1
|
Chen Y, Lin J, Lin J, Gao T, Cai Q, Zhang C, Zhu H, Shen L, Wang Q. Comparison of the effectiveness of intra-infrapatellar fat pad and intra-articular glucocorticoid injection in knee osteoarthritis patients with Hoffa's synovitis: protocol for a multicentre randomised controlled trial. BMJ Open 2025; 15:e087785. [PMID: 39880431 PMCID: PMC11781145 DOI: 10.1136/bmjopen-2024-087785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION The infrapatellar fat pad and synovium are the sites of immune cell infiltration and the origin of proinflammation. Studies have shown that Hoffa's synovitis may be a sign of early-stage osteoarthritis (OA). However, there have been no effective interventions specifically for Hoffa's synovitis. METHODS AND ANALYSIS We will conduct a multicentre, multi-blind (participant, physician, outcome assessor and data analyst blinded) randomised controlled trial to compare the effectiveness of an intra-infrapatellar fat glucocorticoid versus an intra-articular injection for Hoffa's synovitis in patients with knee OA. We will recruit 236 knee OA patients with Hoffa's synovitis at outpatient clinics in three centres. We will randomly allocate them to two groups in a 1:1 ratio. One group will receive ultrasound-guided injection of 40 mg (1 mL) triamcinolone acetonide into the infrapatellar fat pad; the other group will receive ultrasound-guided injection of 40 mg (1 mL) triamcinolone acetonide into the knee joint cavity. All patients will be followed up at 2, 4, 8, 12 and 24 weeks after the injection. Primary outcomes are (1) Hoffa's synovitis improvement rate, measured with the MRI Osteoarthritis Knee Score system (superiority outcome) at 24 weeks and (2) pain intensity, measured with the Western Ontario and McMasters University Osteoarthritis Index (WOMAC) at 2 weeks post-injection. Secondary outcomes include Hoffa's synovitis score at 2 weeks post-injection, pain intensity with the numerical rating scale, WOMAC questionnaire score improvements (function, joint stiffness and total score), improvement rates in effusion synovitis at 2 and 24 weeks, articular cartilage thickness changes at 2 and 24 weeks, Intermittent and Constant Osteoarthritis Pain score, quality of life measured with the EuroQol-5D, OARSI-OMERACT response indicators, co-interventions and side effects at 2, 4, 8, 12 and 24 weeks. ETHICS AND DISSEMINATION Ethical approval has been granted by the Medical Ethics Committee of the Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (2023-178). Written informed consent will be obtained from all patients prior to data collection. The findings of this research will be shared through presentations at academic conferences and publications in peer-reviewed journals. TRIAL REGISTRATION NUMBER ChiCTR2400080474.
Collapse
Affiliation(s)
- Yiwei Chen
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junqing Lin
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiali Lin
- Shanghai Medical college, Fudan university, Shanghai, China
| | - Tao Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianying Cai
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyi Zhu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longxiang Shen
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuke Wang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Sun YY, Wang ML, Zhao RH, Hao MY, Song S, Zhou Y, Zhang N. The Efficacy and Brain Network Mechanism of Acupuncture for Knee Osteoarthritis: A Study Protocol for Randomized Controlled Neuroimaging Trial. J Pain Res 2025; 18:391-404. [PMID: 39872091 PMCID: PMC11771175 DOI: 10.2147/jpr.s500024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/07/2025] [Indexed: 01/29/2025] Open
Abstract
Purpose Knee osteoarthritis (KOA) is a prevalent degenerative bone and joint disease observed in clinical practice. While acupuncture has demonstrated efficacy in treating KOA, the central mechanisms underlying its effects remain ambiguous. Recently, functional magnetic resonance imaging (fMRI) has been extensively applied in studying the brain mechanisms of acupuncture analgesia. Currently, network analysis based on brain connectomics is a focal point in acupuncture imaging research. Therefore, this study uses KOA as the research vehicle, focuses on the abnormal connectivity patterns of brain functional networks, and integrates clinical pain assessments to thoroughly investigate the central mechanisms of acupuncture therapeutic effects on KOA. Patients and Methods In this parallel, randomized, sham-controlled neuroimaging trial, 60 KOA patients will be randomly divided into the acupuncture group and sham acupuncture group in a 1:1 ratio, treated three times weekly for a total of 12 sessions. Patients will undergo clinical symptom assessments and cranial fMRI scans at baseline (-1-0 weeks), post-treatment (4 weeks), and at the follow-up (16 weeks). Forty healthy subjects will be recruited for observation, with a single MRI scan conducted only at baseline (-1-0 week). The primary efficacy indicator will be the change in NRS score after four weeks of treatment, with secondary outcomes including WOMAC, STAI, and safety assessments. fMRI observations will employ independent component analysis, brain network construction, and functional connectivity, complemented by Pearson correlation analysis to explore the relationship between brain responses and clinical improvements. Conclusion This study will initially uncover how acupuncture intervention for chronic KOA pain centrally regulates and exerts therapeutic effects through the modulation of abnormal brain network functional connectivity patterns, with a demonstrated long-term effect. Ethics and Dissemination This study has been approved by the ethics committee of Shandong University of Traditional Chinese Medicine Affiliated Hospital ((2024) Lunshen No. (028) - KY). Clinical Trial Registration This study has been approved by registered in the Chinese Clinical Trial Registry (ChiCTR2400083695).
Collapse
Affiliation(s)
- Yi-Yang Sun
- Department of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Ming-Lin Wang
- Traditional Chinese Medicine Comprehensive Treatment Center, Shengli Oilfield Central Hospital, Jinan, Shandong, People’s Republic of China
| | - Ruo-Han Zhao
- Department of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Ming-Yao Hao
- External Treatment Center of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Shuai Song
- External Treatment Center of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yue Zhou
- External Treatment Center of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Na Zhang
- Department of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
3
|
Wijesinghe SN, Ditchfield C, Flynn S, Agrawal J, Davis ET, Dajas-Bailador F, Chapman V, Jones SW. Immunomodulation and fibroblast dynamics driving nociceptive joint pain within inflammatory synovium: Unravelling mechanisms for therapeutic advancements in osteoarthritis. Osteoarthritis Cartilage 2024; 32:1358-1370. [PMID: 38960140 DOI: 10.1016/j.joca.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/21/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE Synovitis is a widely accepted sign of osteoarthritis (OA), characterised by tissue hyperplasia, where increased infiltration of immune cells and proliferation of resident fibroblasts adopt a pro-inflammatory phenotype, and increased the production of pro-inflammatory mediators that are capable of sensitising and activating sensory nociceptors, which innervate the joint tissues. As such, it is important to understand the cellular composition of synovium and their involvement in pain sensitisation to better inform the development of effective analgesics. METHODS Studies investigating pain sensitisation in OA with a focus on immune cells and fibroblasts were identified using PubMed, Web of Science and SCOPUS. RESULTS In this review, we comprehensively assess the evidence that cellular crosstalk between resident immune cells or synovial fibroblasts with joint nociceptors in inflamed OA synovium contributes to peripheral pain sensitisation. Moreover, we explore whether the elucidation of common mechanisms identified in similar joint conditions may inform the development of more effective analgesics specifically targeting OA joint pain. CONCLUSION The concept of local environment and cellular crosstalk within the inflammatory synovium as a driver of nociceptive joint pain presents a compelling opportunity for future research and therapeutic advancements.
Collapse
Affiliation(s)
- Susanne N Wijesinghe
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Caitlin Ditchfield
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Sariah Flynn
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Jyoti Agrawal
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK.
| | | | | | - Victoria Chapman
- Pain Centre Versus Arthritis, NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
4
|
Jiang P, Hu K, Jin L, Luo Z. A brief review of current treatment options for osteoarthritis including disease-modifying osteoarthritis drugs (DMOADs) and novel therapeutics. Ann Med Surg (Lond) 2024; 86:4042-4048. [PMID: 38989236 PMCID: PMC11230824 DOI: 10.1097/ms9.0000000000002214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024] Open
Abstract
Osteoarthritis (OA) is a chronic disorder caused by degenerative changes in articular cartilage, which are mainly manifests as degeneration of cartilage, subchondral bone remodeling, as well as synovial inflammation. Over the next few decades, OA and its burden will continue to increase worldwide, posing a major public health challenge for the foreseeable future. Treatment for OA includes non-pharmacological, pharmacological, and surgical treatments. Existing conservative treatments and joint surgery can only alleviate the symptoms and cannot be cured, so new therapies for OA are urgently needed. Since advances in the understanding of OA pathophysiology, researchers have identified some potential therapeutic targets against degeneration of cartilage, subchondral bone remodeling and synovial inflammation, enabling development of the disease-modifying OA drugs (DMOADs). Additionally, a number of new technologies are also being investigated for treating OA, such as RNA interference (RNAi), CRISPR/Cas9 and PROTAC. The goal of this review is to describe the current development status of DMOADs and to discuss the potential of emerging therapeutic approaches for treating OA, thus providing a reference for OA treatments.
Collapse
Affiliation(s)
| | | | - Liang Jin
- The Second Affiliated Hospital Zhejiang University School of Medicine Changxing Campus, Taihu Middle Road, Changxing County, Zhejiang Province, China
| | - Zhicheng Luo
- The Second Affiliated Hospital Zhejiang University School of Medicine Changxing Campus, Taihu Middle Road, Changxing County, Zhejiang Province, China
| |
Collapse
|
5
|
Chiang YF, Huang KC, Wang KL, Huang YJ, Chen HY, Ali M, Shieh TM, Hsia SM. Protective Effects of an Oligo-Fucoidan-Based Formula against Osteoarthritis Development via iNOS and COX-2 Suppression following Monosodium Iodoacetate Injection. Mar Drugs 2024; 22:211. [PMID: 38786602 PMCID: PMC11123468 DOI: 10.3390/md22050211] [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: 03/05/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disorder characterized by cartilage degradation and chronic inflammation, accompanied by high oxidative stress. In this study, we utilized the monosodium iodoacetate (MIA)-induced OA model to investigate the efficacy of oligo-fucoidan-based formula (FF) intervention in mitigating OA progression. Through its capacity to alleviate joint bearing function and inflammation, improvements in cartilage integrity following oligo-fucoidan-based formula intervention were observed, highlighting its protective effects against cartilage degeneration and structural damage. Furthermore, the oligo-fucoidan-based formula modulated the p38 signaling pathway, along with downregulating cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expression, contributing to its beneficial effects. Our study provides valuable insights into targeted interventions for OA management and calls for further clinical investigations to validate these preclinical findings and to explore the translational potential of an oligo-fucoidan-based formula in human OA patients.
Collapse
Affiliation(s)
- Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Ko-Chieh Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Kai-Lee Wang
- Department of Nursing, Deh Yu College of Nursing and Health, Keelung 203301, Taiwan
| | - Yun-Ju Huang
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan 710301, Taiwan
| | - Hsin-Yuan Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA;
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Tzong-Ming Shieh
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- School of Food and Safety, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
6
|
Patnaik R, Riaz S, Sivani BM, Faisal S, Naidoo N, Rizzo M, Banerjee Y. Evaluating the potential of Vitamin D and curcumin to alleviate inflammation and mitigate the progression of osteoarthritis through their effects on human chondrocytes: A proof-of-concept investigation. PLoS One 2023; 18:e0290739. [PMID: 38157375 PMCID: PMC10756552 DOI: 10.1371/journal.pone.0290739] [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: 03/14/2023] [Accepted: 08/14/2023] [Indexed: 01/03/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder primarily affecting the elderly, characterized by a prominent inflammatory component. The long-term side effects associated with current therapeutic approaches necessitate the development of safer and more efficacious alternatives. Nutraceuticals, such as Vitamin D and curcumin, present promising therapeutic potentials due to their safety, efficacy, and cost-effectiveness. In this study, we utilized a proinflammatory human chondrocyte model of OA to assess the anti-inflammatory properties of Vitamin D and curcumin, with a particular focus on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway. Employing a robust siRNA approach, we effectively modulated the expression of PAR-2 to understand its role in the inflammatory process. Our results reveal that both Vitamin D and curcumin attenuate the expression of PAR-2, leading to a reduction in the downstream proinflammatory cytokines, such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin 6 (IL-6), and Interleukin 8 (IL-8), implicated in the OA pathogenesis. Concurrently, these compounds suppressed the expression of Receptor Activator of Nuclear Factor kappa-Β Ligand (RANKL) and its receptor RANK, which are associated with PAR-2 mediated TNF-α stimulation. Additionally, Vitamin D and curcumin downregulated the expression of Interferon gamma (IFN-γ), known to elevate RANKL levels, underscoring their potential therapeutic implications in OA. This study, for the first time, provides evidence of the mitigating effect of Vitamin D and curcumin on PAR-2 mediated inflammation, employing an siRNA approach in OA. Thus, our findings pave the way for future research and the development of novel, safer, and more effective therapeutic strategies for managing OA.
Collapse
Affiliation(s)
- Rajashree Patnaik
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Sumbal Riaz
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Bala Mohan Sivani
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Shemima Faisal
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Nerissa Naidoo
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Yajnavalka Banerjee
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
- Centre for Medical Education, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
7
|
Singh N, Bhattacharjee A, Kumar P, Katti DS. Targeting multiple disease hallmarks using a synergistic disease-modifying drug combination ameliorates osteoarthritis via inhibition of senescence and inflammation. Life Sci 2023; 334:122212. [PMID: 37890697 DOI: 10.1016/j.lfs.2023.122212] [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: 05/23/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
AIMS Osteoarthritis (OA), is a debilitating disease characterized by progressive cartilage degradation, synovial inflammation, and chondrocyte senescence. Various treatment agents independently targeting these hallmarks have been investigated. However, due to the complex multifaceted nature of OA, no disease-modifying osteoarthritis drugs are clinically available. In an attempt to overcome this, we developed a combinatorial approach and demonstrated the efficacy of TsC [Tissue inhibitor of metalloproteinase-3 (TIMP3) + sulfated carboxymethylcellulose (sCMC)] and piperlongumine (PL) combination for the amelioration of OA in a goat ex vivo OA model. MAIN METHODS The efficacy of the drug combination was evaluated using the goat ex vivo OA explant model and results were validated in clinically relevant human OA cartilage explants. The chondroprotective effects were evaluated in terms of reduced inflammation and cartilage matrix loss, reduction in chondrosenescence, and reduced oxidative stress. KEY FINDINGS A combination of TsC and PL (TsC-PL) significantly reduced inflammation, cartilage matrix loss, chondrosenescence, and oxidative stress in the goat ex vivo OA model and showed chondroprotective effects. Further, similar chondroprotective effects were observed in human OA cartilage. Additionally, the coefficient of drug interaction analysis indicated that the combination of TsC and PL had a synergistic effect in reducing matrix degrading proteases and inflammation (goat ex vivo OA model) and Reactive oxygen species (ROS) production (human OA cartilage). SIGNIFICANCE Combinatorial treatment with TsC and PL demonstrated potential disease-modifying effects for the treatment of osteoarthritis via inhibition of inflammation and senescence and supports the usage of treatment strategies targeting multiple pathological factors of OA simultaneously.
Collapse
Affiliation(s)
- Nihal Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India; The Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Arijit Bhattacharjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India; The Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Praganesh Kumar
- Ganesh Shankar Vidyarthi Memorial Medical College, Kanpur 208002, Uttar Pradesh, India
| | - Dhirendra S Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India; The Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
8
|
Huang C, Zhao Y, Lin S, Li L, Guo X, Yumiseba S, Yang JD, Hariri R, Ye Q, He S, Kilcoyne A. Characterization of human placenta-derived exosome (pExo) as a potential osteoarthritis disease modifying therapeutic. Arthritis Res Ther 2023; 25:229. [PMID: 38017556 PMCID: PMC10683254 DOI: 10.1186/s13075-023-03219-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE Human placenta-derived exosomes (pExo) were generated, characterized, and evaluated as a therapeutic candidate for the treatment of osteoarthritis (OA). METHODS pExo was generated from full-term human placenta tissues by sequential centrifugation, purification, and sterile filtration. Upon analysis of particle size, cytokine composition, and exosome marker expression, pExo was further tested in cell-based assays to examine its effects on human chondrocytes. In vivo therapeutic efficacies were evaluated in a medial meniscal tear/medial collateral ligament tear (MCLT + MMT) rat model, in which animals received pExo injections intraarticularly and weight bearing tests during in-life stage while histopathology and immunohistochemistry were performed as terminal endpoints. RESULTS pExo displayed typical particle size, expressed maker proteins of exosome, and contained proteins with pro-proliferative, pro-anabolic, anti-catabolic, or anti-inflammatory activities. In vitro, pExo promoted chondrocyte migration and proliferation dose-dependently, which may involve its activation of cell growth-related signaling pathways. Expression of inflammatory and catabolic genes induced in a cellular OA model was significantly suppressed by pExo. In the rat OA model, pExo alleviated pain burden, restored cartilage degeneration, and downregulated expressions of pro-inflammatory, catabolic, or apoptotic proteins in a dose-dependent manner. CONCLUSIONS Our study demonstrates that pExo has multiple potential therapeutic effects including symptom control and disease modifying characteristics. This may make it an attractive candidate for further development as an anti-OA therapeutic.
Collapse
Affiliation(s)
- Chenfei Huang
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Yuechao Zhao
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA.
| | - Shengchen Lin
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Lin Li
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Xuan Guo
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | | | - Jeng-Dar Yang
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Robert Hariri
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Qian Ye
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Shuyang He
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| | - Adrian Kilcoyne
- Celularity Inc., 170 Park Avenue, Florham Park, NJ, 07932, USA
| |
Collapse
|
9
|
Tang Y, Liu Y, Zhu X, Chen Y, Jiang X, Ding S, Zheng Q, Zhang M, Yang J, Ma Y, Xing M, Zhang Z, Ding H, Jin Y, Ma C. ALKBH5-mediated m 6A demethylation of HS3ST3B1-IT1 prevents osteoarthritis progression. iScience 2023; 26:107838. [PMID: 37752950 PMCID: PMC10518728 DOI: 10.1016/j.isci.2023.107838] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 06/15/2023] [Accepted: 09/02/2023] [Indexed: 09/28/2023] Open
Abstract
HS3ST3B1-IT1 was identified as a downregulated long noncoding RNA in osteoarthritic cartilage. However, its roles and mechanisms in the pathogenesis of osteoarthritis (OA) are unclear. In this study, we demonstrated that the expressions of HS3ST3B1-IT1 and its maternal gene HS3ST3B1 were downregulated and positively correlated in osteoarthritic cartilage. Overexpression of HS3ST3B1-IT1 significantly increased chondrocyte viability, inhibited chondrocyte apoptosis, and upregulated extracellular matrix (ECM) proteins, whereas HS3ST3B1-IT1 knockdown had the opposite effects. In addition, HS3ST3B1-IT1 significantly ameliorated monosodium-iodoacetate-induced OA in vivo. Mechanistically, HS3ST3B1-IT1 upregulated HS3ST3B1 expression by blocking its ubiquitination-mediated degradation. Knockdown of HS3ST3B1 reversed the effects of HS3ST3B1-IT1 on chondrocyte viability, apoptosis, and ECM metabolism. AlkB homolog 5 (ALKBH5)-mediated N6-methyladenosine (m6A) demethylation stabilized HS3ST3B1-IT1 RNA. Together, our data revealed that ALKBH5-mediated upregulation of HS3ST3B1-IT1 suppressed OA progression by elevating HS3ST3B1 expression, suggesting that HS3ST3B1-IT1/HS3ST3B1 may serve as potential therapeutic targets for OA treatment.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Yang Liu
- Department of Orthopedics, Nanjing First Hospital, Nanjing, P.R. China
| | - Xiaoshu Zhu
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Yanlin Chen
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Xinluan Jiang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Siyang Ding
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Que Zheng
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Ming Zhang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Jiashu Yang
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Yunfei Ma
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Mengying Xing
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Zongyu Zhang
- Department of Orthopedics, the Traditional Chinese Medical Hospital of Lianyungang, Lianyungang, P.R. China
| | - Huimin Ding
- Department of Orthopedics, BenQ Medical Center, the Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Yucui Jin
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| | - Changyan Ma
- Department of Medical Genetics, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Longmian Road 101, Nanjing, P.R. China
| |
Collapse
|
10
|
Shen L, Yue S. A clinical model to predict the progression of knee osteoarthritis: data from Dryad. J Orthop Surg Res 2023; 18:628. [PMID: 37635226 PMCID: PMC10464113 DOI: 10.1186/s13018-023-04118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/21/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a multifactorial, slow-progressing, non-inflammatory degenerative disease primarily affecting synovial joints. It is usually induced by advanced age and/or trauma and eventually leads to irreversible destruction of articular cartilage and other tissues of the joint. Current research on KOA progression has limited clinical application significance. In this study, we constructed a prediction model for KOA progression based on multiple clinically relevant factors to provide clinicians with an effective tool to intervene in KOA progression. METHOD This study utilized the data set from the Dryad database which included patients with Kellgren-Lawrence (KL) grades 2 and 3. The KL grades was determined as the dependent variable, while 15 potential predictors were identified as independent variables. Patients were randomized into training set and validation set. The training set underwent LASSO analysis, model creation, visualization, decision curve analysis and internal validation using R language. The validation set is externally validated and F1-score, precision, and recall are computed. RESULT A total of 101 patients with KL2 and 94 patients with KL3 were selected. We randomly split the data set into a training set and a validation set by 8:2. We filtered "BMI", "TC", "Hypertension treatment", and "JBS3 (%)" to build the prediction model for progression of KOA. Nomogram used to visualize the model in R language. Area under ROC curve was 0.896 (95% CI 0.847-0.945), indicating high discrimination. Mean absolute error (MAE) of calibration curve = 0.041, showing high calibration. MAE of internal validation error was 0.043, indicating high model calibration. Decision curve analysis showed high net benefit. External validation of the metabolic syndrome column-line graph prediction model was performed by the validation set. The area under the ROC curve was 0.876 (95% CI 0.767-0.984), indicating that the model had a high degree of discrimination. Meanwhile, the calibration curve Mean absolute error was 0.113, indicating that the model had a high degree of calibration. The F1 score is 0.690, the precision is 0.667, and the recall is 0.714. The above metrics represent a good performance of the model. CONCLUSION We found that KOA progression was associated with four variable predictors and constructed a predictive model for KOA progression based on the predictors. The clinician can intervene based on the nomogram of our prediction model. KEY INFORMATION This study is a clinical predictive model of KOA progression. KOA progression prediction model has good credibility and clinical value in the prevention of KOA progression.
Collapse
Affiliation(s)
- Lianwei Shen
- Rehabitation Center, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - Shouwei Yue
- Rehabitation Center, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China.
| |
Collapse
|
11
|
Copp G, Robb KP, Viswanathan S. Culture-expanded mesenchymal stromal cell therapy: does it work in knee osteoarthritis? A pathway to clinical success. Cell Mol Immunol 2023; 20:626-650. [PMID: 37095295 PMCID: PMC10229578 DOI: 10.1038/s41423-023-01020-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative multifactorial disease with concomitant structural, inflammatory, and metabolic changes that fluctuate in a temporal and patient-specific manner. This complexity has contributed to refractory responses to various treatments. MSCs have shown promise as multimodal therapeutics in mitigating OA symptoms and disease progression. Here, we evaluated 15 randomized controlled clinical trials (RCTs) and 11 nonrandomized RCTs using culture-expanded MSCs in the treatment of knee OA, and we found net positive effects of MSCs on mitigating pain and symptoms (improving function in 12/15 RCTs relative to baseline and in 11/15 RCTs relative to control groups at study endpoints) and on cartilage protection and/or repair (18/21 clinical studies). We examined MSC dose, tissue of origin, and autologous vs. allogeneic origins as well as patient clinical phenotype, endotype, age, sex and level of OA severity as key parameters in parsing MSC clinical effectiveness. The relatively small sample size of 610 patients limited the drawing of definitive conclusions. Nonetheless, we noted trends toward moderate to higher doses of MSCs in select OA patient clinical phenotypes mitigating pain and leading to structural improvements or cartilage preservation. Evidence from preclinical studies is supportive of MSC anti-inflammatory and immunomodulatory effects, but additional investigations on immunomodulatory, chondroprotective and other clinical mechanisms of action are needed. We hypothesize that MSC basal immunomodulatory "fitness" correlates with OA treatment efficacy, but this hypothesis needs to be validated in future studies. We conclude with a roadmap articulating the need to match an OA patient subset defined by molecular endotype and clinical phenotype with basally immunomodulatory "fit" or engineered-to-be-fit-for-OA MSCs in well-designed, data-intensive clinical trials to advance the field.
Collapse
Affiliation(s)
- Griffin Copp
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Kevin P Robb
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada.
- Krembil Research Institute, University Health Network, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Department of Medicine, Division of Hematology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Kang L, Yang AL, Lai CH, Chen TJ, Lin SY, Wang YH, Wang CZ, Conway EM, Wu HL, Ho ML, Chang JK, Chen CH, Cheng TL. Chondrocyte Thrombomodulin Protects against Osteoarthritis. Int J Mol Sci 2023; 24:ijms24119522. [PMID: 37298473 DOI: 10.3390/ijms24119522] [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: 05/04/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent form of arthritis that affects over 32.5 million adults worldwide, causing significant cartilage damage and disability. Unfortunately, there are currently no effective treatments for OA, highlighting the need for novel therapeutic approaches. Thrombomodulin (TM), a glycoprotein expressed by chondrocytes and other cell types, has an unknown role in OA. Here, we investigated the function of TM in chondrocytes and OA using various methods, including recombinant TM (rTM), transgenic mice lacking the TM lectin-like domain (TMLeD/LeD), and a microRNA (miRNA) antagomir that increased TM expression. Results showed that chondrocyte-expressed TM and soluble TM [sTM, like recombinant TM domain 1 to 3 (rTMD123)] enhanced cell growth and migration, blocked interleukin-1β (IL-1β)-mediated signaling and protected against knee function and bone integrity loss in an anterior cruciate ligament transection (ACLT)-induced mouse model of OA. Conversely, TMLeD/LeD mice exhibited accelerated knee function loss, while treatment with rTMD123 protected against cartilage loss even one-week post-surgery. The administration of an miRNA antagomir (miR-up-TM) also increased TM expression and protected against cartilage damage in the OA model. These findings suggested that chondrocyte TM plays a crucial role in counteracting OA, and miR-up-TM may represent a promising therapeutic approach to protect against cartilage-related disorders.
