1
|
Wei Y, Qian H, Zhang X, Wang J, Yan H, Xiao N, Zeng S, Chen B, Yang Q, Lu H, Xie J, Xie Z, Qin D, Li Z. Progress in multi-omics studies of osteoarthritis. Biomark Res 2025; 13:26. [PMID: 39934890 DOI: 10.1186/s40364-025-00732-y] [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: 10/05/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
Osteoarthritis (OA), a ubiquitous degenerative joint disorder, is marked by pain and disability, profoundly impacting patients' quality of life. As the population ages, the global prevalence of OA is escalating. Omics technologies have become instrumental in investigating complex diseases like OA, offering comprehensive insights into its pathogenesis and progression by uncovering disease-specific alterations across genomics, transcriptomics, proteomics, and metabolomics levels. In this review, we systematically analyzed and summarized the application and recent achievements of omics technologies in OA research by scouring relevant literature in databases such as PubMed. These studies have shed light on new potential therapeutic targets and biomarkers, charting fresh avenues for OA diagnosis and treatment. Furthermore, in our discussion, we highlighted the immense potential of spatial omics technologies in unraveling the molecular mechanisms of OA and in the development of novel therapeutic strategies, proposing future research directions and challenges. Collectively, this study encapsulates the pivotal advances in current OA research and prospects for future investigation, providing invaluable references for a deeper understanding and treatment of OA. This review aims to synthesize the recent progress of omics technologies in the realm of OA, aspiring to furnish theoretical foundations and research orientations for more profound studies of OA in the future.
Collapse
Affiliation(s)
- Yuanyuan Wei
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - He Qian
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Xiaoyu Zhang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jian Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Heguo Yan
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Niqin Xiao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Sanjin Zeng
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Bingbing Chen
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qianqian Yang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Hongting Lu
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jing Xie
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhaohu Xie
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| | - Zhaofu Li
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Mohd Noor NA, Jun Quan N, Mazlan NAAA, Nurul AA, Ahmad Mohd Zain MR, Azlan M. Synovial Fluid-Derived Exosomes from Osteoarthritis Patients Modulate Cell Surface Phenotypes of Monocytes and Cytokine Secretions. Immunol Invest 2024:1-16. [PMID: 39702926 DOI: 10.1080/08820139.2024.2443244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
BACKGROUND Exosomes can be found in the synovial fluid of inflamed knee joints, which play a significant role in osteoarthritis (OA) progression. However, their role - in modulating the cellular environment within the body, particularly monocytes remain unexplored. This study aimed to evaluate the immunomodulatory effect of exosomes on monocytes. METHODS Exosomes were isolated by ultracentrifugation and characterized using nanoparticle tracking analysis (NTA), scanning electron microscopy (SEM), and Western blot. The effect of exosomes in modulating monocyte phenotypes as well as cytokine secretion were further assessed in a co-culture condition using flow cytometry and ELISA accordingly. RESULTS Exosomes were identified as spherical particles with a size distribution ranging from 30 nm to 150 nm. These nanoparticles intensely expressed exosome protein markers including CD9, CD63, CD81, and HSP70. The expression of HLA-DR, CD14, and CD11b on monocytes decreased in the presence of exosomes after 24 h of incubation, regardless of the dose. Exosomes significantly induced the release of anti-inflammatory cytokines IL-1Ra in a time- and dose-dependent manner, while TNF-α secretion remains unchanged regardless of the presence or absence of exosomes. CONCLUSION This study highlights the immunoregulatory role of exosomes on monocytes, emphasizing the need for further studies into the underlying mechanism.
