zhichao Zhang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Yang Zheng
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Fang Ouyang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Yan Ma
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Weihua Xu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| |
Collapse
|
26
|
Yang H, Zhang W, Luan Q, Liu Y. miR-4284 Promotes Cell Proliferation, Migration, and Invasion in Non-Small Cell Lung Cancer Cells and is Associated with Postoperative Prognosis. Cancer Manag Res 2021; 13:5865-5872. [PMID: 34349559 PMCID: PMC8326286 DOI: 10.2147/cmar.s305379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/18/2021] [Indexed: 12/20/2022] Open
Abstract
PURPOSE MicroRNA-4284 (miR-4284) was demonstrated to be aberrantly expressed and affected cell activities in some types of diseases, including cancer. However, the role of miR-4284 in non-small cell lung cancer (NSCLC) is largely unknown. The aim of this study was to investigate the expression and biological role of miR-4284 in NSCLC. PATIENTS AND METHODS The qRT-PCR assay was applied to detect the expression of miR-4284 in NSCLC tissues and cell lines. Kaplan-Meier curve method and multiple Cox regression analyses were used to explore the prognostic factors for postoperative NSCLC patients. The CCK-8 assay was carried out to measure the proliferative abilities of A549 and H1299 cells. Transwell migration and invasion assays were used to determine the cell migratory and invasive capabilities of NSCLC cells. RESULTS miR-4284 expression was upregulated in NSCLC tissues and cell lines. High expression of miR-4284 was correlated with poor differentiation, positive lymph node metastasis, and advanced TNM stages. In addition, postoperative patients with higher expression of miR-4284 exhibited a shorter overall survival time than those with lower expression of miR-4284. Moreover, the upregulation of miR-4284 accelerated cell proliferative, migratory, and invasive abilities of A549 and H1299 cells, while the downregulation of miR-4284 inhibited these cellular capabilities. CONCLUSION miR-4284 was noticeably upregulated in NSCLC and associated with a poor prognosis of postoperative NSCLC patients. miR-4284 promoted the proliferation, migration, and invasion of A549 and H1299 cells. This study indicated that miR-4284 might serve as a prognostic biomarker and a potential therapeutic target for postoperative NSCLC patients.
Collapse
Affiliation(s)
- Hanbing Yang
- Department of Interventional Thoracic Oncology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, People’s Republic of China
| | - Wenjing Zhang
- Department of Hematology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, People’s Republic of China
| | - Qingxia Luan
- Department of Pediatrics, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, People’s Republic of China
| | - Yanchao Liu
- Department of Hematology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, People’s Republic of China
| |
Collapse
|
27
|
Ma M, Li H, Wang P, Yang W, Mi R, Zhuang J, Jiang Y, Lu Y, Shen X, Wu Y, Shen H. ATF6 aggravates angiogenesis-osteogenesis coupling during ankylosing spondylitis by mediating FGF2 expression in chondrocytes. iScience 2021; 24:102791. [PMID: 34296071 PMCID: PMC8281657 DOI: 10.1016/j.isci.2021.102791] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 01/02/2023] Open
Abstract
Although angiogenesis-osteogenesis coupling is important in ankylosing spondylitis (AS), therapeutic agents targeting the vasculature remain elusive. Here, we identified activating transcription factor 6 (ATF6) as an important regulator of angiogenesis in the pathogenesis of AS. First, we found that ATF6 and fibroblast growth factor 2 (FGF2) levels were higher in SKG mice and in cartilage of pateints with AS1. The proangiogenic activity of human chondrocytes was enhanced by the activation of the ATF6-FGF2 axis following 7 days of stimulation with inflammatory factors, e.g., tumor necrosis factor alpha (TNF-α), interferon-γ (IFN-γ) or interleukin-17 (IL-17). Mechanistically, ATF6 interacted with the FGF2 promotor and promoted its transcription. Treatment with the ATF6 inhibitor Ceapin-A7 inhibited angiogenesis in vitro and angiogenesis-osteogenesis coupling in vivo. ATF6 may aggravate angiogenesis-osteogenesis coupling during AS by mediating FGF2 transcription in chondrocytes, implying that ATF6 represents a promising therapeutic target for AS.
Collapse
Affiliation(s)
- Mengjun Ma
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Hongyu Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Wen Yang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Jiahao Zhuang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Yuhang Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Yixuan Lu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Xin Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, China
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China
| |
Collapse
|
28
|
Kawata K, Koga H, Tsuji K, Miyatake K, Nakagawa Y, Yokota T, Sekiya I, Katagiri H. Extracellular vesicles derived from mesenchymal stromal cells mediate endogenous cell growth and migration via the CXCL5 and CXCL6/CXCR2 axes and repair menisci. Stem Cell Res Ther 2021; 12:414. [PMID: 34294118 PMCID: PMC8296733 DOI: 10.1186/s13287-021-02481-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) are promising candidates for tissue regeneration therapy. However, the therapeutic efficacy of MSC-EVs for meniscus regeneration is uncertain, and the mechanisms underlying MSC-EV-mediated tissue regeneration have not been fully elucidated. The aims of this study were to evaluate the therapeutic efficacy of intra-articular MSC-EV injection in a meniscus defect model and elucidate the mechanism underlying MSC-EV-mediated tissue regeneration via combined bioinformatic analyses. Methods MSC-EVs were isolated from human synovial MSC culture supernatants via ultrafiltration. To evaluate the meniscus regeneration ability, MSC-EVs were injected intra-articularly in the mouse meniscus defect model immediately after meniscus resection and weekly thereafter. After 1 and 3 weeks, their knees were excised for histological and immunohistochemical evaluations. To investigate the mechanisms through which MSC-EVs accelerate meniscus regeneration, cell growth, migration, and chondrogenesis assays were performed using treated and untreated chondrocytes and synovial MSCs with or without MSC-EVs. RNA sequencing assessed the gene expression profile of chondrocytes stimulated by MSC-EVs. Antagonists of the human chemokine CXCR2 receptor (SB265610) were used to determine the role of CXCR2 on chondrocyte cell growth and migration induced by MSC-EVs. Results In the meniscus defect model, MSC-EV injection accelerated meniscus regeneration and normalized the morphology and composition of the repaired tissue. MSC-EVs stimulated chondrocyte and synovial MSC cell growth and migration. RNA sequencing revealed that MSC-EVs induced 168 differentially expressed genes in the chondrocytes and significantly upregulated CXCL5 and CXCL6 in chondrocytes and synovial MSCs. Suppression of CXCL5 and CXCL6 and antagonism of the CXCR2 receptor binding CXCL5 and CXCL6 negated the influence of MSC-EVs on chondrocyte cell growth and migration. Conclusions Intra-articular MSC-EV administration repaired meniscus defects and augmented chondrocyte and synovial MSC cell growth and migration. Comprehensive transcriptome/RNA sequencing data confirmed that MSC-EVs upregulated CXCL5 and CXCL6 in chondrocytes and mediated the cell growth and migration of these cells via the CXCR2 axis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02481-9.
Collapse
Affiliation(s)
- Kazumasa Kawata
- Department of Joint Surgery and Sports Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Kunikazu Tsuji
- Department of Joint Surgery and Sports Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Kazumasa Miyatake
- Department of Joint Surgery and Sports Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Yusuke Nakagawa
- Department of Joint Surgery and Sports Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Hiroki Katagiri
- Department of Joint Surgery and Sports Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan. .,Department of Orthopedics, Dokkyo Medical University Saitama Medical Center, 2-1-50 Minamikoshigaya, Koshigaya, Saitama, 343-8555, Japan.
| |
Collapse
|
29
|
Motta F, Pederzani A, Carena MC, Ceribelli A, Wordsworth PB, De Santis M, Selmi C, Vecellio M. MicroRNAs in Axial Spondylarthritis: an Overview of the Recent Progresses in the Field with a Focus on Ankylosing Spondylitis and Psoriatic Arthritis. Curr Rheumatol Rep 2021; 23:59. [PMID: 34216293 PMCID: PMC8254706 DOI: 10.1007/s11926-021-01027-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/18/2022]
Abstract
Purpose of Review To highlight the recent discoveries and lines of evidence on the role of microRNAs in ankylosing spondylitis (AS) and psoriatic arthritis (PsA), focusing on their expression profiling and mechanisms of action. Recent Findings AS and PsA are chronic inflammatory musculoskeletal diseases with axial manifestations and represent an excellent model for studying microRNAs contribution to the disease pathogenesis, particularly through immunomodulation, inflammation, and bone remodelling, or their value as candidate diagnostic and prognostic biomarkers. Summary MicroRNAs are single-stranded nucleotides able to regulate gene expression. They are a key component of the epigenetic machinery, involved in physiological and pathological processes. The contribution of microRNAs in AS and PsA (such as miR-29a in regulating bone metabolism) is highlighted by several works in the field but their utility as possible markers must be still confirmed, particularly in larger patients’ cohorts.
Collapse
Affiliation(s)
- Francesca Motta
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andrea Pederzani
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Angela Ceribelli
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Paul B Wordsworth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Matteo Vecellio
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy. .,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
30
|
Tian P, Wang Y, Du W. Ultrasound-targeted microbubble destruction enhances the anti-tumor action of miR-4284 inhibitor in non-small cell lung cancer cells. Exp Ther Med 2021; 21:551. [PMID: 33850523 PMCID: PMC8027739 DOI: 10.3892/etm.2021.9983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are known to be involved in various human cancer types. Ultrasound-targeted microbubble destruction (UTMD) may improve the transfection efficiency of exogenous genes into target tissues and organs, thereby improving cancer treatment. In the present study, the role of miR-4284 in non-small cell lung cancer (NSCLC) was investigated and the effect of UTMD-mediated inhibition of miR-4284 on tumor progression was further analyzed. The expression of miR-4284 in NSCLC cells and tissues was detected by reverse transcription-quantitative PCR. UTMD-mediated inhibition of miR-4284 was achieved by co-transfection of microvesicles and miR-4284 inhibitors into NSCLC cells. A Cell Counting Kit-8 assay was used to determine NSCLC cell proliferation, and the migration and invasion of NSCLC cells were examined by Transwell assays. Compared with that in the control group, the expression of miR-4284 was increased in NSCLC tissues and cells. Knockdown of miR-4284 in NSCLC cells inhibited cell proliferation, migration and invasion. UTMD improved the transfection efficiency of miR-4284 inhibitors in NSCLC cells, resulting in more significant inhibition of tumor cell proliferation, migration and invasion. In conclusion, the results indicated that the expression of miR-4284 was increased in clinical samples and cell lines of NSCLC and that knockdown of miR-4284 inhibited the proliferation, migration and invasion of tumor cells. UTMD-mediated miR-4284 inhibition further promoted this effect, indicating that this technique may represent a novel strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Peng Tian
- Department of Ultrasonics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Yanzhen Wang
- Department of Ultrasonics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Wenyan Du
- Department of Science and Education, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
31
|
Inhibition of Allogeneic and Autologous T Cell Proliferation by Adipose-Derived Mesenchymal Stem Cells of Ankylosing Spondylitis Patients. Stem Cells Int 2021; 2021:6637328. [PMID: 33777148 PMCID: PMC7979299 DOI: 10.1155/2021/6637328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/17/2021] [Accepted: 02/15/2021] [Indexed: 01/14/2023] Open
Abstract
Background In ankylosing spondylitis (AS), accompanied by chronic inflammation, T cell expansion plays a pathogenic role; the immunoregulatory properties of bone marrow-derived mesenchymal stem cells (BM-MSCs) are impaired, while functional characteristics of their adipose tissue-derived counterparts are (ASCs) unknown. Methods We evaluated the antiproliferative activity of AS/ASCs, obtained from 20 patients, towards allogeneic and autologous T lymphocytes, using ASCs from healthy donors (HD/ASCs) as the reference cell lines. The PHA-activated peripheral blood mononuclear cells (PBMCs) were cocultured in cell-cell contact and transwell conditions with untreated or TNF + IFNγ- (TI-) licensed ASCs, then analyzed by flow cytometry to identify proliferating and nonproliferating CD4+ and CD8+ T cells. The concentrations of kynurenines, prostaglandin E2 (PGE2), and IL-10 were measured in culture supernatants. Results In an allogeneic system, HD/ASCs and AS/ASCs similarly decreased the proliferation of CD4+ and CD8+ T cells and acted mainly via soluble factors. The concentrations of kynurenines and PGE2 inversely correlated with T cell proliferation, and selective inhibitors of these factors synthesis significantly restored T cell response. AS/ASCs exerted a similar antiproliferative impact also on autologous T cells. Conclusion We report for the first time that despite chronic in vivo exposure to inflammatory conditions, AS/ASCs retain the normal capability to restrain expansion of allogeneic and autologous CD4+ and CD8+ T cells, act primarily via kynurenines and PGE2, and thus may have potential therapeutic value. Some distinctions between the antiproliferative effects of AS/ASCs and HD/ASCs suggest in vivo licensing of AS/ASCs.
Collapse
|
32
|
Mauro D, Simone D, Bucci L, Ciccia F. Novel immune cell phenotypes in spondyloarthritis pathogenesis. Semin Immunopathol 2021; 43:265-277. [PMID: 33569634 PMCID: PMC7990868 DOI: 10.1007/s00281-021-00837-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/06/2021] [Indexed: 02/08/2023]
Abstract
Spondyloarthritis (SpA) is a heterogeneous group of chronic inflammatory diseases of unknown etiology. Over time, the plethora of cellular elements involved in its pathogenesis has progressively enriched together with the definition of specific cytokine pathways. Recent evidence suggests the involvement of new cellular mediators of inflammation in the pathogenesis of SpA or new subgroups of known cellular mediators. The research in this sense is ongoing, and it is clear that this challenge aimed at identifying new cellular actors involved in the perpetuation of the inflammatory process in AxSpA is not a mere academic exercise but rather aims to define a clear cellular hierarchy. Such a definition could pave the way for new targeted therapies, which could interfere with the inflammatory process and specific pathways that trigger immune system dysregulation and stromal cell activity, ultimately leading to significant control of the inflammation and new bone formation in a significant number of patients. In this review, we will describe the recent advances in terms of new cellular actors involved in the pathogenesis of SpA, focusing our attention on stromal cells and innate and adaptive immunity cells.
Collapse
Affiliation(s)
- Daniele Mauro
- Dipartimento di Medicina di Precisione, Section of Rheumatology, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Davide Simone
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Laura Bucci
- Dipartimento di Medicina di Precisione, Section of Rheumatology, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Francesco Ciccia
- Dipartimento di Medicina di Precisione, Section of Rheumatology, Università degli Studi della Campania L. Vanvitelli, Naples, Italy.
| |
Collapse
|
33
|
Liu L, Yuan Y, Zhang S, Xu J, Zou J. Osteoimmunological insights into the pathogenesis of ankylosing spondylitis. J Cell Physiol 2021; 236:6090-6100. [PMID: 33559242 DOI: 10.1002/jcp.30313] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 01/10/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Ankylosing spondylitis (AS) is inflammatory arthritis predominantly affecting the spine, which is involved in the disorders of both immune and skeletal systems. The exact pathogenesis of AS is not fully understood. Osteoimmunology is a new subject of study in inflammatory arthritis, in particular the pathogenic events involved in the cross-regulation of both skeletal and immune systems. In this review, we discuss osteoimmunological and pathological changes of AS in the spine that are characterized by altered osteogenesis and osteolytic bone destruction, accompanied by the changes of the immune system. It was revealed that bone cells like mesenchymal stem cells, osteoblast, and osteoclast in crossing talking with immune cells such as T cells, B cells coregulate to the pathogenesis of AS. Further, an array of cytokines and molecules expressed by both skeletal and immune systems contribute to these complex interplays. Understanding the cellular and molecular mechanisms underlying the pathogenesis of AS will lay a foundation for the exploration of the potential new treatment to AS.
Collapse
Affiliation(s)
- Lifei Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,Department of Rehabilitation, The People's Hospital of Liaoning Province, Shenyang, China
| | - Yu Yuan
- School of Sport and Health, Guangzhou Sport University, Guangzhou, China
| | - Shihua Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
34
|
Ma M, Yang W, Cai Z, Wang P, Li H, Mi R, Jiang Y, Xie Z, Sui P, Wu Y, Shen H. SMAD-specific E3 ubiquitin ligase 2 promotes angiogenesis by facilitating PTX3 degradation in MSCs from patients with ankylosing spondylitis. STEM CELLS (DAYTON, OHIO) 2021; 39:581-599. [PMID: 33547700 PMCID: PMC8248389 DOI: 10.1002/stem.3332] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 11/18/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
Dysregulated angiogenesis of mesenchymal stem cells (MSCs) is closely related to inflammation and disrupted bone metabolism in patients with various autoimmune diseases. However, the role of MSCs in the development of abnormal angiogenesis in patients with ankylosing spondylitis (AS) remains unclear. In this study, we cultured human umbilical vein endothelial cells (HUVECs) with bone marrow-derived MSCs from patients with AS (ASMSCs) or healthy donors (HDMSCs) in vitro. Then, the cocultured HUVECs were assayed using a cell counting kit-8 (CCK-8) to evaluate the cell proliferation. A wound healing assay was performed to investigate cell migration, and a tube formation assay was conducted to determine the angiogenesis efficiency. ASMSCs exhibited increased angiogenesis, and increased expression of SMAD-specific E3 ubiquitin ligase 2 (Smurf2) in MSCs was the main cause of abnormal angiogenesis in patients with AS. Downregulation of Smurf2 in ASMSCs blocked angiogenesis, whereas overexpression of Smurf2 in HDMSCs promoted angiogenesis. The pro-angiogenic effect of Smurf2 was confirmed by the results of a Matrigel plug assay in vivo. By functioning as an E3 ubiquitin ligase in MSCs, Smurf2 regulated the levels of pentraxin 3 (PTX3), which has been shown to suppress angiogenesis through the PTX3-fibroblast growth factor 2 pathway. Moreover, Smurf2 transcription was regulated by activating transcription factor 4-induced endoplasmic reticulum stress. In conclusion, these results identify novel roles of Smurf2 in negatively regulating PTX3 stability and promoting angiogenesis in ASMSCs.
Collapse
Affiliation(s)
- Mengjun Ma
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Wen Yang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Zhaopeng Cai
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Hongyu Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Yuhang Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Pengfei Sui
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
35
|
Oxidative stress-mediated mitochondrial dysfunction facilitates mesenchymal stem cell senescence in ankylosing spondylitis. Cell Death Dis 2020; 11:775. [PMID: 32943613 PMCID: PMC7498590 DOI: 10.1038/s41419-020-02993-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disease possessing a morbid serum microenvironment with enhanced oxidative stress. Long-term exposure to an oxidative environment usually results in cellular senescence alone with cellular dysfunction. Mesenchymal stem cells (MSCs) are a kind of stem cell possessing strong capabilities for immunoregulation, and senescent MSCs may increase inflammation and participate in AS pathogenesis. The objective of this study was to explore whether and how the oxidative serum environment of AS induces MSC senescence. Here, we found that AS serum facilitated senescence of MSCs in vitro, and articular tissues from AS patients exhibited higher expression levels of the cell cycle arrest-related proteins p53, p21 and p16. Importantly, the levels of advanced oxidative protein products (AOPPs), markers of oxidative stress, were increased in AS serum and positively correlated with the extent of MSC senescence induced by AS serum. Furthermore, MSCs cultured with AS serum showed decreased mitochondrial membrane potential and ATP production together with a reduced oxygen consumption rate. Finally, we discovered that AS serum-induced mitochondrial dysfunction resulted in elevated reactive oxygen species (ROS) in MSCs, and ROS inhibition successfully rescued MSCs from senescence. In conclusion, our data demonstrated that the oxidative serum environment of AS facilitated MSC senescence through inducing mitochondrial dysfunction and excessive ROS production. These results may help elucidate the pathogenesis of AS and provide potential targets for AS treatment.
Collapse
|
36
|
Cai Z, Liu W, Chen K, Wang P, Xie Z, Li J, Li M, Cen S, Ye G, Li Z, Su Z, Ma M, Wu Y, Shen H. Aberrantly Expressed lncRNAs and mRNAs of Osteogenically Differentiated Mesenchymal Stem Cells in Ossification of the Posterior Longitudinal Ligament. Front Genet 2020; 11:896. [PMID: 32849851 PMCID: PMC7426401 DOI: 10.3389/fgene.2020.00896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Ectopic bone formation is the chief characteristic of ossification of the posterior longitudinal ligament (OPLL). Emerging evidence has revealed that long non-coding RNAs (lncRNAs) can regulate the osteogenic differentiation of mesenchymal stem cells (MSCs), which are the main cells responsible for bone formation. However, the role of lncRNAs in the pathogenesis of OPLL remains unclear. In this study, 725 aberrantly expressed lncRNAs and 664 mRNAs in osteogenically differentiated MSCs from OPLL patients (OPLL MSCs) were identified by microarrays and confirmed by qRT-PCR assays. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses showed that the most enriched pathways included the p53, JAK-STAT, and PI3K-Akt signaling pathways. The co-expression network showed the interactions between the aberrantly expressed lncRNAs and mRNAs in OPLL MSCs, and the potential targets and transcription factors of the lncRNAs were predicted. Our research demonstrated the aberrantly expressed lncRNA and mRNA and the potential regulatory networks involved in the ectopic bone formation of OPLL. These findings imply that lncRNAs may play a vital role in OPLL, which provides a new perspective on the pathogenesis of OPLL.
Collapse
Affiliation(s)
- Zhaopeng Cai
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Keng Chen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jinteng Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ming Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuizhong Cen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guiwen Ye
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaofeng Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zepeng Su
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Mengjun Ma
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yanfeng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
37
|
Liu W, Li Z, Cai Z, Xie Z, Li J, Li M, Cen S, Tang S, Zheng G, Ye G, Su H, Wang S, Wang P, Shen H, Wu Y. LncRNA-mRNA expression profiles and functional networks in osteoclast differentiation. J Cell Mol Med 2020; 24:9786-9797. [PMID: 32715654 PMCID: PMC7520269 DOI: 10.1111/jcmm.15560] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 01/16/2023] Open
Abstract
Human osteoclasts are differentiated from CD14+ monocytes and are responsible for bone resorption. Long non‐coding RNAs (lncRNAs) have been proved to be significantly involved in multiple biologic processes, especially in cell differentiation. However, the effect of lncRNAs in osteoclast differentiation is less appreciated. In our study, RNA sequencing (RNA‐seq) was used to identify the expression profiles of lncRNAs and mRNAs in osteoclast differentiation. The results demonstrated that expressions of 1117 lncRNAs and 296 mRNAs were significantly altered after osteoclast differentiation. qRT‐PCR assays were performed to confirm the expression profiles, and the results were almost consistent with the RNA‐seq data. GO and KEGG analyses were used to predict the functions of these differentially expressed mRNA and lncRNAs. The Path‐net analysis demonstrated that MAPK pathway, PI3K‐AKT pathway and NF‐kappa B pathway played important roles in osteoclast differentiation. Co‐expression networks and competing endogenous RNA networks indicated that ENSG00000257764.2‐miR‐106a‐5p‐TIMP2 may play a central role in osteoclast differentiation. Our study provides a foundation to further understand the role and underlying mechanism of lncRNAs in osteoclast differentiation, in which many of them could be potential targets for bone metabolic disease.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaofeng Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaopeng Cai
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jinteng Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ming Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuizhong Cen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Su'an Tang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Guiwen Ye
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongjun Su
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shan Wang
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanfeng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
Wang Y, Zhang J, Su Y, Wang C, Zhang G, Liu X, Chen Q, Lv M, Chang Y, Peng J, Hou M, Huang X, Zhang X. miRNA-98-5p Targeting IGF2BP1 Induces Mesenchymal Stem Cell Apoptosis by Modulating PI3K/Akt and p53 in Immune Thrombocytopenia. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:764-776. [PMID: 32428701 PMCID: PMC7232042 DOI: 10.1016/j.omtn.2020.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/15/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Immune thrombocytopenia (ITP) is a common hematological autoimmune disease, in which defective mesenchymal stem cells (MSCs) are potentially involved. Our previous study suggested that MSCs in ITP patients displayed enhanced apoptosis. MicroRNAs (miRNAs) play important roles in ITP by affecting megakaryopoiesis, platelet production and immunoregulation, whereas the roles of miRNAs in ITP-MSCs remain unknown. In a previous study, we performed microarray analysis to obtain mRNA and miRNA profiles of ITP-MSCs. In the present study, we reanalyze the data and identify miR-98-5p as a candidate miRNA contributing to MSC deficiency in ITP. miR-98-5p acts through targeting insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1), and the subsequent downregulation of insulin-like growth factor 2 (IGF-2) causes inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which is involved in the process of MSC deficiency. Furthermore, miR-98-5p upregulates p53 by inhibiting β-transducin repeat-containing protein (β-TrCP)-dependent p53 ubiquitination. Moreover, miR-98-5p overexpression impairs the therapeutic effect of MSCs in ITP mice. All-trans retinoic acid (ATRA) protects MSCs from apoptosis by downregulating miR-98-5p, thus providing a potential therapeutic approach for ITP. Our findings demonstrate that miR-98-5p is a critical regulator of ITP-MSCs, which will help us thoroughly understand the pathogenesis of ITP.
Collapse
Affiliation(s)
- Yanan Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Jiamin Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Yan Su
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Chencong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Gaochao Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Xiao Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Meng Lv
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China.
| |
Collapse
|
39
|
Abstract
Chemokines are a family of small proteins, subdivided by their conserved cysteine residues and common structural features. Chemokines interact with their cognate G-protein-coupled receptors to elicit downstream signals that result in cell migration, proliferation, and survival. This review presents evidence for how the various CXC and CC subfamily chemokines influence bone hemostasis by acting on osteoclasts, osteoblasts, and progenitor cells. Also discussed are the ways in which chemokines contribute to bone loss as a result of inflammatory diseases such as rheumatoid arthritis, HIV infection, and periodontal infection. Both positive and negative effects of chemokines on bone formation and bone loss are presented. In addition, the role of chemokines in altering the bone microenvironment through effects on angiogenesis and tumor invasion is discussed. Very few therapeutic agents that influence bone formation by targeting chemokines or chemokine receptors are available, although a few are currently being evaluated.
Collapse
Affiliation(s)
- Annette Gilchrist
- Department of Pharmaceutical Sciences, Midwestern University, Downers Grove, IL, USA.
| |
Collapse
|