Collapse
Grants
- 110-2314-B-037-022- Ministry of Science and Technology, Executive Yuan, Taiwan
- 111-2314-B-037-055- Ministry of Science and Technology, Executive Yuan, Taiwan
- 110-2314-B-006 -037 -MY3 Ministry of Science and Technology, Executive Yuan, Taiwan
- 110-2314-B-037 -029 -MY3 Ministry of Science and Technology, Executive Yuan, Taiwan
- 111-2314-B-037-106 Ministry of Science and Technology, Executive Yuan, Taiwan
- KMTTH- 111-R002 Kaohsiung Municipal Ta-Tung Hospital
- KMTTH-DK(A)112001 Kaohsiung Municipal Ta-Tung Hospital
- KMU-TC112A02 Kaohsiung Medical University
- KMUH-DK(A)110003 Kaohsiung Medical University
- KMU-DK(B) 110002 Kaohsiung Medical University
Collapse
Affiliation(s)
- Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Ai-Lun Yang
- Institute of Sports Sciences, University of Taipei, Taipei 11153, Taiwan
| | - Chao-Han Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tsan-Ju Chen
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sung-Yen Lin
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yan-Hsiung Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chau-Zen Wang
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Edward M Conway
- Centre for Blood Research, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
| | - Mei-Ling Ho
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 800, Taiwan
| | - Je-Ken Chang
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 800, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Chung-Hwan Chen
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
13
|
Wijesinghe SN, Badoume A, Nanus DE, Sharma‐Oates A, Farah H, Certo M, Alnajjar F, Davis ET, Mauro C, Lindsay MA, Jones SW. Obesity defined molecular endotypes in the synovium of patients with osteoarthritis provides a rationale for therapeutic targeting of fibroblast subsets. Clin Transl Med 2023; 13:e1232. [PMID: 37006170 PMCID: PMC10068310 DOI: 10.1002/ctm2.1232] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA), a multifaceted condition, poses a significant challenge for the successful clinical development of therapeutics due to heterogeneity. However, classifying molecular endotypes of OA pathogenesis could provide invaluable phenotype-directed routes for stratifying subgroups of patients for targeted therapeutics, leading to greater chances of success in trials. This study establishes endotypes in OA soft joint tissue driven by obesity in both load-bearing and non-load bearing joints. METHODS Hand, hip, knee and foot joint synovial tissue was obtained from OA patients (n = 32) classified as obese (BMI > 30) or normal weight (BMI 18.5-24.9). Isolated fibroblasts (OA SF) were assayed by Olink proteomic panel, seahorse metabolic flux assay, Illumina's NextSeq 500 bulk and Chromium 10X single cell RNA-sequencing, validated by Luminex and immunofluorescence. RESULTS Targeted proteomic, metabolic and transcriptomic analysis found the inflammatory landscape of OA SFs are independently impacted by obesity, joint loading and anatomical site with significant heterogeneity between obese and normal weight patients, confirmed by bulk RNAseq. Further investigation by single cell RNAseq identified four functional molecular endotypes including obesity specific subsets defined by an inflammatory endotype related to immune cell regulation, fibroblast activation and inflammatory signaling, with up-regulated CXCL12, CFD and CHI3L1 expression. Luminex confirmed elevated chitase3-like-1(229.5 vs. 49.5 ng/ml, p < .05) and inhibin (20.6 vs. 63.8 pg/ml, p < .05) in obese and normal weight OA SFs, respectively. Lastly, we find SF subsets in obese patients spatially localise in sublining and lining layers of OA synovium and can be distinguished by differential expression of the transcriptional regulators MYC and FOS. CONCLUSION These findings demonstrate the significance of obesity in changing the inflammatory landscape of synovial fibroblasts in both load bearing and non-load bearing joints. Describing multiple heterogeneous OA SF populations characterised by specific molecular endotypes, which drive heterogeneity in OA disease pathogenesis. These molecular endotypes may provide a route for the stratification of patients in clinical trials, providing a rational for the therapeutic targeting of specific SF subsets in specific patient populations with arthritic conditions.
Collapse
Affiliation(s)
- Susanne N. Wijesinghe
- Institute of Inflammation and AgeingMRC‐ARUK Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
| | - Amel Badoume
- Department of Pharmacy and PharmacologyUniversity of Bath, Claverton DownBathUK
| | - Dominika E. Nanus
- Institute of Inflammation and AgeingMRC‐ARUK Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
| | | | - Hussein Farah
- Institute of Inflammation and AgeingMRC‐ARUK Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
| | - Michelangelo Certo
- Institute of Inflammation and AgeingMRC‐ARUK Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
| | - Fawzeyah Alnajjar
- Institute of Inflammation and AgeingMRC‐ARUK Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
| | | | - Claudio Mauro
- Institute of Inflammation and AgeingMRC‐ARUK Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
| | - Mark A. Lindsay
- Department of Pharmacy and PharmacologyUniversity of Bath, Claverton DownBathUK
| | - Simon W. Jones
- Institute of Inflammation and AgeingMRC‐ARUK Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
| |
Collapse
|
14
|
Lee JJ, Namiri NK, Astuto B, Link TM, Majumdar S, Pedoia V. Personalized Risk Model and Leveraging of Magnetic Resonance Imaging-Based Structural Phenotypes and Clinical Factors to Predict Incidence of Radiographic Osteoarthritis. Arthritis Care Res (Hoboken) 2023; 75:501-508. [PMID: 35245407 DOI: 10.1002/acr.24877] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Our study aimed to investigate the association between time to incidence of radiographic osteoarthritis (OA) and magnetic resonance imaging (MRI)-based structural phenotypes proposed by the Rapid Osteoarthritis MRI Eligibility Score (ROAMES). METHODS A retrospective cohort of 2,328 participants without radiographic OA at baseline were selected from the Osteoarthritis Initiative study. Utilizing a deep-learning model, we automatically assessed the presence of inflammatory, meniscus/cartilage, subchondral bone, and hypertrophic phenotypes from MRIs acquired at baseline and 12-, 24-, 36-, 48-, 72-, and 96-month follow-up visits. In addition to 4 structural phenotypes, we examined severe knee injury history and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain scores as time dependent. We used Cox proportional hazards regression to analyze the association between 4 structural phenotypes and radiographic OA disease-free survival, both univariate and adjusted for known risk factors including age, sex, race, body mass index, presence of Heberden's nodes, and knee malalignment. RESULTS Inflammatory (hazard ratio [HR] 3.37 [95% confidence interval (95% CI) 2.45-4.63]), meniscus/cartilage (HR 1.55 [95% CI 1.21-1.98]), and subchondral bone (HR 1.84 [95% CI 1.63-2.09]) phenotypes were associated with time to radiographic OA at P < 0.05 when adjusted for the risk factors. Sex was a modifier of hypertrophic phenotype association with time to radiographic OA. Female participants with the hypertrophic phenotype were associated with 2.8 times higher risk of radiographic OA (95% CI 2.25-7.54) compared to male participants without the hypertrophic phenotype. CONCLUSION Four ROAMES phenotypes may contribute to time to radiographic OA incidence and if validated could be used as a promising tool for personalized OA management.
Collapse
Affiliation(s)
- Jinhee J Lee
- Center for Intelligent Imaging, University of California, San Francisco
| | - Nikan K Namiri
- Center for Intelligent Imaging, University of California, San Francisco
| | - Bruno Astuto
- Center for Intelligent Imaging, University of California, San Francisco
| | - Thomas M Link
- Center for Intelligent Imaging, University of California, San Francisco
| | - Sharmila Majumdar
- Center for Intelligent Imaging, University of California, San Francisco
| | - Valentina Pedoia
- Center for Intelligent Imaging, University of California, San Francisco
| |
Collapse
|
15
|
Nicholson TA, Sagmeister M, Wijesinghe SN, Farah H, Hardy RS, Jones SW. Oligonucleotide Therapeutics for Age-Related Musculoskeletal Disorders: Successes and Challenges. Pharmaceutics 2023; 15:237. [PMID: 36678864 PMCID: PMC9866666 DOI: 10.3390/pharmaceutics15010237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Age-related disorders of the musculoskeletal system including sarcopenia, osteoporosis and arthritis represent some of the most common chronic conditions worldwide, for which there remains a great clinical need to develop safer and more efficacious pharmacological treatments. Collectively, these conditions involve multiple tissues, including skeletal muscle, bone, articular cartilage and the synovium within the joint lining. In this review, we discuss the potential for oligonucleotide therapies to combat the unmet clinical need in musculoskeletal disorders by evaluating the successes of oligonucleotides to modify candidate pathological gene targets and cellular processes in relevant tissues and cells of the musculoskeletal system. Further, we discuss the challenges that remain for the clinical development of oligonucleotides therapies for musculoskeletal disorders and evaluate some of the current approaches to overcome these.
Collapse
Affiliation(s)
- Thomas A. Nicholson
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael Sagmeister
- Institute for Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Susanne N. Wijesinghe
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Hussein Farah
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Rowan S. Hardy
- Institute for Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Simon W. Jones
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
16
|
Rios JL, Sapède D, Djouad F, Rapp AE, Lang A, Larkin J, Ladel C, Mobasheri A. Animal Models of Osteoarthritis Part 1-Preclinical Small Animal Models: Challenges and Opportunities for Drug Development. Curr Protoc 2022; 2:e596. [PMID: 36342311 DOI: 10.1002/cpz1.596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis and a major source of pain and disability in the adult population. There is a significant unmet medical need for the development of effective pharmacological therapies for the treatment of OA. In addition to spontaneously occurring animal models of OA, many experimental animal models have been developed to provide insights into mechanisms of pathogenesis and progression. Many of these animal models are also being used in the drug development pipeline. Here, we provide an overview of commonly used and emerging preclinical small animal models of OA and highlight the strengths and limitations of small animal models in the context of translational drug development. There is limited information in the published literature regarding the technical reliability of these small animal models and their ability to accurately predict clinical drug development outcomes. The cost and complexity of the available models however is an important consideration for pharmaceutical companies, biotechnology startups, and contract research organizations wishing to incorporate preclinical models in target validation, discovery, and development pipelines. Further considerations relevant to industry include timelines, methods of induction, the key issue of reproducibility, and appropriate outcome measures needed to objectively assess outcomes of experimental therapeutics. Preclinical small animal models are indispensable tools that will shine some light on the pathogenesis of OA and its molecular endotypes in the context of drug development. This paper will focus on small animal models used in preclinical OA research. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Jaqueline Lourdes Rios
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Percuros BV, Leiden, The Netherlands
| | - Dora Sapède
- IRMB, Université de Montpellier, INSERM, Montpellier, France
| | - Farida Djouad
- IRMB, Université de Montpellier, INSERM, Montpellier, France
| | - Anna E Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopaedics (Friedrichsheim), University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Annemarie Lang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | | | | | - Ali Mobasheri
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Departments of Orthopaedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Liege, Belgium
| |
Collapse
|
17
|
Cuffaro D, Ciccone L, Rossello A, Nuti E, Santamaria S. Targeting Aggrecanases for Osteoarthritis Therapy: From Zinc Chelation to Exosite Inhibition. J Med Chem 2022; 65:13505-13532. [PMID: 36250680 PMCID: PMC9620172 DOI: 10.1021/acs.jmedchem.2c01177] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Indexed: 11/30/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. In 1999, two members of the A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) family of metalloproteinases, ADAMTS4 and ADAMTS5, or aggrecanases, were identified as the enzymes responsible for aggrecan degradation in cartilage. The first aggrecanase inhibitors targeted the active site by chelation of the catalytic zinc ion. Due to the generally disappointing performance of zinc-chelating inhibitors in preclinical and clinical studies, inhibition strategies tried to move away from the active-site zinc in order to improve selectivity. Exosite inhibitors bind to proteoglycan-binding residues present on the aggrecanase ancillary domains (called exosites). While exosite inhibitors are generally more selective than zinc-chelating inhibitors, they are still far from fulfilling their potential, partly due to a lack of structural and functional data on aggrecanase exosites. Filling this gap will inform the design of novel potent, selective aggrecanase inhibitors.
Collapse
Affiliation(s)
- Doretta Cuffaro
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Lidia Ciccone
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Armando Rossello
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Elisa Nuti
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Salvatore Santamaria
- Department
of Immunology and Inflammation, Imperial
College London, Du Cane Road, London W12
0NN, U.K.
| |
Collapse
|
18
|
Cho KH, Na HS, Jhun J, Woo JS, Lee AR, Lee SY, Lee JS, Um IG, Kim SJ, Park SH, Cho ML. Lactobacillus (LA-1) and butyrate inhibit osteoarthritis by controlling autophagy and inflammatory cell death of chondrocytes. Front Immunol 2022; 13:930511. [PMID: 36325344 PMCID: PMC9619036 DOI: 10.3389/fimmu.2022.930511] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/21/2022] [Indexed: 11/14/2022] Open
Abstract
Osteoarthritis (OA) reduces the quality of life as a result of the pain caused by continuous joint destruction. Inactivated Lactobacillus (LA-1) ameliorated osteoarthritis and protected cartilage by modulating inflammation. In this study, we evaluated the mechanism by which live LA-1 ameliorated OA. To investigate the effect of live LA-1 on OA progression, we administered LA-1 into monosodium iodoacetate (MIA)-induced OA animals. The pain threshold, cartilage damage, and inflammation of the joint synovial membrane were improved by live LA-1. Furthermore, the analysis of intestinal tissues and feces in the disease model has been shown to affect the systems of the intestinal system and improve the microbiome environment. Interestingly, inflammation of the intestinal tissue was reduced, and the intestinal microbiome was altered by live LA-1. Live LA-1 administration led to an increase in the level of Faecalibacterium which is a short-chain fatty acid (SCFA) butyrate-producing bacteria. The daily supply of butyrate, a bacterial SCFA, showed a tendency to decrease necroptosis, a type of abnormal cell death, by inducing autophagy and reversing impaired autophagy by the inflammatory environment. These results suggest that OA is modulated by changes in the gut microbiome, suggesting that activation of autophagy can reduce aberrant cell death. In summary, live LA-1 or butyrate ameliorates OA progression by modulating the gut environment and autophagic flux. Our findings suggest the regulation of the gut microenvironment as a therapeutic target for OA.
Collapse
Affiliation(s)
- Keun-Hyung Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Hyun Sik Na
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - JooYeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
| | - A Ram Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Jeong Su Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - In Gyu Um
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Seok Jung Kim
- Department of Orthopedic Surgery, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
- Department of Medical Life Sciences, College of Medicine, Catholic University of Korea, Seoul, South Korea
- *Correspondence: Mi-La Cho,
| |
Collapse
|
19
|
Nicholson T, Davis L, Davis ET, Newton Ede M, Scott A, Jones SW. e-Cigarette Vapour Condensate Reduces Viability and Impairs Function of Human Osteoblasts, in Part, via a Nicotine Dependent Mechanism. TOXICS 2022; 10:506. [PMID: 36136470 PMCID: PMC9504563 DOI: 10.3390/toxics10090506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 06/16/2023]
Abstract
Cigarette consumption negatively impacts bone quality and is a risk-factor for the development of multiple bone associated disorders, due to the highly vascularised structure of bone being exposed to systemic factors. However, the impact on bone to electronic cigarette (e-cigarette) use, which contains high doses of nicotine and other compounds including flavouring chemicals, metal particulates and carbonyls, is poorly understood. Here, we present the first evidence demonstrating the impact of e-cigarette vapour condensate (replicating changes in e-cigarette liquid chemical structure that occur upon device usage), on human primary osteoblast viability and function. 24 h exposure of osteoblasts to e-cigarette vapour condensate, generated from either second or third generation devices, significantly reduced osteoblast viability in a dose dependent manner, with condensate generated from the more powerful third generation device having greater toxicity. This effect was mediated in-part by nicotine, since exposure to nicotine-free condensate of an equal concentration had a less toxic effect. The detrimental effect of e-cigarette vapour condensate on osteoblast viability was rescued by co-treatment with the antioxidant N-Acetyl-L-cysteine (NAC), indicating toxicity may also be driven by reactive species generated upon device usage. Finally, non-toxic doses of either second or third generation condensate significantly blunted osteoblast osteoprotegerin secretion after 24 h, which was sustained for up to 7 days. In summary we demonstrate that e-cigarette vapour condensate, generated from commonly used second and third generation devices, can significantly reduce osteoblast viability and impair osteoblast function, at physiologically relevant doses. These data highlight the need for further investigation to inform users of the potential risks of e-cigarette use on bone health, including, accelerating bone associated disease progression, impacting skeletal development in younger users and to advise patients following orthopaedic surgery, dental surgery, or injury to maximise bone healing.
Collapse
Affiliation(s)
- Thomas Nicholson
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Lauren Davis
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Edward T. Davis
- Royal Orthopaedic Hospital, Bristol Road South, Birmingham B15 2TT, UK
| | | | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Simon W. Jones
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
20
|
Anderson JR, Jacobsen S, Walters M, Bundgaard L, Diendorfer A, Hackl M, Clarke EJ, James V, Peffers MJ. Small non-coding RNA landscape of extracellular vesicles from a post-traumatic model of equine osteoarthritis. Front Vet Sci 2022; 9:901269. [PMID: 36003409 PMCID: PMC9393553 DOI: 10.3389/fvets.2022.901269] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/18/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles comprise an as yet inadequately investigated intercellular communication pathway in the field of early osteoarthritis. We hypothesised that the small non-coding RNA expression pattern in synovial fluid and plasma would change during progression of experimental osteoarthritis. In this study, we conducted small RNA sequencing to provide a comprehensive overview of the temporal expression profiles of small non-coding transcripts carried by extracellular vesicles derived from plasma and synovial fluid for the first time in a posttraumatic model of equine osteoarthritis. Additionally, we characterised synovial fluid and plasma-derived extracellular vesicles with respect to quantity, size, and surface markers. The different temporal expressions of seven microRNAs in plasma and synovial fluid-derived extracellular vesicles, eca-miR-451, eca-miR-25, eca-miR-215, eca-miR-92a, eca-miR-let-7c, eca-miR-486-5p, and eca-miR-23a, and four snoRNAs, U3, snord15, snord46, and snord58, represent potential biomarkers for early osteoarthritis. Bioinformatics analysis of the differentially expressed microRNAs in synovial fluid highlighted that in early osteoarthritis these related to the inhibition of cell cycle, cell cycle progression, DNA damage and cell proliferation as well as increased cell viability and differentiation of stem cells. Plasma and synovial fluid-derived extracellular vesicle small non-coding signatures have been established for the first time in a temporal model of osteoarthritis. These could serve as novel biomarkers for evaluation of osteoarthritis progression or act as potential therapeutic targets.
Collapse
Affiliation(s)
- James R. Anderson
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Marie Walters
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Louise Bundgaard
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | | | | | - Emily J. Clarke
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Mandy J. Peffers
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
21
|
Tramś E, Malesa K, Pomianowski S, Kamiński R. Role of Platelets in Osteoarthritis-Updated Systematic Review and Meta-Analysis on the Role of Platelet-Rich Plasma in Osteoarthritis. Cells 2022; 11:1080. [PMID: 35406644 PMCID: PMC8997794 DOI: 10.3390/cells11071080] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/13/2022] [Accepted: 03/17/2022] [Indexed: 01/15/2023] Open
Abstract
Platelets are an essential component of hemostasis, with an increasing role in host inflammatory processes in injured tissues. The reaction between receptors and vascular endothelial cells results in the recruitment of platelets in the immune response pathway. The aim of the present review is to describe the role of platelets in osteoarthritis. Platelets induce secretion of biological substances, many of which are key players in the inflammatory response in osteoarthritis. Molecules involved in cartilage degeneration, or being markers of inflammation in osteoarthritis, are cytokines, such as tumor necrosis factor α (TNFα), interleukins (IL), type II collagen, aggrecan, and metalloproteinases. Surprisingly, platelets may also be used as a treatment modality for osteoarthritis. Multiple randomized controlled trials included in our systematic review and meta-analyses prove the effectiveness of platelet-rich plasma (PRP) as a minimally invasive method of pain alleviation in osteoarthritis treatment.
Collapse
Affiliation(s)
| | | | | | - Rafał Kamiński
- Centre of Postgraduate Medical Education, Department of Orthopaedics and Trauma Surgery, Professor A. Gruca Teaching Hospital, Konarskiego 13, 05-400 Otwock, Poland; (E.T.); (K.M.); (S.P.)
| |
Collapse
|
22
|
Farah H, Wijesinghe SN, Nicholson T, Alnajjar F, Certo M, Alghamdi A, Davis ET, Young SP, Mauro C, Jones SW. Differential Metabotypes in Synovial Fibroblasts and Synovial Fluid in Hip Osteoarthritis Patients Support Inflammatory Responses. Int J Mol Sci 2022; 23:ijms23063266. [PMID: 35328687 PMCID: PMC8950319 DOI: 10.3390/ijms23063266] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/02/2023] Open
Abstract
Changes in cellular metabolism have been implicated in mediating the activated fibroblast phenotype in a number of chronic inflammatory disorders, including pulmonary fibrosis, renal disease and rheumatoid arthritis. The aim of this study was therefore to characterise the metabolic profile of synovial joint fluid and synovial fibroblasts under both basal and inflammatory conditions in a cohort of obese and normal-weight hip OA patients. Furthermore, we sought to ascertain whether modulation of a metabolic pathway in OA synovial fibroblasts could alter their inflammatory activity. Synovium and synovial fluid was obtained from hip OA patients, who were either of normal-weight or obese and were undergoing elective joint replacement surgery. The synovial fluid metabolome was determined by 1H NMR spectroscopy. The metabolic profile of isolated synovial fibroblasts in vitro was characterised by lactate secretion, oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) using the Seahorse XF Analyser. The effects of a small molecule pharmacological inhibitor and siRNA targeted at glutaminase-1 (GLS1) were assessed to probe the role of glutamine metabolism in OA synovial fibroblast function. Obese OA patient synovial fluid (n = 5) exhibited a different metabotype, compared to normal-weight patient fluid (n = 6), with significantly increased levels of 1, 3-dimethylurate, N-Nitrosodimethylamine, succinate, tyrosine, pyruvate, glucose, glycine and lactate, and enrichment of the glutamine-glutamate metabolic pathway, which correlated with increasing adiposity. In vitro, isolated obese OA fibroblasts exhibited greater basal lactate secretion and aerobic glycolysis, and increased mitochondrial respiration when stimulated with pro-inflammatory cytokine TNFα, compared to fibroblasts from normal-weight patients. Inhibition of GLS1 attenuated the TNFα-induced expression and secretion of IL-6 in OA synovial fibroblasts. These findings suggest that altered cellular metabolism underpins the inflammatory phenotype of OA fibroblasts, and that targeted inhibition of glutamine-glutamate metabolism may provide a route to reducing the pathological effects of joint inflammation in OA patients who are obese.
Collapse
Affiliation(s)
- Hussein Farah
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Susanne N. Wijesinghe
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Thomas Nicholson
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Fawzeyah Alnajjar
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Abdullah Alghamdi
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Edward T. Davis
- The Royal Orthopaedic Hospital, NHS Foundation Trust, Bristol Road South, Northfield, Birmingham B31 2AP, UK;
| | - Stephen P. Young
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Claudio Mauro
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Simon W. Jones
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
- Correspondence:
| |
Collapse
|
23
|
Carpintero-Fernández P, Varela-Eirín M, García-Yuste A, López-Díaz I, Caeiro JR, Mayán MD. Osteoarthritis: Mechanistic Insights, Senescence, and Novel Therapeutic Opportunities. Bioelectricity 2022; 4:39-47. [PMID: 39355566 PMCID: PMC11441363 DOI: 10.1089/bioe.2021.0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease. In the last years, the research community has focused on understanding the molecular mechanisms that led to the pathogenesis of the disease, trying to identify different molecular and clinical phenotypes along with the discovery of new therapeutic opportunities. Different types of cell-to-cell communication mechanisms have been proposed to contribute to OA progression, including mechanisms mediated by connexin43 (Cx43) channels or by small extracellular vesicles. Furthermore, changes in the chondrocyte phenotype such as cellular senescence have been proposed as new contributors of the OA progression, changing the paradigm of the disease. The use of different drugs able to restore chondrocyte phenotype, to reduce cellular senescence and senescence-associated secretory phenotype components, and to modulate ion channel activity or Cx43 appears to be promising therapeutic strategies for the different types of OA. In this review, we aim to summarize the current knowledge in OA phenotypes related with aging and tissue damage and the new therapeutic opportunities currently available.
Collapse
Affiliation(s)
- Paula Carpintero-Fernández
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Marta Varela-Eirín
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Alejandro García-Yuste
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Iñaki López-Díaz
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - José Ramón Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| |
Collapse
|
24
|
Zhu J, Guo Y. Circ_0020093 Overexpression Alleviates Interleukin-1 Beta-induced Inflammation, Apoptosis and Extracellular Matrix Degradation in Human Chondrocytes by Targeting the miR-181a-5p/ERG Pathway. Immunol Invest 2022; 51:1660-1677. [PMID: 35012421 DOI: 10.1080/08820139.2021.2021938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Osteoarthritis (OA) is a well-known chronic degenerative joint disease, with multiple changes in the phenotype of chondrocytes. Circular RNAs (circRNAs) have been shown to be involved in various human diseases, including OA. The purpose of this study was to determine the role of circ_0020093 in OA pathological changes in vitro. C28/I2 cells were treated with interleukin-1 beta (IL-1β) to mimic OA pathological conditions. The expression levels of circ_0020093, miR-181a-5p and ETS-related gene (ERG) mRNA were measured by quantitative real-time PCR (qRT-PCR). For functional analyses, cell proliferative capacity was detected using EdU assay and CCK-8 assay. Inflammatory response was assessed by determining the release of pro-inflammatory factors using ELISA kits. Cell apoptosis was examined by flow cytometry assay. The levels of apoptosis-related proteins and extracellular matrix (ECM)-associated proteins were assessed by Western blot. The binding relationship between miR-181a-5p and circ_0020093 or ERG was confirmed by RNA pull-down assay, dual-luciferase reporter assay or RIP assay. The expression level of circ_0020093 was decreased in IL-1β-treated C28/I2 cells. Circ_0020093 overexpression relieved inflammatory responses, cell apoptosis and ECM degradation in IL-1β-induced C28/I2 cells. Circ_0020093 directly targeted miR-181a-5p, and miR-181a-5p bound to the 3' -untranslated region (3'UTR) of ERG to regulate ERG expression. Circ_0020093 overexpression promoted the expression of ERG by sponging miR-181a-5p. Rescue experiments showed that miR-181a-5p overexpression or ERG knockdown could reverse the inhibitory effects of circ_0020093 overexpression on the pathological changes in IL-1β-induced C28/I2 cells. Circ_0020093 overexpression alleviated IL-1β-induced human chondrocyte inflammatory injury, apoptosis and ECM degradation by targeting miR-181a-5p/ERG pathway.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang City, Hubei Province, China
| | - Yongchun Guo
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang City, Hubei Province, China
| |
Collapse
|
25
|
Abstract
The last decade has seen an enormous increase in long non-coding RNA (lncRNA) research within rheumatology. LncRNAs are arbitrarily classed as non-protein encoding RNA transcripts that exceed 200 nucleotides in length. These transcripts have tissue and cell specific patterns of expression and are implicated in a variety of biological processes. Unsurprisingly, numerous lncRNAs are dysregulated in rheumatoid conditions, correlating with disease activity and cited as potential biomarkers and targets for therapeutic intervention. In this chapter, following an introduction into each condition, we discuss the lncRNAs involved in rheumatoid arthritis, osteoarthritis and systemic lupus erythematosus. These inflammatory joint conditions share several inflammatory signalling pathways and therefore not surprisingly many commonly dysregulated lncRNAs are shared across these conditions. In the interest of translational research only those lncRNAs which are strongly conserved have been addressed. The lncRNAs discussed here have diverse roles in regulating inflammation, proliferation, migration, invasion and apoptosis. Understanding the molecular basis of lncRNA function in rheumatology will be crucial in fully determining the inflammatory mechanisms that drive these conditions.
Collapse
|
26
|
Panikkar M, Attia E, Dardak S. Osteoarthritis: A Review of Novel Treatments and Drug Targets. Cureus 2021; 13:e20026. [PMID: 34873554 PMCID: PMC8630567 DOI: 10.7759/cureus.20026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis affects over 10% of our population over the age of 60 years old, significantly reducing their quality of life and increasing morbidity. A number of aetiological factors contribute to the development of osteoarthritis including obesity, genetic factors, injury and increasing age. Many of the pathological processes which underlie the condition remain poorly understood and therefore limited progress has been made in developing effective disease modifying treatments. This review article aims to summarise our current understanding of osteoarthritis, the molecular mechanisms which drive the disease and current progress in developing therapeutic strategies to target these.
Collapse
Affiliation(s)
- Mohini Panikkar
- Trauma and Orthopaedics, Royal Shrewsbury Hospital, Shrewsbury, GBR
| | | | - Sara Dardak
- Trauma and Orthopaedics, St George's Hospital, London, GBR
| |
Collapse
|
27
|
Maqbool M, Fekadu G, Jiang X, Bekele F, Tolossa T, Turi E, Fetensa G, Fanta K. An up to date on clinical prospects and management of osteoarthritis. Ann Med Surg (Lond) 2021; 72:103077. [PMID: 34868573 PMCID: PMC8626656 DOI: 10.1016/j.amsu.2021.103077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
The rising prevalence of osteoarthritis (OA) in the general population has necessitated the development of novel treatment options. It is critical to recognize the joint as a separate entity participating in degenerative processes, as well as the multifaceted nature of OA. OA is incurable because there is currently no medication that can stop or reverse cartilage or bone loss. As this point of view has attracted attention, more research is being directed toward determining how the various joint components are impacted and how they contribute to OA pathogenesis. Over the next few years, several prospective therapies focusing on inflammation, cartilage metabolism, subchondral bone remodelling, cellular senescence, and the peripheral nociceptive pathway are predicted to transform the OA therapy landscape. Stem cell therapies and the use of various biomaterials to target articular cartilage (AC) and osteochondral tissues are now being investigated in considerable detail. Currently, laboratory-made cartilage tissues are on the verge of being used in clinical settings. This review focuses on the update of clinical prospects and management of osteoarthritis, as well as future possibilities for the treatment of OA. Osteoarthritis (OA) is a general term that incorporates several different joint diseases. The exact pathophysiology of OA remains unclear. OA is incurable because there is currently no medication that can stop or reverse cartilage or bone loss. Nonsteroidal anti-inflammatory drugs are the most frequently prescribed medications to alleviate arthritic discomfort. Stem cell therapies to target articular cartilage and osteochondral tissues are now under investigation.
Collapse
Affiliation(s)
- Mudasir Maqbool
- Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal Srinagar, 190006, Jammu and Kashmir, India
| | - Ginenus Fekadu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong.,School of Pharmacy, Institute of Health Sciences, Wollega University, Nekemte, Ethiopia
| | - Xinchan Jiang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong
| | - Firomsa Bekele
- Department of Pharmacy, College of Health Science, Mettu University, Mettu, Ethiopia
| | - Tadesse Tolossa
- Department of Public Health, Institute of Health Sciences, Wollega University, Nekemte, Ethiopia
| | - Ebisa Turi
- Department of Public Health, Institute of Health Sciences, Wollega University, Nekemte, Ethiopia
| | - Getahun Fetensa
- School of Nursing and Midwifery, Institute of Health Sciences, Wollega University, Nekemte, Ethiopia
| | - Korinan Fanta
- School of Pharmacy, Institute of Health Science, Jimma University, Jimma, Ethiopia
| |
Collapse
|
28
|
Nanus DE, Badoume A, Wijesinghe SN, Halsey AM, Hurley P, Ahmed Z, Botchu R, Davis ET, Lindsay MA, Jones SW. Synovial tissue from sites of joint pain in knee osteoarthritis patients exhibits a differential phenotype with distinct fibroblast subsets. EBioMedicine 2021; 72:103618. [PMID: 34628351 PMCID: PMC8511845 DOI: 10.1016/j.ebiom.2021.103618] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Synovial inflammation is associated with pain severity in patients with knee osteoarthritis (OA). The aim here was to determine in a population with knee OA, whether synovial tissue from areas associated with pain exhibited different synovial fibroblast subsets, compared to synovial tissue from sites not associated with pain. A further aim was to compare differences between early and end-stage disease synovial fibroblast subsets. METHODS Patients with early knee OA (n = 29) and end-stage knee OA (n = 22) were recruited. Patient reported pain was recorded by questionnaire and using an anatomical knee pain map. Proton density fat suppressed MRI axial and sagittal sequences were analysed and scored for synovitis. Synovial tissue was obtained from the medial and lateral parapatellar and suprapatellar sites. Fibroblast single cell RNA sequencing was performed using Chromium 10X and analysed using Seurat. Transcriptomes were functionally characterised using Ingenuity Pathway Analysis and the effect of fibroblast secretome on neuronal growth assessed using rat DRGN. FINDINGS Parapatellar synovitis was significantly associated with the pattern of patient-reported pain in knee OA patients. Synovial tissue from sites of patient-reported pain exhibited a differential transcriptomic phenotype, with distinct synovial fibroblast subsets in early OA and end-stage OA. Functional pathway analysis revealed that synovial tissue and fibroblast subsets from painful sites promoted fibrosis, inflammation and the growth and activity of neurons. The secretome of fibroblasts from early OA painful sites induced greater survival and neurite outgrowth in dissociated adult rodent dorsal root ganglion neurons. INTERPRETATION Sites of patient-reported pain in knee OA exhibit a different synovial tissue phenotype and distinct synovial fibroblast subsets. Further interrogation of these fibroblast pathotypes will increase our understanding of the role of synovitis in OA joint pain and provide a rationale for the therapeutic targeting of fibroblast subsets to alleviate pain in patients. FUNDING This study was funded by Versus Arthritis, UK (21530; 21812).
Collapse
Affiliation(s)
- Dominika E Nanus
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Amel Badoume
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Susanne N Wijesinghe
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Andrea M Halsey
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Patrick Hurley
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, United Kingdom,Centre for Trauma Sciences Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Rajesh Botchu
- The Royal Orthopaedic Hospital, Birmingham B31 2AP, United Kingdom
| | - Edward T Davis
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom,The Royal Orthopaedic Hospital, Birmingham B31 2AP, United Kingdom
| | - Mark A Lindsay
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom,Corresponding author.
| |
Collapse
|
29
|
Elucidating mechano-pathology of osteoarthritis: transcriptome-wide differences in mechanically stressed aged human cartilage explants. Arthritis Res Ther 2021; 23:215. [PMID: 34399844 PMCID: PMC8365911 DOI: 10.1186/s13075-021-02595-8] [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] [Received: 06/15/2021] [Accepted: 07/30/2021] [Indexed: 11/25/2022] Open
Abstract
Background Failing of intrinsic chondrocyte repair after mechanical stress is known as one of the most important initiators of osteoarthritis. Nonetheless, insight into these early mechano-pathophysiological processes in age-related human articular cartilage is still lacking. Such insights are needed to advance clinical development. To highlight important molecular processes of osteoarthritis mechano-pathology, the transcriptome-wide changes following injurious mechanical stress on human aged osteochondral explants were characterized. Methods Following mechanical stress at a strain of 65% (65%MS) on human osteochondral explants (n65%MS = 14 versus ncontrol = 14), RNA sequencing was performed. Differential expression analysis between control and 65%MS was performed to determine mechanical stress-specific changes. Enrichment for pathways and protein-protein interactions was analyzed with Enrichr and STRING. Results We identified 156 genes significantly differentially expressed between control and 65%MS human osteochondral explants. Of note, IGFBP5 (FC = 6.01; FDR = 7.81 × 10−3) and MMP13 (FC = 5.19; FDR = 4.84 × 10−2) were the highest upregulated genes, while IGFBP6 (FC = 0.19; FDR = 3.07 × 10−4) was the most downregulated gene. Protein-protein interactions were significantly higher than expected by chance (P = 1.44 × 10−15 with connections between 116 out of 156 genes). Pathway analysis showed, among others, enrichment for cellular senescence, insulin-like growth factor (IGF) I and II binding, and focal adhesion. Conclusions Our results faithfully represent transcriptomic wide consequences of mechanical stress in human aged articular cartilage with MMP13, IGF binding proteins, and cellular senescence as the most notable results. Acquired knowledge on the as such identified initial, osteoarthritis-related, detrimental responses of chondrocytes may eventually contribute to the development of effective disease-modifying osteoarthritis treatments. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02595-8.
Collapse
|
30
|
Oligonucleotide Therapies in the Treatment of Arthritis: A Narrative Review. Biomedicines 2021; 9:biomedicines9080902. [PMID: 34440106 PMCID: PMC8389545 DOI: 10.3390/biomedicines9080902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/23/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are two of the most common chronic inflammatory joint diseases, for which there remains a great clinical need to develop safer and more efficacious pharmacological treatments. The pathology of both OA and RA involves multiple tissues within the joint, including the synovial joint lining and the bone, as well as the articular cartilage in OA. In this review, we discuss the potential for the development of oligonucleotide therapies for these disorders by examining the evidence that oligonucleotides can modulate the key cellular pathways that drive the pathology of the inflammatory diseased joint pathology, as well as evidence in preclinical in vivo models that oligonucleotides can modify disease progression.
Collapse
|
31
|
Oo WM, Little C, Duong V, Hunter DJ. The Development of Disease-Modifying Therapies for Osteoarthritis (DMOADs): The Evidence to Date. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2921-2945. [PMID: 34262259 PMCID: PMC8273751 DOI: 10.2147/dddt.s295224] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) is a complex heterogeneous articular disease with multiple joint tissue involvement of varying severity and no regulatory-agency-approved disease-modifying drugs (DMOADs). In this review, we discuss the reasons necessitating the development of DMOADs for OA management, the classifications of clinical phenotypes or molecular/mechanistic endotypes from the viewpoint of targeted drug discovery, and then summarize the efficacy and safety profile of a range of targeted drugs in Phase 2 and 3 clinical trials directed to cartilage-driven, bone-driven, and inflammation-driven endotypes. Finally, we briefly put forward the reasons for failures in OA clinical trials and possible steps to overcome these barriers.