Collapse
Affiliation(s)
- Nur Azira Mohd Noor
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Ng Jun Quan
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Asma Abdullah Nurul
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Maryam Azlan
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
3
|
Xu Y, Szilagyi IA, Boer CG, Sedaghati-Khayat B, Visser WE, van Meurs JB, Chaker L. Association between thyroid function and osteoarthritis: A population-based cohort study. Osteoarthritis Cartilage 2024:S1063-4584(24)01450-X. [PMID: 39537017 DOI: 10.1016/j.joca.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 10/10/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Previous genetic and animal studies indicated a causal role of thyroid hormones in osteoarthritis (OA), which has not been observed in the general population. We aimed to investigate whether thyroid-stimulating hormone (TSH) and free thyroxine (FT4) were associated with hand, hip, or knee OA. METHODS Participants from the Rotterdam Study with baseline TSH, FT4, and joint radiographs were included. We used multivariable regression models to investigate the association of thyroid function with the prevalence, severity, incidence, and progression of OA. We conducted stratified analyses by sex, age, body mass index (BMI) and weight-bearing physical activity. RESULTS Among 9054 participants included (mean age 65 years, 56.3% women), higher FT4 concentrations were associated with an increased risk of prevalent knee OA (odd ratio [OR] 1.04 per pmol/L, 95% CI 1.01-1.06, corresponding to an OR of 1.62 across the reference range [i.e., 14pmol/L changes] of FT4) and more severe knee OA. There was a positive association between FT4 and overall progression of knee OA (OR 1.03 per pmol/L, 1.00-1.07). No association of TSH with hand, hip, or knee OA was identified. Stratified analysis revealed an association between FT4 and prevalent knee OA among individuals with BMI ≥30 kg/m2 (OR 1.05 per pmol/L, 1.01-1.08) and those with high levels of weight-bearing physical activity (OR 1.05 per pmol/L, 1.01-1.10). CONCLUSIONS Our study indicated that higher FT4 concentrations may increase the risk of knee OA. This association might be greater in individuals with extra joint loading, such as those with obesity.
Collapse
Affiliation(s)
- Yanning Xu
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ingrid A Szilagyi
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Cindy G Boer
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bahar Sedaghati-Khayat
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joyce B van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Layal Chaker
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
4
|
Zhang X, Wang Y, Zheng M, Wei Q, Zhang R, Zhu K, Zhai Q, Xu Y. IMPC-based screening revealed that ROBO1 can regulate osteoporosis by inhibiting osteogenic differentiation. Front Cell Dev Biol 2024; 12:1450215. [PMID: 39439909 PMCID: PMC11494888 DOI: 10.3389/fcell.2024.1450215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction The utilization of denosumab in treating osteoporosis highlights promising prospects for osteoporosis intervention guided by gene targets. While omics-based research into osteoporosis pathogenesis yields a plethora of potential gene targets for clinical transformation, identifying effective gene targets has posed challenges. Methods We first queried the omics data of osteoporosis clinical samples on PubMed, used International Mouse Phenotyping Consortium (IMPC) to screen differentially expressed genes, and conducted preliminary functional verification of candidate genes in human Saos2 cells through osteogenic differentiation and mineralization experiments. We then selected the candidate genes with the most significant effects on osteogenic differentiation and further verified the osteogenic differentiation and mineralization functions in mouse 3T3-E1 and bone marrow mesenchymal stem cells (BMSC). Finally, we used RNA-seq to explore the regulation of osteogenesis by the target gene. Results We identified PPP2R2A, RRBP1, HSPB6, SLC22A15, ADAMTS4, ATP8B1, CTNNB1, ROBO1, and EFR3B, which may contribute to osteoporosis. ROBO1 was the most significant regulator of osteogenesis in both human and mouse osteoblast. The inhibitory effect of Robo1 knockdown on osteogenic differentiation may be related to the activation of inflammatory signaling pathways. Conclusion Our study provides several novel molecular mechanisms involved in the pathogenesis of osteoporosis. ROBO1 is a potential target for osteoporosis intervention.
Collapse
Affiliation(s)
- Xiangzheng Zhang
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yike Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Zheng
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Wei
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ruizhi Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Keyu Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiaocheng Zhai
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Youjia Xu
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
5
|
Wang M, Zhang C, Yang Z, Cheng T, Lan C, Mo F. Muscle activation patterns and gait changes in unilateral knee osteoarthritis patients: a comparative study with healthy controls. Clin Rheumatol 2024; 43:2963-2972. [PMID: 39088118 DOI: 10.1007/s10067-024-07057-5] [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: 12/13/2023] [Revised: 06/23/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024]
Abstract
The objective of this study was to investigate the differences in muscle activation and kinematic parameters between patients with unilateral knee osteoarthritis (OA) and healthy individuals. Additionally, the study aimed to determine the correlation between muscle activation and kinematic parameters with knee OA symptoms. Participants with unilateral knee OA (n = 32) and healthy individuals (n = 32) completed the gait test. Electromyography (EMG) and motion capture were employed to collect muscle activation data and kinematic parameters. Spearman's correlation coefficient was used to analysis the correlation between BMI, symptomatic side EMG parameters, kinematic parameters, and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores. Furthermore, a multiple linear regression analysis of WOMAC pain was also conducted. The peak root mean square, integrated electromyography, and co-activation index (CCI) were increased bilaterally in the unilateral knee OA group compared to the healthy group. Furthermore, these values were higher on the symptomatic side than on the asymptomatic side. Compared with the healthy group, the knee OA group had lower gait speed, decreased stride length and cadence on bilateral sides, longer total stance time and double-stance time, and shorter single stance time and swing time. The maximum knee flexion angle of the swing phase on the symptomatic side of the knee OA group was smaller than that on the asymptomatic side and healthy group. Changes in EMG and gait parameters on the symptomatic side correlated with WOMAC scores. The main factors influencing WOMAC pain were the CCI values of the lateral femoral and biceps femoris muscles and gait speed. Muscle activation and kinematic parameters in the lower limbs of patients with unilateral knee OA were altered bilaterally during walking. These alterations on the symptomatic side were associated with knee OA-related pain. ChiCTR2200064958. Date of registration: 2022-10-24. Key Points • Unilateral symptomatic knee OA leads to bilateral alterations in muscle activation and gait parameters. • Symptomatic muscle activation and gait parameter changes in knee OA patients are associated with knee OA symptoms. • Correcting abnormal muscle activation conditions and gait training may reduce knee OA-related pain.
Collapse
Affiliation(s)
- Meiyi Wang
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Changjie Zhang
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zhi Yang
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Tiefeng Cheng
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, Hunan, China
| | - Chunna Lan
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| | - Fuhao Mo
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, Hunan, China.
| |
Collapse
|
6
|
Cheng F, Li M, Hua H, Zhang R, Zhu Y, Zhu Y, Zhang Y, Tong P. Identification of biomarkers and potential drug targets in osteoarthritis based on bioinformatics analysis and mendelian randomization. Front Pharmacol 2024; 15:1439289. [PMID: 39268462 PMCID: PMC11390638 DOI: 10.3389/fphar.2024.1439289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Background Osteoarthritis (OA) can lead to chronic joint pain, and currently there are no methods available for complete cure. Utilizing the Gene Expression Omnibus (GEO) database for bioinformatics analysis combined with Mendelian randomization (MR) has been widely employed for drug repurposing and discovery of novel therapeutic targets. Therefore, our research focus is to identify new diagnostic markers and improved drug target sites. Methods Gene expression data from different tissues of synovial membrane, cartilage and subchondral bone were collected through GEO data to screen out differential genes. Two-sample MR Analysis was used to estimate the causal effect of expression quantitative trait loci (eQTL) on OA. Through the intersection of the two, core genes were obtained, which were further screened by bioinformatics analysis for in vitro and in vivo molecular experimental verification. Finally, drug prediction and molecular docking further verified the medicinal value of drug targets. Results In the joint analysis utilizing the GEO database and MR approach, five genes exhibited significance across both analytical methods. These genes were subjected to bioinformatics analysis, revealing their close association with immunological functions. Further refinement identified two core genes (ARL4C and GAPDH), whose expression levels were found to decrease in OA pathology and exhibited a protective effect in the MR analysis, thus demonstrating consistent trends. Support from in vitro and in vivo molecular experiments was also obtained, while molecular docking revealed favorable interactions between the drugs and proteins, in line with existing structural data. Conclusion This study identified potential diagnostic biomarkers and drug targets for OA through the utilization of the GEO database and MR analysis. The findings suggest that the ARL4C and GAPDH genes may serve as therapeutic targets, offering promise for personalized treatment of OA.
Collapse
Affiliation(s)
- Feng Cheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Orthopedics, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mengying Li
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haotian Hua
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ruikun Zhang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yiwen Zhu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yingjia Zhu
- Department of Gynecology, Hangzhou Women's Hospital, Hangzhou, Zhejiang, China
| | - Yang Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Deng W, Zhou Y, Wan Q, Li L, Deng H, Yin Y, Zhou Q, Li Q, Cheng D, Hu X, Wang Y, Feng G. Nano-enzyme hydrogels for cartilage repair effectiveness based on ternary strategy therapy. J Mater Chem B 2024; 12:6242-6256. [PMID: 38842217 DOI: 10.1039/d4tb00307a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Designing artificial nano-enzymes for scavenging reactive oxygen species (ROS) in chondrocytes (CHOs) is considered the most feasible pathway for the treatment of osteoarthritis (OA). However, the accumulation of ROS due to the amount of nano-enzymatic catalytic site exposure and insufficient oxygen supply seriously threatens the clinical application of this therapy. Although metal-organic framework (MOF) immobilization of artificial nano-enzymes to enhance active site exposure has been extensively studied, artificial nano-enzymes/MOFs for ROS scavenging in OA treatment are still lacking. In this study, a biocompatible lubricating hydrogel-loaded iron-doped zeolitic imidazolate framework-8 (Fe/ZIF-8/Gel) centrase was engineered to scavenge endogenous overexpressed ROS synergistically generating dissolved oxygen and enhancing sustained lubrication for CHOs as a ternary artificial nano-enzyme. This property enabled the nano-enzymatic hydrogels to mitigate OA hypoxia and inhibit oxidative stress damage successfully. Ternary strategy-based therapies show excellent cartilage repair in vivo. The experimental results suggest that nano-enzyme-enhanced lubricating hydrogels are a potentially effective OA treatment and a novel strategy.
Collapse
Affiliation(s)
- Wei Deng
- Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 17 Gaopeng Avenue, Chengdu 610041, China.
- Department of Orthopedics, Pidu District People's Hospital, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, 611730, China
| | - Yue Zhou
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinlin Wan
- Medical College of Soochow University, Suzhou, 215123, China
| | - Lei Li
- National Engineering Research Center for Biomaterials & College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China.
| | - Hui Deng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yong Yin
- Department of Orthopedics, Pidu District People's Hospital, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, 611730, China
| | - Qingsong Zhou
- Department of Orthopedics, Pidu District People's Hospital, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, 611730, China
| | - Qiujiang Li
- Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 17 Gaopeng Avenue, Chengdu 610041, China.
| | - Duo Cheng
- Department of Orthopedics, Pidu District People's Hospital, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, 611730, China
| | - Xuefeng Hu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials & College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China.
| | - Ganjun Feng
- Department of Orthopedics Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 17 Gaopeng Avenue, Chengdu 610041, China.
| |
Collapse
|
8
|
Chen K, Zhao Y, Zhao W, Mao X, Li D, Wang Y, Shang S, Zhang H. Lubricating Microneedles System with Multistage Sustained Drug Delivery for the Treatment of Osteoarthritis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307281. [PMID: 38225701 DOI: 10.1002/smll.202307281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/31/2023] [Indexed: 01/17/2024]
Abstract
Osteoarthritis (OA) is a typical joint degenerative disease that is prevalent worldwide and significantly affects the normal activities of patients. Traditional treatments using diclofenac (DCF) as an anti-inflammatory drug by oral administration and transdermal delivery have many inherent deficiencies. In this study, a lubricating microneedles (MNs) system for the treatment of osteoarthritis with multistage sustained drug delivery and great reduction in skin damage during MNs penetration is developed. The bilayer dissolvable MNs system, namely HA-DCF@PDMPC, is prepared by designating the composite material of hyaluronic acid (HA) and covalently conjugated drug compound (HA-DCF) as the MNs tips and then modifying the surface of MNs tips with a self-adhesive lubricating copolymer (PDMPC). The MNs system is designed to achieve sustained drug release of DCF via ester bond hydrolysis, physical diffusion from MNs tips, and breakthrough of lubrication coating. Additionally, skin damage is reduced due to the presence of the lubrication coating on the superficial surface. Therefore, the lubricating MNs with multistage sustained drug delivery show good compliance as a transdermal patch for OA treatment, which is validated from anti-inflammatory cell tests and therapeutic animal experiments, down-regulating the expression levels of pro-inflammatory factors and alleviating articular cartilage destruction.
Collapse
Affiliation(s)
- Kexin Chen
- Center of Digital Dentistry/Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, NHC Research Center of Engineering and Technology for Computerized Dentistry, Beijing, 100081, China
- School of Nursing, Peaking of University, Beijing, 100191, China
| | - Yanlong Zhao
- State Key Laboratory of Tribology in Advanced Equipment, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Weiwei Zhao
- Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, China
| | - Xiaowei Mao
- State Key Laboratory of Tribology in Advanced Equipment, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Dan Li
- School of Nursing, Peaking of University, Beijing, 100191, China
| | - Yuguang Wang
- Center of Digital Dentistry/Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, NHC Research Center of Engineering and Technology for Computerized Dentistry, Beijing, 100081, China
| | - Shaomei Shang
- School of Nursing, Peaking of University, Beijing, 100191, China
| | - Hongyu Zhang
- State Key Laboratory of Tribology in Advanced Equipment, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
9
|
Pratt SJP, Plunkett CM, Kuzu G, Trinh T, Barbara J, Choconta P, Quackenbush D, Huynh T, Smith A, Barnes SW, New J, Pierce J, Walker JR, Mainquist J, King FJ, Elliott J, Hammack S, Decker RS. A high throughput cell stretch device for investigating mechanobiology in vitro. APL Bioeng 2024; 8:026129. [PMID: 38938688 PMCID: PMC11210978 DOI: 10.1063/5.0206852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Mechanobiology is a rapidly advancing field, with growing evidence that mechanical signaling plays key roles in health and disease. To accelerate mechanobiology-based drug discovery, novel in vitro systems are needed that enable mechanical perturbation of cells in a format amenable to high throughput screening. Here, both a mechanical stretch device and 192-well silicone flexible linear stretch plate were designed and fabricated to meet high throughput technology needs for cell stretch-based applications. To demonstrate the utility of the stretch plate in automation and screening, cell dispensing, liquid handling, high content imaging, and high throughput sequencing platforms were employed. Using this system, an assay was developed as a biological validation and proof-of-concept readout for screening. A mechano-transcriptional stretch response was characterized using focused gene expression profiling measured by RNA-mediated oligonucleotide Annealing, Selection, and Ligation with Next-Gen sequencing. Using articular chondrocytes, a gene expression signature containing stretch responsive genes relevant to cartilage homeostasis and disease was identified. The possibility for integration of other stretch sensitive cell types (e.g., cardiovascular, airway, bladder, gut, and musculoskeletal), in combination with alternative phenotypic readouts (e.g., protein expression, proliferation, or spatial alignment), broadens the scope of high throughput stretch and allows for wider adoption by the research community. This high throughput mechanical stress device fills an unmet need in phenotypic screening technology to support drug discovery in mechanobiology-based disease areas.
Collapse
Affiliation(s)
- Stephen J. P. Pratt
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | | | - Guray Kuzu
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Ton Trinh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joshua Barbara
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Paula Choconta
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Doug Quackenbush
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Truc Huynh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Anders Smith
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - S. Whitney Barnes
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joel New
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Pierce
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - John R. Walker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Mainquist
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Frederick J. King
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Jimmy Elliott
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Scott Hammack
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Rebekah S. Decker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| |
Collapse
|
10
|
Yi G, Zhang R, Li M, Song X, Li S. Atractylenolide-III attenuates osteoarthritis by repolarizing macrophages through inactivating TLR4/NF-κB signaling. Int Immunopharmacol 2024; 129:111629. [PMID: 38346377 DOI: 10.1016/j.intimp.2024.111629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND As a common chronic musculoskeletal condition, osteoarthritis (OA) presently lacks particular treatment strategies. The aim of this study was to examine how AT-III therapies affected macrophage repolarity in order to slow down the advancement of OA. METHODS RAW264.7 macrophages were polarized to M1 subtypes then administered with different concentrations of AT-III. Immunofluorescence, qRT-PCR and flow cytometry were used to assess the polarization of the macrophages. The mechanism of AT-III repolarize macrophages was evaluated by western blot. Furthermore, the effects of macrophage conditioned media (CM) on the migration, proliferation, and chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) were investigated using CCK-8 assays, the scratch test, and alcian blue staining. The effects of macrophage CM on chondrocyte proliferation and degeneration were investigated using CCK-8 and qRT-PCR. In vivo micro-CT and histological observations were performed on rats with anterior cruciate ligament transection and partial medial meniscectomy, either with or without AT-III treatment. RESULTS AT-III repolarized M1 macrophages to M2 phenotype. Mechanistically, AT-III reduced the expression of Toll-like receptor(TLR) 4 induced by lipopolysaccharide in RAW264.7 and lowered nuclear factor-κB (NF-κB) signaling molecules p-p65 and p-IκBα. The TLR4 agonist RS09 reversed the effects of AT-III on macrophage repolarization. AT-III-induced macrophages CM stimulated BMSCs migration, proliferation and chondrogenic differentiation. AT-III-treated macrophage CM promoted chondrocyte proliferation while inhibiting chondrocyte degeneration. In vivo, AT-III treatment alleviated the degree of synovitis, inhibited subchondral bone remodeling and reduced cartilage destruction in the rat OA model. CONCLUSIONS AT-III attenuates OA by repolarizing macrophages through inactivating TLR4/NF-κB signaling. These data suggest that AT-III may be an effective therapeutic candidate for OA treatment.
Collapse
Affiliation(s)
- Guoliang Yi
- Guizhou Medical University, Guiyang 550004, China; Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Runmin Zhang
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Min Li
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xizheng Song
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Siming Li
- Guizhou Medical University, Guiyang 550004, China; Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou 510220, China.
| |
Collapse
|
11
|
Li Y, Yang J, Chen X, Hu H, Lan N, Zhao J, Zheng L. Mitochondrial-targeting and NIR-responsive Mn 3O 4@PDA@Pd-SS31 nanozymes reduce oxidative stress and reverse mitochondrial dysfunction to alleviate osteoarthritis. Biomaterials 2024; 305:122449. [PMID: 38194734 DOI: 10.1016/j.biomaterials.2023.122449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
Mitochondrial reactive oxygen species (mROS) play a crucial role in the process of osteoarthritis (OA), which may be a promising target for therapy of OA. In this study, novel mitochondrial-targeting and SOD-mimic Mn3O4@PDA@Pd-SS31 nanozymes with near-infrared (NIR) responsiveness and synergistic cascade to scavenge mROS were designed for the therapy of OA. Results showed that the nanozymes accelerated the release of Pd and Mn3O4 under NIR irradiation, exhibiting enhanced activities of SOD and CAT mimic enzymes with reversed mitochondrial dysfunction and promoted mitophagy to effectively scavenge mROS from chondrocytes, modulate the microenvironment of oxidative stress, and eventually inhibit the inflammatory response. Nanozymes were excreted in vivo through intestinal metabolic pathway and had good biocompatibility, effectively reducing the inflammatory response and relieving articular cartilage degeneration in OA joints, with a reduction of 93.7 % and 93.8 % in OARSCI scores for 4 and 8 weeks respectively. Thus, this study demonstrated that the mitochondria targeting and NIR responsive Mn3O4@PDA@Pd-SS31 nanozymes could efficiently scavenge mROS, repair damaged mitochondrial function and promote cartilage regeneration, which are promising for the treatment of OA in clinical applications.
Collapse
Affiliation(s)
- Yuquan Li
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530005, China
| | - Junxu Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiaoming Chen
- Department of Spine Osteopathia, The First Affifiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hao Hu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441100, China
| | - Nihan Lan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
12
|
Iacobescu GL, Iacobescu L, Popa MIG, Covache-Busuioc RA, Corlatescu AD, Cirstoiu C. Genomic Determinants of Knee Joint Biomechanics: An Exploration into the Molecular Basis of Locomotor Function, a Narrative Review. Curr Issues Mol Biol 2024; 46:1237-1258. [PMID: 38392197 PMCID: PMC10888373 DOI: 10.3390/cimb46020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
In recent years, the nexus between genetics and biomechanics has garnered significant attention, elucidating the role of genomic determinants in shaping the biomechanical attributes of human joints, specifically the knee. This review seeks to provide a comprehensive exploration of the molecular basis underlying knee joint locomotor function. Leveraging advancements in genomic sequencing, we identified specific genetic markers and polymorphisms tied to key biomechanical features of the knee, such as ligament elasticity, meniscal resilience, and cartilage health. Particular attention was devoted to collagen genes like COL1A1 and COL5A1 and their influence on ligamentous strength and injury susceptibility. We further investigated the genetic underpinnings of knee osteoarthritis onset and progression, as well as the potential for personalized rehabilitation strategies tailored to an individual's genetic profile. We reviewed the impact of genetic factors on knee biomechanics and highlighted the importance of personalized orthopedic interventions. The results hold significant implications for injury prevention, treatment optimization, and the future of regenerative medicine, targeting not only knee joint health but joint health in general.
Collapse
Affiliation(s)
- Georgian-Longin Iacobescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Loredana Iacobescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Mihnea Ioan Gabriel Popa
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Antonio-Daniel Corlatescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Catalin Cirstoiu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| |
Collapse
|
13
|
Liu ZF, Zhang Y, Liu J, Wang YY, Chen M, Liu EY, Guo JM, Wang YH, Weng ZW, Liu CX, Yu CH, Wang XY. Effect of Traditional Chinese Non-Pharmacological Therapies on Knee Osteoarthritis: A Narrative Review of Clinical Application and Mechanism. Orthop Res Rev 2024; 16:21-33. [PMID: 38292459 PMCID: PMC10826518 DOI: 10.2147/orr.s442025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
Knee osteoarthritis (KOA) stands as a degenerative ailment with a substantial and escalating prevalence. The practice of traditional Chinese non-pharmacological therapy has become a prevalent complementary and adjunctive approach. A mounting body of evidence suggests its efficacy in addressing KOA. