1
|
Hridayanka KSN, Duttaroy AK, Basak S. Bioactive Compounds and Their Chondroprotective Effects for Osteoarthritis Amelioration: A Focus on Nanotherapeutic Strategies, Epigenetic Modifications, and Gut Microbiota. Nutrients 2024; 16:3587. [PMID: 39519419 PMCID: PMC11547880 DOI: 10.3390/nu16213587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
In degenerative joint disease like osteoarthritis (OA), bioactive compounds like resveratrol, epigallocatechin gallate, curcumin, and other polyphenols often target various signalling pathways, including NFκB, TGFβ, and Wnt/β-catenin by executing epigenetic-modifying activities. Epigenetic modulation can target genes of disease pathophysiology via histone modification, promoter DNA methylation, and non-coding RNA expression, some of which are directly involved in OA but have been less explored. OA patients often seek options that can improve the quality of their life in addition to existing treatment with nonsteroidal anti-inflammatory drugs (NSAIDs). Although bioactive and natural compounds exhibit therapeutic potential against OA, several disadvantages loom, like insolubility and poor bioavailability. Nanoformulated bioactive compounds promise a better way to alleviate OA since they also control systemic events, including metabolic, immunological, and inflammatory responses, by modulating host gut microbiota that can regulate OA pathogenesis. Recent data suggest gut dysbiosis in OA. However, limited evidence is available on the role of bioactive compounds as epigenetic and gut modulators in ameliorating OA. Moreover, it is not known whether the effects of polyphenolic bioactive compounds on gut microbial response are mediated by epigenetic modulatory activities in OA. This narrative review highlights the nanotherapeutic strategies utilizing bioactive compounds, reporting their effects on chondrocyte growth, metabolism, and epigenetic modifications in osteoarthritis amelioration.
Collapse
Affiliation(s)
- Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway;
| | - Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| |
Collapse
|
2
|
Liu S, Li G, Xu H, Wang Q, Wei Y, Yang Q, Xiong A, Yu F, Weng J, Zeng H. "Cross-talk" between gut microbiome dysbiosis and osteoarthritis progression: a systematic review. Front Immunol 2023; 14:1150572. [PMID: 37180142 PMCID: PMC10167637 DOI: 10.3389/fimmu.2023.1150572] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/27/2023] [Indexed: 05/15/2023] Open
Abstract
Objectives The aim of this systematic review was to summarize the available literature on gut microbiome (GMB) and osteoarthritis (OA), analyze the correlation between GMB and OA, and explore potential underlying mechanisms. Methods A systematic search of the PubMed, Embase, Cochrane, and Web of Science with the keywords "Gut Microbiome" and "Osteoarthritis" was conducted to identify the human and animal studies exploring the association between GMB and OA. The retrieval time range was from the database inception to July 31, 2022. Studies reported the other arthritic diseases without OA, reviews, and studies focused on the microbiome in other parts of the body with OA, such as oral or skin, were excluded. The included studies were mainly reviewed for GMB composition, OA severity, inflammatory factors, and intestinal permeability. Results There were 31 studies published met the inclusion criteria and were analyzed, including 10 human studies and 21 animal studies. Human and animal studies have reached a consistent conclusion that GMB dysbiosis could aggravate OA. In addition, several studies have found that alterations of GMB composition can increase intestinal permeability and serum levels of inflammatory factors, while regulating GMB can alleviate the changes. Owing to the susceptibility of GMB to internal and external environments, genetics, and geography, the included studies were not consistent in GMB composition analysis. Conclusion There is a lack of high-quality studies evaluating the effects of GMB on OA. Available evidence indicated that GMB dysbiosis aggravated OA through activating the immune response and subsequent induction of inflammation. Future studies should focus on more prospective, cohort studies combined with multi-omics to further clarify the correlation.
Collapse
Affiliation(s)
- Su Liu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guoqing Li
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Huihui Xu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qichang Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yihao Wei
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qi Yang
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ao Xiong
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jian Weng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Zeng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
3
|
Xie S, Choudhari S, Wu CL, Abramson K, Corcoran D, Gregory SG, Thimmapuram J, Guilak F, Little D. Aging and obesity prime the methylome and transcriptome of adipose stem cells for disease and dysfunction. FASEB J 2023; 37:e22785. [PMID: 36794668 PMCID: PMC10561192 DOI: 10.1096/fj.202201413r] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/20/2022] [Accepted: 01/09/2023] [Indexed: 02/17/2023]
Abstract
The epigenome of stem cells occupies a critical interface between genes and environment, serving to regulate expression through modification by intrinsic and extrinsic factors. We hypothesized that aging and obesity, which represent major risk factors for a variety of diseases, synergistically modify the epigenome of adult adipose stem cells (ASCs). Using integrated RNA- and targeted bisulfite-sequencing in murine ASCs from lean and obese mice at 5- and 12-months of age, we identified global DNA hypomethylation with either aging or obesity, and a synergistic effect of aging combined with obesity. The transcriptome of ASCs in lean mice was relatively stable to the effects of age, but this was not true in obese mice. Functional pathway analyses identified a subset of genes with critical roles in progenitors and in diseases of obesity and aging. Specifically, Mapt, Nr3c2, App, and Ctnnb1 emerged as potential hypomethylated upstream regulators in both aging and obesity (AL vs. YL and AO vs. YO), and App, Ctnnb1, Hipk2, Id2, and Tp53 exhibited additional effects of aging in obese animals. Furthermore, Foxo3 and Ccnd1 were potential hypermethylated upstream regulators of healthy aging (AL vs. YL), and of the effects of obesity in young animals (YO vs. YL), suggesting that these factors could play a role in accelerated aging with obesity. Finally, we identified candidate driver genes that appeared recurrently in all analyses and comparisons undertaken. Further mechanistic studies are needed to validate the roles of these genes capable of priming ASCs for dysfunction in aging- and obesity-associated pathologies.