Collapse
Affiliation(s)
- Win Min Oo
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar
| | - Christopher Little
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Vicky Duong
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David J Hunter
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
32
|
eNAMPT Is Localised to Areas of Cartilage Damage in Patients with Hip Osteoarthritis and Promotes Cartilage Catabolism and Inflammation. Int J Mol Sci 2021; 22:ijms22136719. [PMID: 34201564 PMCID: PMC8269388 DOI: 10.3390/ijms22136719] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity increases the risk of hip osteoarthritis (OA). Recent studies have shown that adipokine extracellular nicotinamide phosphoribosyltransferase (eNAMPT or visfatin) induces the production of IL-6 and matrix metalloproteases (MMPs) in chondrocytes, suggesting it may promote articular cartilage degradation. However, neither the functional effects of extracellular visfatin on human articular cartilage tissue, nor its expression in the joint of hip OA patients of varying BMI, have been reported. Hip OA joint tissues were collected from patients undergoing joint replacement surgery. Cartilage explants were stimulated with recombinant human visfatin. Pro-inflammatory cytokines and MMPs were measured by ELISA and Luminex. Localisation of visfatin expression in cartilage tissue was determined by immunohistochemistry. Cartilage matrix degradation was determined by quantifying proteoglycan release. Expression of visfatin was elevated in the synovial tissue of hip OA patients who were obese, and was co-localised with MMP-13 in areas of cartilage damage. Visfatin promoted the degradation of hip OA cartilage proteoglycan and induced the production of pro-inflammatory cytokines (IL-6, MCP-1, CCL20, and CCL4) and MMPs. The elevated expression of visfatin in the obese hip OA joint, and its functional effects on hip cartilage tissue, suggests it plays a central role in the loss of cartilage integrity in obese patients with hip OA.
Collapse
|
33
|
Grässel S, Zaucke F, Madry H. Osteoarthritis: Novel Molecular Mechanisms Increase Our Understanding of the Disease Pathology. J Clin Med 2021; 10:jcm10091938. [PMID: 33946429 PMCID: PMC8125020 DOI: 10.3390/jcm10091938] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Although osteoarthritis (OA) is the most common musculoskeletal condition that causes significant health and social problems worldwide, its exact etiology is still unclear. With an aging and increasingly obese population, OA is becoming even more prevalent than in previous decades. Up to 35% of the world’s population over 60 years of age suffers from symptomatic (painful, disabling) OA. The disease poses a tremendous economic burden on the health-care system and society for diagnosis, treatment, sick leave, rehabilitation, and early retirement. Most patients also experience sleep disturbances, reduced capability for exercising, lifting, and walking and are less capable of working, and maintaining an independent lifestyle. For patients, the major problem is disability, resulting from joint tissue destruction and pain. So far, there is no therapy available that effectively arrests structural deterioration of cartilage and bone or is able to successfully reverse any of the existing structural defects. Here, we elucidate novel concepts and hypotheses regarding disease progression and pathology, which are relevant for understanding underlying the molecular mechanisms as a prerequisite for future therapeutic approaches. Emphasis is placed on topographical modeling of the disease, the role of proteases and cytokines in OA, and the impact of the peripheral nervous system and its neuropeptides.
Collapse
Affiliation(s)
- Susanne Grässel
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, 93053 Regensburg, Germany
- Correspondence:
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, 60528 Frankfurt am Main, Germany;
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany;
| |
Collapse
|
34
|
Zeng N, Chen XY, Yan ZP, Li JT, Liao T, Ni GX. Efficacy and safety of sprifermin injection for knee osteoarthritis treatment: a meta-analysis. Arthritis Res Ther 2021; 23:107. [PMID: 33836824 PMCID: PMC8034149 DOI: 10.1186/s13075-021-02488-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/22/2021] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE To perform a meta-analysis comparing the structural progression and clinical symptom outcomes as well as adverse events experienced from intra-articular injections of sprifermin compared to a placebo treatment for patients with knee osteoarthritis (KOA). METHOD We systematically searched the literature for studies that compared long-term outcomes between sprifermin and placebo injections for KOA treatment. Meta-analysis was performed with RevMan5.3 using an inverse variance approach with fixed or random effects models. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated. RESULTS Eight studies were included. Overall, there was significantly less improvement of WOMAC total scores in patients receiving sprifermin, compared with the placebo (mean difference (MD) = 3.23, 95% CI 0.76-5.69; I2 = 0%; P = 0.01). Further, sprifermin injection patients gained more, and lost less, cartilage thickness and volume in total femorotibial joint (cartilage thickness: standardized mean differences (SMD) = 0.55, 95% CI 0.26-0.84; I2 = 78%; P = 0.0002; cartilage volume: SMD = 0.39, 95% CI 0.20-0.58; I2 = 49%; P < 0.0001). Changes in the cartilage surface morphology of the medial tibio-femoral joint (MD = -0.30, 95% CI -0.44 to -0.16; I2 = 0%; P < 0.0001) and patello-femoral joint (MD = -0.22; 95% CI -0.37 to -0.07; I2 = 0%; P = 0.004) showed a significant difference between the sprifermin and placebo injections. Moreover, there were no significant differences between sprifermin and the placebo in the risk of treatment-emergent adverse events (OR = 1.05; 95% CI 0.52-2.14; I2 = 48%; P = 0.89). CONCLUSION The data from the included studies provide strong evidence to determine the effect of intra-articular sprifermin on joint structure in individuals with KOA and show no specific adverse effects. Nevertheless, intra-articular sprifermin did not likely have any positive effect on symptom alleviation.
Collapse
Affiliation(s)
- Ni Zeng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xin-Yuan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhi-Peng Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jie-Ting Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Tao Liao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guo-Xin Ni
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
35
|
Affiliation(s)
- Francesca Oliviero
- Department of Medicine - DIMED, Rheumatology Unit, University of Padua, Padua, Italy
| | - Roberta Ramonda
- Department of Medicine - DIMED, Rheumatology Unit, University of Padua, Padua, Italy
| |
Collapse
|
36
|
The Expression and Function of Metastases Associated Lung Adenocarcinoma Transcript-1 Long Non-Coding RNA in Subchondral Bone and Osteoblasts from Patients with Osteoarthritis. Cells 2021; 10:cells10040786. [PMID: 33916321 PMCID: PMC8066176 DOI: 10.3390/cells10040786] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/02/2023] Open
Abstract
Metastasis Associated Lung Adenocarcinoma Transcript-1 (MALAT1) is implicated in regulating the inflammatory response and in the pathology of several chronic inflammatory diseases, including osteoarthritis (OA). The purpose of this study was to examine the relationship between OA subchondral bone expression of MALAT1 with parameters of joint health and biomarkers of joint inflammation, and to determine its functional role in human OA osteoblasts. Subchondral bone and blood were collected from hip and knee OA patients (n = 17) and bone only from neck of femur fracture patients (n = 6) undergoing joint replacement surgery. Cytokines were determined by multiplex assays and ELISA, and gene expression by qPCR. MALAT1 loss of function was performed in OA patient osteoblasts using locked nucleic acids. The osteoblast transcriptome was analysed by RNASeq and pathway analysis. Bone expression of MALAT1 positively correlated to serum DKK1 and galectin-1 concentrations, and in OA patient osteoblasts was induced in response to IL-1β stimulation. Osteoblasts depleted of MALAT1 exhibited differential expression (>1.5 fold change) of 155 genes, including PTGS2. Both basal and IL-1β-mediated PGE2 secretion was greater in MALAT1 depleted osteoblasts. The induction of MALAT1 in human OA osteoblasts upon inflammatory challenge and its modulation of PGE2 production suggests that MALAT1 may play a role in regulating inflammation in OA subchondral bone.
Collapse
|
37
|
Na HS, Kwon JY, Lee SY, Lee SH, Lee AR, Woo JS, Jung K, Cho KH, Choi JW, Lee DH, Min HK, Park SH, Kim SJ, Cho ML. Metformin Attenuates Monosodium-Iodoacetate-Induced Osteoarthritis via Regulation of Pain Mediators and the Autophagy-Lysosomal Pathway. Cells 2021; 10:681. [PMID: 33808727 PMCID: PMC8003384 DOI: 10.3390/cells10030681] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative arthritis associated with pain and cartilage destruction in the elderly; it is known to be involved in inflammation as well. A drug called celecoxib is commonly used in patients with osteoarthritis to control pain. Metformin is used to treat type 2 diabetes but also exhibits regulation of the autophagy pathway. The purpose of this study is to investigate whether metformin can treat monosodium iodoacetate (MIA)-induced OA in rats. Metformin was administered orally every day to rats with OA. Paw-withdrawal latency and threshold were used to assess pain severity. Cartilage damage and pain mediators in dorsal root ganglia were evaluated by histological analysis and a scoring system. Relative mRNA expression was measured by real-time PCR. Metformin reduced the progression of experimental OA and showed both antinociceptive properties and cartilage protection. The combined administration of metformin and celecoxib controlled cartilage damage more effectively than metformin alone. In chondrocytes from OA patients, metformin reduced catabolic factor gene expression and inflammatory cell death factor expression, increased LC3Ⅱb, p62, and LAMP1 expression, and induced an autophagy-lysosome fusion phenotype. We investigated if metformin treatment reduces cartilage damage and inflammatory cell death of chondrocytes. The results suggest the potential for the therapeutic use of metformin in OA patients based on its ability to suppress pain and protect cartilage.
Collapse
Affiliation(s)
- Hyun Sik Na
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Ji Ye Kwon
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Seon-Yeong Lee
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Seung Hoon Lee
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - A Ram Lee
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Korea
| | - Jin Seok Woo
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - KyungAh Jung
- Impact Biotech, Korea 505 Banpo-Dong, Seocho-Ku, Seoul 06591, Korea;
| | - Keun-Hyung Cho
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Jeong-Won Choi
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Dong Hwan Lee
- Department of Orthopedic Surgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Korea;
| | - Hong-Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul 05030, Korea;
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seok Jung Kim
- Department of Orthopedic Surgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Korea;
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
- Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
38
|
van Hoolwerff M, Metselaar PI, Tuerlings M, Suchiman HED, Lakenberg N, Ramos YFM, Cats D, Nelissen RGHH, Broekhuis D, Mei H, de Almeida RC, Meulenbelt I. Elucidating Epigenetic Regulation by Identifying Functional cis-Acting Long Noncoding RNAs and Their Targets in Osteoarthritic Articular Cartilage. Arthritis Rheumatol 2020; 72:1845-1854. [PMID: 32840049 PMCID: PMC7702083 DOI: 10.1002/art.41396] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/02/2020] [Indexed: 12/23/2022]
Abstract
Objective To identify robustly differentially expressed long noncoding RNAs (lncRNAs) with osteoarthritis (OA) pathophysiology in cartilage and to explore potential target messenger RNA (mRNA) by establishing coexpression networks, followed by functional validation. Methods RNA sequencing was performed on macroscopically lesioned and preserved OA cartilage from patients who underwent joint replacement surgery due to OA (n = 98). Differential expression analysis was performed on lncRNAs that were annotated in GENCODE and Ensembl databases. To identify potential interactions, correlations were calculated between the identified differentially expressed lncRNAs and the previously reported differentially expressed protein‐coding genes in the same samples. Modulation of chondrocyte lncRNA expression was achieved using locked nucleic acid GapmeRs. Results By applying our in‐house pipeline, we identified 5,053 lncRNAs that were robustly expressed, of which 191 were significantly differentially expressed (according to false discovery rate) between lesioned and preserved OA cartilage. Upon integrating mRNA sequencing data, we showed that intergenic and antisense differentially expressed lncRNAs demonstrate high, positive correlations with their respective flanking sense genes. To functionally validate this observation, we selected P3H2‐AS1, which was down‐regulated in primary chondrocytes, resulting in the down‐regulation of P3H2 gene expression levels. As such, we can confirm that P3H2‐AS1 regulates its sense gene P3H2. Conclusion By applying an improved detection strategy, robustly differentially expressed lncRNAs in OA cartilage were detected. Integration of these lncRNAs with differential mRNA expression levels in the same samples provided insight into their regulatory networks. Our data indicate that intergenic and antisense lncRNAs play an important role in regulating the pathophysiology of OA.
Collapse
Affiliation(s)
| | | | | | | | - Nico Lakenberg
- Leiden University Medical Center, Leiden, The Netherlands
| | | | - Davy Cats
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Hailiang Mei
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
39
|
Drug delivery in intervertebral disc degeneration and osteoarthritis: Selecting the optimal platform for the delivery of disease-modifying agents. J Control Release 2020; 328:985-999. [PMID: 32860929 DOI: 10.1016/j.jconrel.2020.08.041] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) and intervertebral disc degeneration (IVDD) as major cause of chronic low back pain represent the most common degenerative joint pathologies and are leading causes of pain and disability in adults. Articular cartilage (AC) and intervertebral discs are cartilaginous tissues with a similar biochemical composition and pathophysiological aspects of degeneration. Although treatments directed at reversing these conditions are yet to be developed, many promising disease-modifying drug candidates are currently under investigation. Given the localized nature of these chronic diseases, drug delivery systems have the potential to enhance therapeutic outcomes by providing controlled and targeted release of bioactives, minimizing the number of injections needed and increasing drug concentration in the affected areas. This review provides a comprehensive overview of the currently most promising disease-modifying drugs as well as potential drug delivery systems for OA and IVDD therapy.
Collapse
|
40
|
Guo Q, Xu J, Shi Q, Wu S. PDLIM2 protects articular chondrocytes from lipopolysaccharide-induced apoptosis, degeneration and inflammatory injury through down-regulation of nuclear factor (NF)-κB signaling. Int Immunopharmacol 2020; 88:106883. [PMID: 32805696 DOI: 10.1016/j.intimp.2020.106883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022]
Abstract
Excessive inflammatory response-induced apoptosis and the degeneration of articular chondrocytes contribute to the development and progression of osteoarthritis. PDZ and LIM domain containing protein 2 (PDLIM2) has emerged as one of the pivotal regulators in orchestrating an inflammatory response through regulating the activity of transcription factor nuclear factor (NF)-κB. However, whether PDLIM2 participates in the articular chondrocyte-associated inflammatory response in osteoarthritis remains unknown. In the current study, we aimed to explore the biological function of PDLIM2 in lipopolysaccharide (LPS)-stimulated articular chondrocytes, an in vitro model of osteoarthritis. Herein, we found that PDLIM2 expression was significantly down-regulated in chondrocytes in response to LPS exposure. Functional experiments revealed that PDLIM2 overexpression increased the viability and decreased the apoptosis of chondrocytes following LPS treatment. Moreover, PDLIM2 overexpression attenuated LPS-induced degeneration of chondrocytes via the down-regulation of matrix metalloproteinase (MMP)-3 and MMP-13 and the up-regulation of COL2A1 and ACAN. In addition, the overexpression of PDLIM2 decreased LPS-induced production of interleukin (IL)-1β, IL-6 and TNF-α. In contrast, depletion of PDLIM2 exhibited the opposite effect. Mechanism research elucidated that PDLIM2 repressed the activation of NF-κB signaling associated with the down-regulation of NF-κB p65 protein expression. PDLIM2 depletion-exacerbated LPS-induced injury was significantly reversed by NF-κB inhibition. Taken together, these results demonstrate that PDLIM2 overexpression attenuates LPS-induced injury of articular chondrocytes through the inactivation of NF-κB signaling.
Collapse
Affiliation(s)
- Qinyue Guo
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Street, Xi'an, Shaanxi 710061, China
| | - Jing Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Street, Xi'an, Shaanxi 710061, China
| | - Qindong Shi
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Street, Xi'an, Shaanxi 710061, China
| | - Shufang Wu
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Street, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
41
|
Cheng HJ, Hsu WT, Chen CN, Li C. Activation of NOTCH1 by Shear Force Elicits Immediate Cytokine Expression in Human Chondrocytes. Int J Mol Sci 2020; 21:ijms21144958. [PMID: 32674293 PMCID: PMC7404062 DOI: 10.3390/ijms21144958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/04/2020] [Accepted: 07/10/2020] [Indexed: 01/01/2023] Open
Abstract
Osteoarthritis is caused by overloading of joints and is characterized by inflammation-induced disruption of cartilage structure. Current treatment strategy aims to relieve inflammation and prevent further deterioration of joint function. However, how mechanical force leads to inflammation and deterioration of chondrocyte function still remains incompletely understood. To explore the force-regulated molecular mechanism, an in vitro hydraulic shear force experiment to simulate the condition of force loading was required. The result demonstrated that multiple cytokines and immune regulators, including interleukin 8, interferon β, TRAF1 and TNFAIP3, were significantly increased by shear force within two hours of treatment. Moreover, JAG1 and HES1 were drastically upregulated as well, suggesting that NOTCH1 signaling is activated by shear force. Short-term expression of NOTCH1 intracellular domain activated a similar set of cytokines, indicating that NOTCH1 responds to shear force and activates downstream genes. When incubated under the medium conditioned by NOTCH1-activated chondrocyte, osteoblasts expressed higher levels of interferon β and interferon λ. Together, our results indicated that NOTCH1 functions as a force sensor and promotes expression of cytokines and immune regulators from shear-force bearing chondrocytes.
Collapse
Affiliation(s)
- Hao-Jen Cheng
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan; (H.-J.C.); (W.-T.H.)
- Department of Orthopedics, Shinnhomei Clinic, Chiayi 600, Taiwan
| | - Wan-Ting Hsu
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan; (H.-J.C.); (W.-T.H.)
| | - Cheng-Nan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600, Taiwan;
| | - Chin Li
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan; (H.-J.C.); (W.-T.H.)
- Correspondence: ; Tel.: 886-5-272-0411; Fax: 886-5-272-2871
| |
Collapse
|
42
|
Wan Y, Li W, Liao Z, Yan M, Chen X, Tang Z. Selective MMP-13 Inhibitors: Promising Agents for the Therapy of Osteoarthritis. Curr Med Chem 2020; 27:3753-3769. [PMID: 30556497 DOI: 10.2174/0929867326666181217153118] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/15/2018] [Accepted: 12/11/2018] [Indexed: 01/02/2023]
Abstract
Osteoarthritis (OA) is an age-related degenerative disease, which is characterized by chronic joint pain, inflammation and the damage of joint cartilage. At present, steroidal drugs and nonsteroidal anti-inflammatory drugs (NSAIDS), selective cyclooxygenase-2 (COX-2) inhibitors, are the first-line drugs for the treatment of OA. However, these drugs could lead to some cardiovascular side effects. Therefore, it is urgent to develop novel agents for the treatment of OA. Matrix metalloproteinase-13 (MMP-13), an important member of matrix metalloproteinases (MMPs) family, plays a vital role by degrading type II collagen in articular cartilage and bone in OA. It is noted that MMP-13 is specially expressed in the OA patients, and not in normal adults. In addition, broadspectrum MMP inhibitors could result in some painful and joint-stiffening side effects, called musculoskeletal syndrome (MSS) in the clinical trials. Thus, developing selective MMP-13 inhibitors is a potential strategy for the therapy of OA. In this review, we summarize the recent progress of selective MMP-13 inhibitors including two subfamilies, namely zinc-binding and non-zinc-binding selective MMP-13 inhibitors.
Collapse
Affiliation(s)
- Yichao Wan
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, Hunan Provincial Key Laboratory of Controllable Preparation and Functional Application of Fine Polymers, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China.,Hunan Provincial College Key Laboratory of QSAR/QSPR, Hunan Provincial Key Lab of Advanced Materials for New Energy Storage and conversion, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China
| | - Wei Li
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, Hunan Provincial Key Laboratory of Controllable Preparation and Functional Application of Fine Polymers, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China.,Hunan Provincial College Key Laboratory of QSAR/QSPR, Hunan Provincial Key Lab of Advanced Materials for New Energy Storage and conversion, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China
| | - Zhipeng Liao
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, Hunan Provincial Key Laboratory of Controllable Preparation and Functional Application of Fine Polymers, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China.,Hunan Provincial College Key Laboratory of QSAR/QSPR, Hunan Provincial Key Lab of Advanced Materials for New Energy Storage and conversion, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China
| | - Mi Yan
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Xuwang Chen
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Zilong Tang
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, Hunan Provincial Key Laboratory of Controllable Preparation and Functional Application of Fine Polymers, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China.,Hunan Provincial College Key Laboratory of QSAR/QSPR, Hunan Provincial Key Lab of Advanced Materials for New Energy Storage and conversion, Hunan University of Science and Technology, Xiangtan, Hunan 411201, China
| |
Collapse
|
43
|
Frischholz S, Berberich O, Böck T, Meffert RH, Blunk T. Resveratrol counteracts IL-1β-mediated impairment of extracellular matrix deposition in 3D articular chondrocyte constructs. J Tissue Eng Regen Med 2020; 14:897-908. [PMID: 32181981 DOI: 10.1002/term.3031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/05/2020] [Accepted: 03/06/2020] [Indexed: 11/09/2022]
Abstract
When aiming at cell-based therapies in osteoarthritis (OA), proinflammatory conditions mediated by cytokines such as IL-1β need to be considered. In recent studies, the phytoalexin resveratrol (RSV) has exhibited potent anti-inflammatory properties. However, long-term effects on 3D cartilaginous constructs under inflammatory conditions with regard to tissue quality, especially extracellular matrix (ECM) composition, have remained unexplored. Therefore, we employed long-term model cultures for cell-based therapies in an in vitro OA environment and evaluated effects of RSV. Pellet constructs made from expanded porcine articular chondrocytes were cultured with either IL-1β (1-10 ng/ml) or RSV (50 μM) alone, or a cotreatment with both agents. Treatments were applied for 14 days, either directly after pellet formation or after a preculture period of 7 days. Culture with IL-1β (10 ng/ml) decreased pellet size and DNA amount and severely compromised glycosaminoglycan (GAG) and collagen content. Cotreatment with RSV distinctly counteracted the proinflammatory catabolism and led to partial rescue of the ECM composition in both culture systems, with especially strong effects on GAG. Marked MMP13 expression was detected in IL-1β-treated pellets, but none upon RSV cotreatment. Expression of collagen type I was increased upon IL-1β treatment and still observed when adding RSV, whereas collagen type X, indicating hypertrophy, was detected exclusively in pellets treated with RSV alone. In conclusion, RSV can counteract IL-1β-mediated degradation and distinctly improve cartilaginous ECM deposition in 3D long-term inflammatory cultures. Nevertheless, potential hypertrophic effects should be taken into account when considering RSV as cotreatment for articular cartilage repair techniques.
Collapse
Affiliation(s)
- Sebastian Frischholz
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Oliver Berberich
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Thomas Böck
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Rainer H Meffert
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| |
Collapse
|
44
|
Abstract
Osteoarthritis (OA) is one of the most debilitating diseases and is associated with a high personal and socioeconomic burden. So far, there is no therapy available that effectively arrests structural deterioration of cartilage and bone or is able to successfully reverse any of the existing structural defects. Efforts to identify more tailored treatment options led to the development of strategies that enabled the classification of patient subgroups from the pool of heterogeneous phenotypes that display distinct common characteristics. To this end, the classification differentiates the structural endotypes into cartilage and bone subtypes, which are predominantly driven by structure-related degenerative events. In addition, further classifications have highlighted individuals with an increased inflammatory contribution (inflammatory phenotype) and pain-driven phenotypes as well as senescence and metabolic syndrome phenotypes. Most probably, it will not be possible to classify individuals by a single definite subtype, but it might help to identify groups of patients with a predominant pathology that would more likely benefit from a specific drug or cell-based therapy. Current clinical trials addressed mainly regeneration/repair of cartilage and bone defects or targeted pro-inflammatory mediators by intra-articular injections of drugs and antibodies. Pain was treated mostly by antagonizing nerve growth factor (NGF) activity and its receptor tropomyosin-related kinase A (TrkA). Therapies targeting metabolic disorders such as diabetes mellitus and senescence/aging-related pathologies are not specifically addressing OA. However, none of these therapies has been proven to modify disease progression significantly or successfully prevent final joint replacement in the advanced disease stage. Within this review, we discuss the recent advances in phenotype-specific treatment options and evaluate their applicability for use in personalized OA therapy.
Collapse
Affiliation(s)
- Susanne Grässel
- Department of Orthopedic Surgery, Exp. Orthopedics, ZMB/Biopark 1, Am Biopark 9, University of Regensburg, Regensburg, 93053, Germany
| | - Dominique Muschter
- Department of Orthopedic Surgery, Exp. Orthopedics, ZMB/Biopark 1, Am Biopark 9, University of Regensburg, Regensburg, 93053, Germany
| |
Collapse
|
45
|
Cowan KJ, Kleinschmidt-Dörr K, Gigout A, Moreau F, Kraines J, Townsend R, Dolgos H, DeMartino J. Translational strategies in drug development for knee osteoarthritis. Drug Discov Today 2020; 25:1054-1064. [PMID: 32251777 DOI: 10.1016/j.drudis.2020.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/03/2020] [Accepted: 03/23/2020] [Indexed: 10/24/2022]
Abstract
Osteoarthritis (OA) is a common disease worldwide with large unmet medical needs. To bring innovative treatments to OA patients, we at Merck have implemented a comprehensive strategy for drug candidate evaluation. We have a clear framework for decision-making in our preclinical pipeline, to design our clinical proof-of-concept trials for OA patients. We have qualified our strategy to define and refine dose and dosing regimen, for treatments administered either systemically or intra-articularly (IA). We do this through preclinical in vitro and in vivo studies, and by back-translating results from clinical studies in OA patients.
Collapse
Affiliation(s)
| | | | | | - Flavie Moreau
- EMD Serono Research and Development Institute, Billerica, MA, USA (A business of Merck, Darmstadt, Germany)
| | - Jeff Kraines
- EMD Serono Research and Development Institute, Billerica, MA, USA (A business of Merck, Darmstadt, Germany)
| | - Robert Townsend
- EMD Serono Research and Development Institute, Billerica, MA, USA (A business of Merck, Darmstadt, Germany)
| | | | - Julie DeMartino
- EMD Serono Research and Development Institute, Billerica, MA, USA (A business of Merck, Darmstadt, Germany)
| |
Collapse
|
46
|
Mohammadinejad R, Ashrafizadeh M, Pardakhty A, Uzieliene I, Denkovskij J, Bernotiene E, Janssen L, Lorite GS, Saarakkala S, Mobasheri A. Nanotechnological Strategies for Osteoarthritis Diagnosis, Monitoring, Clinical Management, and Regenerative Medicine: Recent Advances and Future Opportunities. Curr Rheumatol Rep 2020; 22:12. [PMID: 32248371 PMCID: PMC7128005 DOI: 10.1007/s11926-020-0884-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW In this review article, we discuss the potential for employing nanotechnological strategies for the diagnosis, monitoring, and clinical management of osteoarthritis (OA) and explore how nanotechnology is being integrated rapidly into regenerative medicine for OA and related osteoarticular disorders. RECENT FINDINGS We review recent advances in this rapidly emerging field and discuss future opportunities for innovations in enhanced diagnosis, prognosis, and treatment of OA and other osteoarticular disorders, the smart delivery of drugs and biological agents, and the development of biomimetic regenerative platforms to support cell and gene therapies for arresting OA and promoting cartilage and bone repair. Nanotubes, magnetic nanoparticles, and other nanotechnology-based drug and gene delivery systems may be used for targeting molecular pathways and pathogenic mechanisms involved in OA development. Nanocomposites are also being explored as potential tools for promoting cartilage repair. Nanotechnology platforms may be combined with cell, gene, and biological therapies for the development of a new generation of future OA therapeutics. Graphical Abstract.
Collapse
Affiliation(s)
- Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania
| | - Jaroslav Denkovskij
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania
| | - Lauriane Janssen
- Microelectronics Research Unit, Faculty of Information Technology and Electrical Engineering, University of Oulu, PL 4500, 3FI-90014, Oulu, Finland
| | - Gabriela S Lorite
- Microelectronics Research Unit, Faculty of Information Technology and Electrical Engineering, University of Oulu, PL 4500, 3FI-90014, Oulu, Finland
| | - Simo Saarakkala
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania.
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland.
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Centre for Sport, Exercise and Osteoarthritis Versus Arthritis, Queen's Medical Centre, Nottingham, UK.
- Sheik Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis with Stem Cells, King AbdulAziz University, Jeddah, Saudi Arabia.
- University Medical Center Utrecht, Department of Orthopedics and Department of Rheumatology & Clinical Immunology, 508 GA, Utrecht, The Netherlands.
| |
Collapse
|
47
|
Nanus DE, Wijesinghe SN, Pearson MJ, Hadjicharalambous MR, Rosser A, Davis ET, Lindsay MA, Jones SW. Regulation of the Inflammatory Synovial Fibroblast Phenotype by Metastasis-Associated Lung Adenocarcinoma Transcript 1 Long Noncoding RNA in Obese Patients With Osteoarthritis. Arthritis Rheumatol 2020; 72:609-619. [PMID: 31682073 DOI: 10.1002/art.41158] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To identify long noncoding RNAs (lncRNAs) associated with the inflammatory phenotype of synovial fibroblasts from obese patients with osteoarthritis (OA), and to explore the expression and function of these lncRNAs. METHODS Synovium was collected from normal-weight patients with hip fracture (non-OA; n = 6) and from normal-weight (n = 8) and obese (n = 8) patients with hip OA. Expression of RNA was determined by RNA-sequencing and quantitative reverse transcription-polymerase chain reaction. Knockdown of lncRNA was performed using LNA-based GapmeRs. Synovial fibroblast cytokine production was measured by enzyme-linked immunosorbent assay. RESULTS Synovial fibroblasts from obese patients with OA secreted greater levels of interleukin-6 (IL-6) (mean ± SEM 162 ± 21 pg/ml; P < 0.001) and CXCL8 (262 ± 67 pg/ml; P < 0.05) compared to fibroblasts from normal-weight patients with OA (IL-6, 51 ± 4 pg/ml; CXCL8, 78 ± 11 pg/ml) or non-OA patients (IL-6, 35 ± 3 pg/ml; CXCL8, 56 ± 6 pg/ml) (n = 6 patients per group). RNA-sequencing revealed that fibroblasts from obese OA patients exhibited an inflammatory transcriptome, with increased expression of proinflammatory messenger RNAs (mRNAs) as compared to that in fibroblasts from normal-weight OA or non-OA patients (>2-fold change, P < 0.05; n = 4 patients per group). A total of 19 lncRNAs were differentially expressed between normal-weight OA and non-OA patient fibroblasts, and a further 19 lncRNAs were differentially expressed in fibroblasts from obese OA patients compared to normal-weight OA patients (>2-fold change, P < 0.05 for each), which included the lncRNA for metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). MALAT1 was rapidly induced upon stimulation of OA synovial fibroblasts with proinflammatory cytokines, and was up-regulated in the synovium from obese OA patients as compared to normal-weight OA patients (1.6-fold change, P < 0.001) or non-OA patients (6-fold change, P < 0.001). MALAT1 knockdown in OA synovial fibroblasts (n = 4 patients) decreased the levels of mRNA expression and protein secretion of CXCL8 (>1.5-fold change, P < 0.01), whereas it increased expression of mRNAs for TRIM6 (>2-fold change, P < 0.01), IL7R (<2-fold change, P < 0.01), HIST1H1C (>1.5-fold change, P < 0.001), and MAML3 (>1.5-fold change, P < 0.001). In addition, MALAT1 knockdown inhibited the proliferation of synovial fibroblasts from obese patients with OA. CONCLUSION Synovial fibroblasts from obese patients with hip OA exhibit an inflammatory phenotype. MALAT1 lncRNA may mediate joint inflammation in obese OA patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mark A Lindsay
- University of Birmingham, Birmingham, UK, and University of Bath, Bath, UK
| | | |
Collapse
|
48
|
Veronesi F, Vandenbulcke F, Ashmore K, Di Matteo B, Nicoli Aldini N, Martini L, Fini M, Kon E. Meniscectomy-induced osteoarthritis in the sheep model for the investigation of therapeutic strategies: a systematic review. INTERNATIONAL ORTHOPAEDICS 2020; 44:779-793. [PMID: 32025798 DOI: 10.1007/s00264-020-04493-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE One of the major risk factors for OA is meniscectomy (Mx) that causes a rapid and progressive OA. Mx has been employed in various animal models, especially in large ones, to study preclinical safety and strategy effectiveness to counteract OA. The aim of the present study is to review in vivo studies, performed in sheep and published in the last ten years. METHODS The search strategy was performed in three websites: www.scopus.com, www.pubmed.com, and www.webofknowledge.com, using "Meniscectomy and osteoarthritis in sheep" keywords. RESULTS The 25 included studies performed unilateral total medial Mx (MMx), unilateral partial MMx, bilateral MMx, unilateral total lateral Mx (LMx), unilateral partial LMx, and bilateral LMx and MMx combined with anterior cruciate ligament transaction. The most frequently performed is the unilateral total MMx that increases changes in cartilage and subchondral bone more than the other techniques. Gross evaluations, histology, radiography, and biochemical tests are used to assess the degree of OA. The most widely tested treatments are related to scaffolds with or without mesenchymal stem cells. CONCLUSION OA therapeutic strategies require the use of large animal models due to similarities with human joint anatomy. A protocol for future in vivo studies on post-traumatic OA is clarified.
Collapse
Affiliation(s)
- Francesca Veronesi
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy.
| | - Filippo Vandenbulcke
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele - Milan, Italy.,Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano - Milan, Italy
| | - Kevin Ashmore
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele - Milan, Italy.,Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano - Milan, Italy
| | - Berardo Di Matteo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele - Milan, Italy.,Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano - Milan, Italy
| | - Nicolò Nicoli Aldini
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Lucia Martini
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Milena Fini
- Laboratory of Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Elizaveta Kon
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele - Milan, Italy.,Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano - Milan, Italy
| |
Collapse
|
49
|
Jansen MP, Mastbergen SC, van Heerwaarden RJ, Spruijt S, van Empelen MD, Kester EC, Lafeber FPJG, Custers RJH. Knee joint distraction in regular care for treatment of knee osteoarthritis: A comparison with clinical trial data. PLoS One 2020; 15:e0227975. [PMID: 31968005 PMCID: PMC6975543 DOI: 10.1371/journal.pone.0227975] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/03/2020] [Indexed: 01/10/2023] Open
Abstract
Objectives Knee joint distraction (KJD) has been evaluated as a joint-preserving treatment to postpone total knee arthroplasty in knee osteoarthritis patients in three clinical trials. Since 2014 the treatment is used in regular care in some hospitals, which might lead to a deviation from the original indication and decreased treatment outcome. In this study, baseline characteristics, complications and clinical benefit are compared between patients treated in regular care and in clinical trials. Methods In our hospital, 84 patients were treated in regular care for 6 weeks with KJD. Surgical details, complications, and range of motion were assessed from patient hospital charts. Patient-reported outcome measures were evaluated in regular care before and one year after treatment. Trial patients (n = 62) were treated and followed as described in literature. Results Patient characteristics were not significantly different between groups, except for distraction duration (regular care 45.3±4.3; clinical trials 48.1±8.1 days; p = 0.019). Pin tract infections were the most occurring complication (70% regular care; 66% clinical trials), but there was no significant difference in treatment complications between groups (p>0.1). The range of motion was recovered within a year after treatment for both groups. WOMAC questionnaires showed statistically and clinically significant improvement for both groups (both p<0.001 and >15 points in all subscales) and no significant differences between groups (all differences p>0.05). After one year, 70% of patients were responders (regular care 61%, trial 75%; p = 0.120). Neither regular care compared to clinical trial, nor any other characteristic could predict clinical response. Conclusions KJD as joint-preserving treatment in clinical practice, to postpone arthroplasty for end-stage knee osteoarthritis patient below the age of 65, results in an outcome similar to that thus far demonstrated in clinical trials. Longer follow-up in regular care is needed to test whether also long-term results remain beneficial and comparable to trial data.
Collapse
Affiliation(s)
- Mylène P Jansen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Simon C Mastbergen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ronald J van Heerwaarden
- Centre for Deformity Correction and Joint Preserving Surgery, Kliniek ViaSana, Mill, The Netherlands
| | - Sander Spruijt
- Department of Orthopedics, HagaZiekenhuis, Den Haag, The Netherlands
| | - Michelle D van Empelen
- Department of Orthopedic Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Esmee C Kester
- Department of Orthopedic Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Floris P J G Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel J H Custers
- Department of Orthopedic Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
50
|
Mobasheri A, Saarakkala S, Finnilä M, Karsdal MA, Bay-Jensen AC, van Spil WE. Recent advances in understanding the phenotypes of osteoarthritis. F1000Res 2019; 8. [PMID: 31885861 PMCID: PMC6913225 DOI: 10.12688/f1000research.20575.1] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2019] [Indexed: 01/15/2023] Open
Abstract
Recent research in the field of osteoarthritis (OA) has focused on understanding the underlying molecular and clinical phenotypes of the disease. This narrative review article focuses on recent advances in our understanding of the phenotypes of OA and proposes that the disease represents a diversity of clinical phenotypes that are underpinned by a number of molecular mechanisms, which may be shared by several phenotypes and targeted more specifically for therapeutic purposes. The clinical phenotypes of OA supposedly have different underlying etiologies and pathogenic pathways and they progress at different rates. Large OA population cohorts consist of a majority of patients whose disease progresses slowly and a minority of individuals whose disease may progress faster. The ability to identify the people with relatively rapidly progressing OA can transform clinical trials and enhance their efficiency. The identification, characterization, and classification of molecular phenotypes of rapidly progressing OA, which represent patients who may benefit most from intervention, could potentially serve as the basis for precision medicine for this disabling condition. Imaging and biochemical markers (biomarkers) are important diagnostic and research tools that can assist with this challenge.
Collapse
Affiliation(s)
- Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, 08661, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland.,Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, UK.,ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, DK-2730, Denmark
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland
| | - Mikko Finnilä
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland
| | - Morten A Karsdal
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, DK-2730, Denmark
| | | | - Willem Evert van Spil
- Division of Internal Medicine & Dermatology, Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Rheumatology, Dijklander Hospital, 1620 AR Hoorn, The Netherlands
| |
Collapse
|