Collapse
Affiliation(s)
- Shaojun Xie
- Bioinformatics Core, Purdue University, 1022 Young Hall, 155 S. Grant Street, West Lafayette, IN 47907
| | - Sulbha Choudhari
- Bioinformatics Core, Purdue University, 1022 Young Hall, 155 S. Grant Street, West Lafayette, IN 47907
- Advanced Biomedical Computational Science, Bioinformatics and Computational Science, Frederick National Laboratory for Cancer Research, 8560 Progress Drive, Frederick, MD 2170
| | - Chia-Lung Wu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, NY, 14611
| | - Karen Abramson
- Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701
| | - David Corcoran
- Genomic Analysis and Bioinformatics Shared Resource, Duke Center for Genomic and Computational Biology, 101 Science Drive, Duke University Medical Center Box 3382, Durham, NC 27708
- Lineberger Bioinformatics Core, 5200 Marsico Hall, University of North Carolina-Chapel Hill, Chapel Hill, NC 27516
| | - Simon G. Gregory
- Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701
- Department of Neurology, Duke University School of Medicine, 311 Research Drive, Durham, NC 27710
| | - Jyothi Thimmapuram
- Bioinformatics Core, Purdue University, 1022 Young Hall, 155 S. Grant Street, West Lafayette, IN 47907
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, 4515 McKinley Ave., St. Louis, MO 63110
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis Missouri 63110
| | - Dianne Little
- Departments of Basic Medical Sciences and Biomedical Engineering, Purdue University, 2186 Lynn Hall, 625 Harrison St, West Lafayette, IN 47907-2026
| |
Collapse
|
4
|
Zhang ZJ, Hou YK, Chen MW, Yu XZ, Chen SY, Yue YR, Guo XT, Chen JX, Zhou Q. A pH-responsive metal-organic framework for the co-delivery of HIF-2α siRNA and curcumin for enhanced therapy of osteoarthritis. J Nanobiotechnology 2023; 21:18. [PMID: 36650517 PMCID: PMC9847079 DOI: 10.1186/s12951-022-01758-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
The occurrence of osteoarthritis (OA) is highly correlated with the reduction of joint lubrication performance, in which persistent excessive inflammation and irreversible destruction of cartilage dominate the mechanism. The inadequate response to monotherapy methods, suboptimal efficacy caused by undesirable bioavailability, short retention, and lack of stimulus-responsiveness, are few unresolved issues. Herein, we report a pH-responsive metal-organic framework (MOF), namely, MIL-101-NH2, for the co-delivery of anti-inflammatory drug curcumin (CCM) and small interfering RNA (siRNA) for hypoxia inducible factor (HIF-2α). CCM and siRNA were loaded via encapsulation and surface coordination ability of MIL-101-NH2. Our vitro tests showed that MIL-101-NH2 protected siRNA from nuclease degradation by lysosomal escape. The pH-responsive MIL-101-NH2 gradually collapsed in an acidic OA microenvironment to release the CCM payloads to down-regulate the level of pro-inflammatory cytokines, and to release the siRNA payloads to cleave the target HIF-2α mRNA for gene-silencing therapy, ultimately exhibiting the synergetic therapeutic efficacy by silencing HIF-2α genes accompanied by inhibiting the inflammation response and cartilage degeneration of OA. The hybrid material reported herein exhibited promising potential performance for OA therapy as supported by both in vitro and in vivo studies and may offer an efficacious therapeutic strategy for OA utilizing MOFs as host materials.
Collapse
Affiliation(s)
- Zi-Jian Zhang
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Ying-Ke Hou
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Ming-Wa Chen
- grid.284723.80000 0000 8877 7471NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 People’s Republic of China
| | - Xue-Zhao Yu
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Si-Yu Chen
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Ya-Ru Yue
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Xiong-Tian Guo
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Jin-Xiang Chen
- grid.284723.80000 0000 8877 7471NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 People’s Republic of China
| | - Quan Zhou
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| |
Collapse
|
5
|
Yang H, Yang Z, Yu Z, Xiong C, Zhang Y, Zhang J, Huang Y, Zhou X, Li J, Xu N. SEMA6D, Negatively Regulated by miR-7, Contributes to C28/I2 chondrocyte's Catabolic and Anabolic Activities via p38 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9674221. [PMID: 35757507 PMCID: PMC9225841 DOI: 10.1155/2022/9674221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 05/16/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
Abstract
MiR-7 has been recognized as an osteoarthritis (OA-)-promoting factor, but the specific downstream pathway of miR-7 still remains unknown. Further investigation of the molecular regulatory mechanism of miR-7 might help develop a novel therapeutic method for OA. In this study, we revealed that Semaphorin 6D (SEMA6D) was a direct target gene of miR-7 and presented a negative regulatory relation with SEMA6D in vitro and in vivo. SEMA6D could improve OA in rat OA models, as indicated by H&E and Safranin O-Fast green staining, and also μCT analysis. Further evaluation of SEMA6D suggested that SEMA6D promotes the anabolism and reduces the catabolism of C28/I2 chondrocytes via inhibiting the activation of the p38 pathway. The present research illustrated that SEMA6D is a negatively regulatory factor of miR-7 and a pivotal mediator of catabolism and anabolism in C28/I2 chondrocytes. SEMA6D exerts its function via inhibiting the activation of the p38 pathway.
Collapse
Affiliation(s)
- Haoyu Yang
- Department of Orthopedics, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi 214000, China
| | - Zhicheng Yang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Zhentang Yu
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Chenwei Xiong
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yi Zhang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Junjie Zhang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Jin Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Nanwei Xu
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| |
Collapse
|
6
|
Sun X, Mi L, Du G, Sun C, He S. Platelet-rich plasma treatment alleviates osteoarthritis-related pain, inflammation, and apoptosis by upregulating the expression levels of microRNA-375 and microRNA-337. Immunopharmacol Immunotoxicol 2021; 44:87-98. [PMID: 34845965 DOI: 10.1080/08923973.2021.2007263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The present study was designed to determine the molecular mechanism by which platelet-rich plasma (PRP) acts on Osteoarthritis (OA) -related pain, inflammation, and apoptosis in vivo and in vitro. MATERIALS AND METHODS An in vivo OA model was established in rats using anterior cruciate ligament transection, and an in vitro OA model was created by treating chondrocytes with IL-1β. Then, the induced rats and chondrocytes were treated with PRP. Real-time PCR were used to examine the expression of micorRNAs (miRs) and mRNAs of inflammatory cytokines. WB were performed to detect the expression of apoptotic factors and Wnt/β-catenin signals. Structural damage of the cartilage and pain in OA rats were analyzed and represented by Mankin Score, OARSIS score, Tender threshold, and Thermal pain threshold. CCK-8 assay and flow cytometry were used to determine cell viability and apoptosis. RESULTS The expression levels of miR-337 and miR-375 were downregulated in the in vivo and vitro OA models; however, PRP treatment elevated their levels. miR-337 and miR-375 inhibition reversed the effects of PRP of reducing tenderness and thermal pain thresholds in OA rats. Moreover, PRP decreased the mRNA expression levels of MMP-13, Bax, and inflammatory factors, such as IL-1β, IL-18, and TNF-α, as well as increased the expression levels of collagen II and antiapoptotic Bcl-2. The decrease in inflammation and apoptosis was reversed by miR-337 and miR-375 inhibition, respectively. DISCUSSION AND CONCLUSIONS In conclusion, miR-337 and miR-375 are involved in PRP-delayed OA progression by affecting inflammation and apoptosis.
Collapse
Affiliation(s)
- Xuegang Sun
- Department of Orthopedic Surgery, The Second Hospital of Dalian Medical University, Liaoning, China
| | - Lidong Mi
- Department of Orthopedic Surgery, The Second Hospital of Dalian Medical University, Liaoning, China
| | - Guangyu Du
- Department of Orthopedic Surgery, The Second Hospital of Dalian Medical University, Liaoning, China
| | - Chuanxiu Sun
- Department of Orthopedic Surgery, The Second Hospital of Dalian Medical University, Liaoning, China
| | - Shengwei He
- Department of Orthopedic Surgery, The Second Hospital of Dalian Medical University, Liaoning, China
| |
Collapse
|
7
|
Wang Z, Zhou N, Wang W, Yu Y, Xia L, Li N. HDAC2 interacts with microRNA-503-5p to regulate SGK1 in osteoarthritis. Arthritis Res Ther 2021; 23:78. [PMID: 33750441 PMCID: PMC7941997 DOI: 10.1186/s13075-020-02373-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/13/2020] [Indexed: 01/15/2023] Open
Abstract
Background Osteoarthritis (OA) is a disabling joint disease that causes articular cartilage degeneration. It has been implicated that altered expression of histone deacetylase 2 (HDAC2) is found in patients with OA. However, the specific role of HDAC2 in the development of OA still remains enigmatic. Hence, we sought to characterize the functional relevance of HDAC2 in the development of OA. Methods Anterior cruciate ligament surgery was performed to generate the rat model of OA. Luciferase assay was performed to evaluate the relationship between microRNA-503-5p (miR-503-5p) and serum- and glucocorticoid-inducible kinase-1 (SGK1). Functional experiments were conducted to examine the functional significance of miR-503-5p, histone deacetylase 2 (HDAC2), and SGK1 on the progression of OA by determining proliferation, apoptosis, and expression of apoptosis-associated proteins and inflammatory cytokines. Results HDAC2 could inhibit miR-503-5p expression. SGK1 was the target gene of miR-503-5p. Upregulation of miR-503-5p or silencing of HDAC2 contributed to enhanced proliferation, suppressed apoptosis (reduced expression of Caspase-3 and Bax but elevated expression of Bcl2), and promoted inflammation in chondrocytes of OA rats. Conclusion In conclusion, our study demonstrated that HDAC2 could promote OA through miR-503-5p/SGK1 axis, which might function as a therapeutic target for OA treatment.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Eastern Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - Nan Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Eastern Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - Wengang Wang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Eastern Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - Yangke Yu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Eastern Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China
| | - Lei Xia
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Eastern Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China.
| | - Ning Li
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Eastern Jianshe Road, Zhengzhou, 450000, Henan Province, People's Republic of China.
| |
Collapse
|
8
|
Shao LT, Gou Y, Fang JK, Hu YP, Lian QQ, Zhang YY, Wang YD, Tian FM, Zhang L. Parathyroid hormone (1-34) ameliorates cartilage degeneration and subchondral bone deterioration in collagenase-induced osteoarthritis model in mice. Bone Joint Res 2020; 9:675-688. [PMID: 33101657 PMCID: PMC7563035 DOI: 10.1302/2046-3758.910.bjr-2020-0018.r1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Aims Parathyroid hormone (PTH) (1-34) exhibits potential in preventing degeneration in both cartilage and subchondral bone in osteoarthritis (OA) development. We assessed the effects of PTH (1-34) at different concentrations on bone and cartilage metabolism in a collagenase-induced mouse model of OA and examined whether PTH (1-34) affects the JAK2/STAT3 signalling pathway in this process. Methods Collagenase-induced OA was established in C57Bl/6 mice. Therapy with PTH (1-34) (10 μg/kg/day or 40 μg/kg/day) was initiated immediately after surgery and continued for six weeks. Cartilage pathology was evaluated by gross visual, histology, and immunohistochemical assessments. Cell apoptosis was analyzed by TUNEL staining. Microcomputed tomography (micro-CT) was used to evaluate the bone mass and the microarchitecture in subchondral bone. Results Enhanced matrix catabolism, increased apoptosis of chondrocytes in cartilage, and overexpressed JAK2/STAT3 and p-JAK2/p-STAT3 were observed in cartilage in this model. All of these changes were prevented by PTH (1-34) treatment, with no significant difference between the low-dose and high-dose groups. Micro-CT analysis indicated that bone mineral density (BMD), bone volume/trabecular volume (BV/TV), and trabecular thickness (Tb.Th) levels were significantly lower in the OA group than those in the Sham, PTH 10 μg, and PTH 40 μg groups, but these parameters were significantly higher in the PTH 40 μg group than in the PTH 10 μg group. Conclusion Intermittent administration of PTH (1-34) exhibits protective effects on both cartilage and subchondral bone in a dose-dependent manner on the latter in a collagenase-induced OA mouse model, which may be involved in regulating the JAK2/STAT3 signalling pathway. Cite this article: Bone Joint Res 2020;9(10):675–688.
Collapse
Affiliation(s)
- Li-Tao Shao
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China.,Medical Research Center, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, China
| | - Yu Gou
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Jia-Kang Fang
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, China
| | - Yun-Peng Hu
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, China
| | - Qiang-Qiang Lian
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, China
| | - Yu-Ying Zhang
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, China
| | - Yu-Dan Wang
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, China
| | - Fa-Ming Tian
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, China
| | - Liu Zhang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China.,Department of Orthopedic Surgery, Emergency General Hospital, Beijing, China
| |
Collapse
|
9
|
Comparative Analysis of the Occurrence and Role of CX3CL1 (Fractalkine) and Its Receptor CX3CR1 in Hemophilic Arthropathy and Osteoarthritis. J Immunol Res 2020; 2020:2932696. [PMID: 32884948 PMCID: PMC7455839 DOI: 10.1155/2020/2932696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Hemophilic arthropathy is characterized by recurrent bleeding episodes in patients with hemophilia leading to irreversible joint degeneration. The involvement of CX3CL1 (fractalkine) and its receptor CX3CR1 was observed in the pathogenesis of numerous arthritis-associated diseases. Taking this into account, we have presented a study investigating the role of the CX3CL1/CX3XR1 axis in the course of hemophilic arthropathy, including the CX3CL1-dependent expression of CD56+, CD68+, and CD31+ cells along with evaluation of articular cartilage and synovial membrane morphology. Methods The study was carried out using cases (n = 20) of end-stage hemophilic arthropathy with a severe type of hemophilia A and control cases (n = 20) diagnosed with osteoarthritis. The biofluids including blood serum and synovial fluid were obtained intraoperatively for the evaluation of CX3CL1 using the ELISA test. Tissue specimens including articular cartilage and synovial membrane were similarly collected during surgery and stained immunohistologically using selected antibodies including anti-CX3CR1, anti-CD56, anti-CD68, and anti-CD31. Additionally, the analysis included the assessment of articular cartilage, synovial membrane, and blood vessel morphology. Results In our study, we have documented increased average concentration of CX3CL1 in the blood serum of the study group (7.16 ± 0.53 ng/ml) compared to the control group (5.85 ± 0.70 ng/ml) without statistically significant difference in synovial fluid concentration at the same time. We have observed an increased macrophage presence with more marked proliferation and fibrosis of the synovial membrane in the study group. Remaining results such as expression of CX3CR1 presence of NK cells and larger surface area of blood vessels within the synovial membrane were noted also without statistical significance. Conclusions This study has demonstrated collective CX3CL1/CX3CR1 axis involvement in hemophilic arthropathy pathogenesis introducing new interesting diagnostics and a therapeutic target.
Collapse
|
10
|
Bellini M, Pest MA, Miranda-Rodrigues M, Qin L, Jeong JW, Beier F. Overexpression of MIG-6 in the cartilage induces an osteoarthritis-like phenotype in mice. Arthritis Res Ther 2020; 22:119. [PMID: 32430054 PMCID: PMC7236969 DOI: 10.1186/s13075-020-02213-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common form of arthritis and characterized by degeneration of the articular cartilage. Mitogen-inducible gene 6 (Mig-6) has been identified as a negative regulator of the epidermal growth factor receptor (EGFR). Cartilage-specific Mig-6 knockout (KO) mice display increased EGFR signaling, an anabolic buildup of the articular cartilage, and formation of chondro-osseous nodules. Since our understanding of the EGFR/Mig-6 network in the cartilage remains incomplete, we characterized mice with cartilage-specific overexpression of Mig-6 in this study. METHODS Utilizing knee joints from cartilage-specific Mig-6-overexpressing (Mig-6over/over) mice (at multiple time points), we evaluated the articular cartilage using histology, immunohistochemical staining, and semi-quantitative histopathological scoring (OARSI) at multiple ages. MicroCT analysis was employed to examine skeletal morphometry, body composition, and bone mineral density. RESULTS Our data show that cartilage-specific Mig-6 overexpression did not cause any major developmental abnormalities in the articular cartilage, although Mig-6over/over mice have slightly shorter long bones compared to the control group. Moreover, there was no significant difference in bone mineral density and body composition in any of the groups. However, our results indicate that Mig-6over/over male mice show accelerated cartilage degeneration at 12 and 18 months of age. Immunohistochemistry for SOX9 demonstrated that the number of positively stained cells in Mig-6over/over mice was decreased relative to controls. Immunostaining for MMP13 appeared increased in areas of cartilage degeneration in Mig-6over/over mice. Moreover, staining for phospho-EGFR (Tyr-1173) and lubricin (PRG4) was decreased in the articular cartilage of Mig-6over/over mice. CONCLUSION Overexpression of Mig-6 in the articular cartilage causes no major developmental phenotype; however, these mice develop earlier OA during aging. These data demonstrate that Mig-6/EGFR pathways are critical for joint homeostasis and might present a promising therapeutic target for OA.
Collapse
Affiliation(s)
- Melina Bellini
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
| | - Michael A Pest
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
| | - Manuela Miranda-Rodrigues
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Frank Beier
- Department of Physiology and Pharmacology, Western University, London, ON, Canada.
- Western University Bone and Joint Institute, London, ON, Canada.
- Children's Health Research Institute, London, ON, Canada.
| |
Collapse
|
11
|
Jun Z, Xinmeng J, Yue L, Zhi W, Yan Z, Tieyi Y, Jiangan T. Jumonji domain containing-3 (JMJD3) inhibition attenuates IL-1β-induced chondrocytes damage in vitro and protects osteoarthritis cartilage in vivo. Inflamm Res 2020; 69:657-666. [PMID: 32394143 DOI: 10.1007/s00011-020-01356-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 04/01/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES This study aimed to explore the effects and relative mechanism of JMJD3 on knee osteoarthritis (OA). METHODS In this study, we first analyzed the expression of JMJD3 in OA cartilage using western blot and immunohistochemistry. In an in vitro study, the effects of GSK-J4, JMJD3 inhibitor, on ATDC-5 chondrocytes were evaluated by CCK-8 assay. Real-time PCR and western blot were used to examine the inhibitory effect of GSK-J4 on the inflammation and ECM degradation of chondrocytes. NF-κB p65 phosphorylation and nuclear translocation were measured by western blot and immunofluorescence. In the animal study, twenty mice were randomized into four experimental groups: sham group, DMM-induced OA + DMSO group, OA + low-dose GSK-J4 group, and OA + high-dose GSK-J4 group. After the treatment, hematoxylin-eosin and safranin O/fast green staining were used to evaluate cartilage degradation of knee joint, with OARSI scores for quantitative assessment of cartilage damage. RESULTS Our results revealed that JMJD3 was overexpressed in OA cartilage and GSK-J4 could suppress the IL-1β-induced production of pro-inflammatory cytokines and catabolic enzymes, including IL-6, IL-8, MMP-9 and ADAMTS-5. Consistent with these findings, GSK-J4 could inhibit IL-1β-induced degradation of collagen II and aggrecan. Mechanistically, GSK-J4 dramatically suppressed IL-1β-stimulated NF-κB signal pathway activation. In vivo, GSK-J4 prevented cartilage damage in mouse DMM-induced OA model. CONCLUSIONS This study elucidates the important role of JMJD3 in cartilage degeneration in OA, and our results indicate that JDJM3 may become a novel therapeutic target in OA therapy.
Collapse
Affiliation(s)
- Zhou Jun
- Department of Orthopaedics, Gongli Hospital, The Second Military Medical University, No. 219, Miaopu Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Jin Xinmeng
- Department of Orthopaedics, Gongli Hospital, The Second Military Medical University, No. 219, Miaopu Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Liu Yue
- Department of Orthopaedics, Gongli Hospital, The Second Military Medical University, No. 219, Miaopu Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Wang Zhi
- Department of Orthopaedics, Gongli Hospital, The Second Military Medical University, No. 219, Miaopu Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Zhang Yan
- Department of Orthopaedics, Gongli Hospital, The Second Military Medical University, No. 219, Miaopu Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Yang Tieyi
- Department of Orthopaedics, Gongli Hospital, The Second Military Medical University, No. 219, Miaopu Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Tang Jiangan
- Department of Orthopaedics, Gongli Hospital, The Second Military Medical University, No. 219, Miaopu Road, Pudong New Area, Shanghai, 200135, People's Republic of China.
| |
Collapse
|
12
|
Ukita M, Matsushita K, Tamura M, Yamaguchi T. Histone H3K9 methylation is involved in temporomandibular joint osteoarthritis. Int J Mol Med 2019; 45:607-614. [PMID: 31894302 DOI: 10.3892/ijmm.2019.4446] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/11/2019] [Indexed: 11/05/2022] Open
Abstract
The morbidity of temporomandibular joint osteoarthritis (TMJOA) increases with age. Condylar articular cartilage degradation, which causes TMJOA, is known to be involved in articular chondrocyte metabolic imbalances in the temporomandibular joint (TMJ) and in other joints of the body. Epigenetic regulation, such as the chemical modification of DNA and histones, is implicated in cartilage homeostasis. However, few studies have been conducted on the epigenetic regulation of condylar articular cartilage degradation. The present study investigated the regulation of histone H3 lysine 9 (H3K9) methylation and its effects on the pathogenesis of degenerative TMJ cartilage disorders. The histone H3K9 methylation level was decreased in degenerated condylar articular cartilage in aged mice. Treatment with chaetocin (a selective H3K9 methylation inhibitor) reduced cell viability and promoted caspase‑3/7 activity in ATDC5 mouse chondroprogenitor cells. The inhibition of H3K9 methylation increased matrix metalloproteinase (Mmp)1 and Mmp13 mRNA expression in these cells. Furthermore, the expression levels of Sox9 and collagen α1(II) (Col2a1) mRNA, which are anabolic factors for chondrogenic differentiation, were also decreased by treatment with chaetocin, which is an inhibitor of histone methyltransferases. These results indicated that histone H3K9 methylation regulates chondrocyte homeostasis in terms of cell growth, apoptosis and gene expression, and highlighted a possible future therapy option for TMJOA.
Collapse
Affiliation(s)
- Mayumi Ukita
- Crown and Bridge Prosthodontics, Department of Oral Functional Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8586, Japan
| | - Kenji Matsushita
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi 474‑8511, Japan
| | - Masato Tamura
- Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8586, Japan
| | - Taihiko Yamaguchi
- Crown and Bridge Prosthodontics, Department of Oral Functional Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8586, Japan
| |
Collapse
|
13
|
Du X, Ouyang H. [Correlation between histone methylation level and pathological development of osteoarthritis]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:682-687. [PMID: 31955544 PMCID: PMC8800784 DOI: 10.3785/j.issn.1008-9292.2019.12.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/03/2019] [Indexed: 06/10/2023]
Abstract
Osteoarthritis is the most common degenerative cartilage disease. A large number of studies have shown the close association between epigenetics and osteoarthritis. Histone methylation is a type of epigenetic modification, and the link between histone methylation and osteoarthritis has also been revealed. In this article, we summarize the correlation between methylation levels of different histones and osteoarthritis in an attempt to explore the changes and regulation mechanisms of histone methylation in osteoarthritis. It has been shown that there are possible relations between the methylation levels of different amino acids on histone H3 and the pathological development of osteoarthritis; specifically, the rise of methylation level at the lysine 4 would aggravate the pathological development of osteoarthritis, while the the pattern of lysine 9 and 27 would be the opposite. These results indicate the possible existence of a complex network of histone methylation modifications. And the specific regulation of histone methylation levels in different positions may delay or prevent the occurrence and development of osteoarthritis.
Collapse
Affiliation(s)
- Xiaotian Du
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
- Zhejiang University-University of Edinburgh Institute, International Campus of Zhejiang University, Haining 314400, Zhejiang Province, China
| |
Collapse
|
14
|
Cai C, Min S, Yan B, Liu W, Yang X, Li L, Wang T, Jin A. MiR-27a promotes the autophagy and apoptosis of IL-1β treated-articular chondrocytes in osteoarthritis through PI3K/AKT/mTOR signaling. Aging (Albany NY) 2019; 11:6371-6384. [PMID: 31460867 PMCID: PMC6738432 DOI: 10.18632/aging.102194] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a common degenerative joint disorder, which involves articular cartilage degeneration as well as joint inflammatory reactions. The recent studies have identified microRNA (miRNA) as one of the epigenetic mechanisms for the regulation of gene expression. Here we aim to reveal the role of miRNA in the regulation of gene expression in articular chondrocytes and its significance in the OA pathogenesis. In the present study, miRNA profiling was performed using OA cartilage and normal healthy cartilage tissues. As compared to their levels in normal cells and tissues, miR-27a expression was found to be upregulated in OA cartilage and IL-1β-treated articular chondrocytes. TUNEL staining, as well as flow cytometry with Annexin V-FITC/PI double labeling indicated that miR-27a inhibition reduced the apoptosis of IL-1β-treated articular chondrocytes. Bioinformatics prediction and the dual-luciferase reporter assay indicated that miR-27a targeted the 3'-UTR of the PI3K gene to silence it. The PI3K mRNA level in OA cartilage and IL-1β-treated articular chondrocytes was also downregulated, comparing with normal cells and tissues. Transfection of chondrocytes transfected with the miR-27a inhibitor upregulated the PI3K expression. This study demonstrated miR-27a is a regulator of the PI3K-Akt-mTOR axis in human chondrocytes and could participate in OA pathogenesis.
Collapse
Affiliation(s)
- Chen Cai
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shaoxiong Min
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Yan
- Department of Spine Surgery, The Third Affliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Wen Liu
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao Yang
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Liuxun Li
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Wang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, China
| | - Anmin Jin
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Grote C, Reinhardt D, Zhang M, Wang J. Regulatory mechanisms and clinical manifestations of musculoskeletal aging. J Orthop Res 2019; 37:1475-1488. [PMID: 30919498 PMCID: PMC9202363 DOI: 10.1002/jor.24292] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/13/2019] [Indexed: 02/04/2023]
Abstract
Aging is the strongest risk factor for degenerative bone and joint diseases. Clinical therapies for age-related musculoskeletal disorders face significant challenges as their pathogenic mechanisms remain largely unclear. This review article focuses on the recent advances in the understanding of regulatory mechanisms of musculoskeletal aging and their clinical relevance. We begin with the prevalence and socioeconomic impacts of major age-related musculoskeletal disorders such as sarcopenia, osteoporosis, osteoarthritis, and degenerative tendinopathy. The current understanding of responsible biological mechanisms involved in general aging is then summarized. Proposed molecular, cellular, and biomechanical mechanisms relevant to the clinical manifestations of aging in the musculoskeletal system are discussed in detail, with a focus on the disorders affecting muscle, bone, articular cartilage, and tendon. Although musculoskeletal aging processes share many common pathways with the aging of other body systems, unique molecular and cellular mechanisms may be involved in the aging processes of musculoskeletal tissues. Advancements in the understanding of regulatory mechanisms of musculoskeletal aging may promote the development of novel treatments for age-related musculoskeletal disorders. Finally, future research directions for major musculoskeletal tissues including functional interaction between the tissues and their clinical relevance to age-related musculoskeletal disorders are highlighted in the Future Prospects section. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1475-1488, 2019.
Collapse
Affiliation(s)
- Caleb Grote
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Daniel Reinhardt
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mingcai Zhang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
16
|
Bei M, Tian F, Liu N, Zheng Z, Cao X, Zhang H, Wang Y, Xiao Y, Dai M, Zhang L. A Novel Rat Model of Patellofemoral Osteoarthritis Due to Patella Baja, or Low-Lying Patella. Med Sci Monit 2019; 25:2702-2717. [PMID: 30979862 PMCID: PMC6476235 DOI: 10.12659/msm.915018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Patella baja, or patella infera, consists of a low-lying patella that results in a limited range of motion, joint pain, and crepitations. Patellofemoral joint osteoarthritis (PFJOA) is a subtype OA of the knee. This study aimed to develop a reproducible and reliable rat model of PFJOA. Material/Methods Three-month-old female Sprague-Dawley rats (n=24) included a baseline group (n=8) that were euthanized at the beginning of the study. The sham group (n=8), and the patella ligament shortening (PLS) group (n=8) were euthanized and evaluated at ten weeks. The PLS model group (n=8) underwent insertion of a Kirschner wire under the patella tendon to induce patella baja. At ten weeks, the sham group and the PLS group were compared using X-ray imaging, macroscopic appearance, histology, immunohistochemistry, TUNEL staining for apoptosis, and micro-computed tomography (micro-CT). The patella height was determined using the modified Insall-Salvati (MIS) ratio. Results The establishment of the rat model of patella baja in the PLS group at ten weeks was confirmed by X-ray. In the PLS group, patella volume, sagittal length, and cross-sectional area were significantly increased compared with the sham group. The PFJ showed typical lesions of OA, confirmed macroscopically and histologically. Compared with the sham group, in the rat model of PFJOA, there was increased cell apoptosis, and immunohistochemistry showed increased expression of biomarkers of osteoarthritis, compared with the sham group. Conclusions A rat model of PFJOA was developed that was confirmed by changes in cartilage and subchondral bone.
Collapse
Affiliation(s)
- Mingjian Bei
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Faming Tian
- Meical Research Center, North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Ning Liu
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Zhiyuan Zheng
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Xuehui Cao
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Hongfei Zhang
- Meical Research Center, North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Yudan Wang
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Yaping Xiao
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China (mainland)
| | - Muwei Dai
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Liu Zhang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Department of Orthopedic Surgery, Meitan General Hospital, Beijing, China (mainland)
| |
Collapse
|
17
|
Zhang M, Theleman JL, Lygrisse KA, Wang J. Epigenetic Mechanisms Underlying the Aging of Articular Cartilage and Osteoarthritis. Gerontology 2019; 65:387-396. [PMID: 30970348 PMCID: PMC9150844 DOI: 10.1159/000496688] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 01/08/2019] [Indexed: 10/29/2023] Open
Abstract
Aging is a progressive and complicated bioprocess with overall decline in physiological function. Osteoarthritis (OA) is the most common joint disease in middle-aged and older populations. Since the prevalence of OA increases with age and breakdown of articular cartilage is its major hallmark, OA has long been thought of as "wear and tear" of joint cartilage. Nevertheless, recent studies have revealed that changes in the chondrocyte function and matrix components may reduce the material properties of articular cartilage and predispose the joint to OA. The aberrant gene expression in aging articular cartilage that is regulated by various epigenetic mechanisms plays an important role in age-related OA pathogenesis. This review begins with an introduction to the current understanding of epigenetic mechanisms, followed by mechanistic studies on the aging of joint tissues, epigenetic regulation of age-dependent gene expression in articular cartilage, and the significance of epigenetic mechanisms in OA pathogenesis. Our recent findings on age-dependent expression of 2 transcription factors, nuclear factor of activated T cell 1 (NFAT1) and SOX9, and their roles in the formation and aging of articular cartilage are summarized in the review. Chondrocyte dysfunction in aged mice, which is mediated by epigenetically regulated spontaneous reduction of NFAT1 expression in articular cartilage, is highlighted as an important advance in epigenetics and cartilage aging. Potential therapeutic strategies for age-related cartilage degeneration and OA using epigenetic molecular tools are discussed at the end.
Collapse
Affiliation(s)
- Mingcai Zhang
- Department of Orthopedic Surgery, Harrington Laboratory for Molecular Orthopedics, Kansas City, Kansas, USA
- Department of Medicine, Clinical Immunology and Rheumatology, Kansas City, Kansas, USA
| | - Justin L Theleman
- Department of Orthopedic Surgery, Harrington Laboratory for Molecular Orthopedics, Kansas City, Kansas, USA
| | - Katherine A Lygrisse
- Department of Orthopedic Surgery, Harrington Laboratory for Molecular Orthopedics, Kansas City, Kansas, USA
| | - Jinxi Wang
- Department of Orthopedic Surgery, Harrington Laboratory for Molecular Orthopedics, Kansas City, Kansas, USA,
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA,
| |
Collapse
|
18
|
Song D, Qi W, Lv M, Yuan C, Tian K, Zhang F. Combined bioinformatics analysis reveals gene expression and DNA methylation patterns in osteoarthritis. Mol Med Rep 2018; 17:8069-8078. [PMID: 29658578 DOI: 10.3892/mmr.2018.88.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 02/15/2018] [Indexed: 01/03/2025] Open
Abstract
Osteoarthritis (OA) is a common type of arthritis, which may cause pain and disability. Alterations in gene expression and DNA methylation have been proven to be associated with the development of OA. The aim of the present study was to identify potential therapeutic targets and associated processes for OA via the combined analysis of gene expression and DNA methylation datasets. The gene expression and DNA methylation profiles were obtained from the Gene Expression Omnibus, and differentially expressed genes (DEGs) and differentially methylated sites (DMSs) were identified in the present study, using R programming software. The enriched functions of DEGs and DMSs were obtained via the Database for Annotation, Visualization and Integrated Discovery. Finally, cross analysis of DEGs and DMSs was performed to identify genes that exhibited differential expression and methylation simultaneously. The protein‑protein interaction (PPI) network of overlaps between DEGs and DMSs was obtained using the Human Protein Reference Database; the topological properties of PPI network overlaps were additionally obtained. Hub genes in the PPI network were further confirmed via reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). The results of the present study revealed that the majority of DEGs and DMSs were upregulated and hypomethylated in patients with OA, respectively. DEGs and DMSs were primarily involved in inflammatory, immune and gene expression regulation‑associated processes and pathways. Cross analysis revealed 30 genes that exhibited differential expression and methylation in OA simultaneously. Topological analysis of the PPI network revealed that numerous genes, including G protein subunit α1 (GNAI1), runt related transcription factor 2 (RUNX2) and integrin subunit β2 (ITGB2), may be involved in the development of OA. Additionally, RT‑qPCR analysis of GNAI1, RUNX2 and ITGB2 provided further confirmation. Numerous known and novel therapeutic targets were obtained via network analysis. The results of the present study may be beneficial for the diagnosis and treatment of OA.
Collapse
Affiliation(s)
- Delei Song
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Wei Qi
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Ming Lv
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Chun Yuan
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Kangsong Tian
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Feng Zhang
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
19
|
Song D, Qi W, Lv M, Yuan C, Tian K, Zhang F. Combined bioinformatics analysis reveals gene expression and DNA methylation patterns in osteoarthritis. Mol Med Rep 2018; 17:8069-8078. [PMID: 29658578 PMCID: PMC5983981 DOI: 10.3892/mmr.2018.8874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 02/15/2018] [Indexed: 12/30/2022] Open
Abstract
Osteoarthritis (OA) is a common type of arthritis, which may cause pain and disability. Alterations in gene expression and DNA methylation have been proven to be associated with the development of OA. The aim of the present study was to identify potential therapeutic targets and associated processes for OA via the combined analysis of gene expression and DNA methylation datasets. The gene expression and DNA methylation profiles were obtained from the Gene Expression Omnibus, and differentially expressed genes (DEGs) and differentially methylated sites (DMSs) were identified in the present study, using R programming software. The enriched functions of DEGs and DMSs were obtained via the Database for Annotation, Visualization and Integrated Discovery. Finally, cross analysis of DEGs and DMSs was performed to identify genes that exhibited differential expression and methylation simultaneously. The protein‑protein interaction (PPI) network of overlaps between DEGs and DMSs was obtained using the Human Protein Reference Database; the topological properties of PPI network overlaps were additionally obtained. Hub genes in the PPI network were further confirmed via reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). The results of the present study revealed that the majority of DEGs and DMSs were upregulated and hypomethylated in patients with OA, respectively. DEGs and DMSs were primarily involved in inflammatory, immune and gene expression regulation‑associated processes and pathways. Cross analysis revealed 30 genes that exhibited differential expression and methylation in OA simultaneously. Topological analysis of the PPI network revealed that numerous genes, including G protein subunit α1 (GNAI1), runt related transcription factor 2 (RUNX2) and integrin subunit β2 (ITGB2), may be involved in the development of OA. Additionally, RT‑qPCR analysis of GNAI1, RUNX2 and ITGB2 provided further confirmation. Numerous known and novel therapeutic targets were obtained via network analysis. The results of the present study may be beneficial for the diagnosis and treatment of OA.
Collapse
Affiliation(s)
- Delei Song
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Wei Qi
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Ming Lv
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Chun Yuan
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Kangsong Tian
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Feng Zhang
- Trauma Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
20
|
Raman S, FitzGerald U, Murphy JM. Interplay of Inflammatory Mediators with Epigenetics and Cartilage Modifications in Osteoarthritis. Front Bioeng Biotechnol 2018; 6:22. [PMID: 29594113 PMCID: PMC5861204 DOI: 10.3389/fbioe.2018.00022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 02/22/2018] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA), a degenerative disease of diarthrodial joints, is influenced by mechanical and inflammatory factors with aging, obesity, chronic injuries, and secondary diseases thought to be major factors driving the process of articular cartilage degeneration. Chondrocytes, the cellular component of cartilage, reside in an avascular environment and normally have limited potential to replicate. However, extrinsic factors such as injury to the joint or intrinsic alterations to the chondrocytes themselves can lead to an altered phenotype and development of OA. Synovial inflammation is also a pivotal element of the osteoarthritic, degenerative process: influx of pro-inflammatory cytokines and production of matrix metalloproteinases accelerate advanced cellular processes such as synovitis and cartilage damage. As well as a genetic input, recent data have highlighted epigenetic factors as contributing to disease. Studies conducted over the last decade have focused on three key aspects in OA; inflammation and the immune response, genome-wide association studies that have identified important genes undergoing epigenetic modifications, and finally how chondrocytes transform in their function during development and disease. Data highlighted here have identified critical inflammatory genes involved in OA and how these factors impact chondrocyte hypertrophy in the disease. This review also addresses key inflammatory factors in synovial inflammation, epigenetics, and chondrocyte fate, and how agents that inhibit epigenetic mechanisms like DNA methylation and histone modifications could aid in development of long-term treatment strategies for the disease.
Collapse
Affiliation(s)
- Swarna Raman
- Orthobiology, Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Una FitzGerald
- School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - J Mary Murphy
- Orthobiology, Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
21
|
Zhi LQ, Yao SX, Liu HL, Li M, Duan N, Ma JB. Hydroxytyrosol inhibits the inflammatory response of osteoarthritis chondrocytes via SIRT6-mediated autophagy. Mol Med Rep 2017; 17:4035-4042. [PMID: 29286133 DOI: 10.3892/mmr.2017.8353] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 11/24/2017] [Indexed: 11/05/2022] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease. Inflammation may exaggerate the catabolism and degeneration in the pathogenesis of OA. Hydroxytyrosol (HT) has been used in the management of inflammatory diseases. In addition, reports have revealed that autophagy was a therapeutic target of diseases caused by inflammation. Sirtuin 6 (SIRT6) has also been demonstrated to prevent OA development by reducing both the inflammatory response and chondrocyte senescence. However, the roles of SIRT6 and autophagy in cartilage and its underlying anti‑inflammatory mechanism are unknown. Therefore, the present study aimed to determine the effects of HT on autophagy and inflammation in chondrocytes, and clarify whether HT regulates the inflammatory response through SIRT6‑mediated autophagy. The expression of protein and mRNA were determined by western blot analysis and reverse transcription‑quantitative polymerase chain reaction. The production of cytokines was detected by ELISA. It was demonstrated that HT inhibited the levels of interleukin (IL)‑1β and IL‑6 in tumor necrosis factor (TNF)‑α‑stimulated chondrocytes in a concentration‑dependent manner. In addition, HT promoted cell autophagy and increased the mRNA and protein expression levels of SIRT6 in chondrocytes stimulated with TNF-α. Autophagy inhibitor 3-methyladenine or knockdown of SIRT6 decreased the inhibitory effects of HT on the inflammatory response in chondrocytes. In addition, knockdown of SIRT6 attenuated the expression of microtubule-associated protein 1A/1B‑light chain 3 and Beclin1 in chondrocytes. Overall, these findings suggested that HT inhibits the inflammatory response of chondrocytes through SIRT6‑mediated autophagy. The present study provided a new drug target for the clinical treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Li-Qiang Zhi
- Department of Joint Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| | - Shu-Xin Yao
- Department of Joint Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| | - Hong-Liang Liu
- Department of Orthopaedic Trauma, Hong‑Hui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| | - Meng Li
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ning Duan
- Department of Traumatic Osteopathic, Hong‑Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Jian-Bing Ma
- Department of Joint Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| |
Collapse
|
22
|
Abstract
Although the potential effect of aberrant expression of catabolic and
anabolic genes on the development of osteoarthritis (OA) is well-documented, the
regulatory mechanism for the expression of these genes in articular chondrocytes
remains to be elucidated. The recent advances in epigenetic studies have
identified microRNA (miRNA) as one of the epigenetic mechanisms for the
regulation of gene expression. This mini review highlights the role of miRNA in
the regulation of gene expression in articular chondrocytes and its significance
in the pathogenesis of OA, with a discussion on the potential of miRNA as a new
biomarker and therapeutic target for OA. Further investigations are required to
determine the specificity, sensitivity, and efficacy of miRNA for clinical
applications.
Collapse
Affiliation(s)
- Mingcai Zhang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kate Lygrisse
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
23
|
Gabay O, Clouse KA. Epigenetics of cartilage diseases. Joint Bone Spine 2016; 83:491-4. [DOI: 10.1016/j.jbspin.2015.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/28/2015] [Indexed: 02/06/2023]
|
24
|
Jiang LB, Lee S, Wang Y, Xu QT, Meng DH, Zhang J. Adipose-derived stem cells induce autophagic activation and inhibit catabolic response to pro-inflammatory cytokines in rat chondrocytes. Osteoarthritis Cartilage 2016; 24:1071-81. [PMID: 26778531 DOI: 10.1016/j.joca.2015.12.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/18/2015] [Accepted: 12/27/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Adipose-derived stem cells (ADSCs) have been demonstrated to have an anti-apoptosis effect on chondrocytes; However, their effect on autophagic activation remains unclear. We sought to explore whether ADSCs can activate autophagy and inhibit IL-1β- and lipopolysaccharide (LPS)-induced catabolism in chondrocytes. METHODS ADSCs and chondrocytes were collected from SD rats. The biologic characteristics of ADSCs were analyzed by flow cytometric analysis, Oil red O and Alizarin Red staining. Autophagic level and autophagic flux were revealed by Western blotting for LC3-II and SQSTM1/P62, MDC (monodansylcadaverine) staining and mRFP-GFP-LC3 analysis. The mTOR pathway was investigated by Western blotting for p-mTOR. The mRNA level of matrix metalloproteinases (MMPs) and thrombospondin motifs (ADAMTSs) was detected by real-time PCR. RESULTS The typical surface markers and differentiation potentials of ADSCs were proved. ADSCs enhanced the expression of LC3-II/LC3-I and reduced SQSTM1 levels in IL-1β-induced chondrocytes after 24 and 48 h co-culturing and in LPS-induced chondrocytes after 48 h co-culturing respectively. mRFP-GFP-LC3 analysis suggested that autophagosomes and autolysosomes were formed earlier in IL-1β-treated chondrocytes than in LPS-treated chondrocytes. Bafilomycin A1 treatment further increased the LC3-II/LC3-I level in chondrocytes in co-culture with ADSCs. The mTOR pathway was inhibited in the chondrocytes in co-culture with ADSCs. Finally, ADSCs inhibited the increase of MMPs and ADAMTSs in chondrocytes induced by IL-1β and LPS. CONCLUSIONS ADSCs seem able to activate autophagy and inhibit catabolism in chondrocytes in an inflammation environment, and the mTOR pathway might be involved in the autophagy activation.
Collapse
Affiliation(s)
- Li-Bo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Soomin Lee
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qin-Tong Xu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - De-Hua Meng
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Lee S, Nemeño JGE, Lee JI. Repositioning Bevacizumab: A Promising Therapeutic Strategy for Cartilage Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:341-357. [PMID: 26905221 DOI: 10.1089/ten.teb.2015.0300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug discovery and development has been garnering an increasing trend of research due to the growing incidence of the diverse types of diseases. Recently, drug repositioning, also known as drug repurposing, has been emerging parallel to cancer and tissue engineering studies. Drug repositioning involves the application of currently approved or even abandoned drugs as alternative treatments to other diseases or as biomaterials in other fields including cell therapy and tissue engineering. In this review, the advancement of the antiangiogenesis drugs that were used as treatment for cancer and other diseases, with particular focus on bevacizumab, will be described. This will include an overview of the nature and progression of osteoarthritis (OA), one of the leading global degenerative diseases that cause morbidity, and the development of its therapeutic strategies. In addition, this will also feature the nonsteroidal anti-inflammatory drugs that are commonly prescribed for OA and the benefits of repositioning bevacizumab as alternative treatments for other diseases and as biomaterials for cartilage regeneration. To date, a few number of studies, employing different modes of administration and varying dosages in diverse animal models, have shown that bevacizumab can be used as a signal and can promote both in vitro and in vivo cartilage regeneration. However, other antiangiogenesis drugs and their effects in chondrogenesis and cartilage regeneration are also worth investigating.
Collapse
Affiliation(s)
- Soojung Lee
- 1 Regenerative Medicine Laboratory, Department of Biomedical Science and Technology, Center for Stem Cell Research, Institute of Biomedical Science & Technology, Konkuk University , Seoul, Republic of Korea
| | - Judee Grace E Nemeño
- 1 Regenerative Medicine Laboratory, Department of Biomedical Science and Technology, Center for Stem Cell Research, Institute of Biomedical Science & Technology, Konkuk University , Seoul, Republic of Korea
| | - Jeong Ik Lee
- 1 Regenerative Medicine Laboratory, Department of Biomedical Science and Technology, Center for Stem Cell Research, Institute of Biomedical Science & Technology, Konkuk University , Seoul, Republic of Korea.,2 Deparment of Veterinary Medicine, College of Veterinary Medicine, Konkuk University , Seoul, Republic of Korea
| |
Collapse
|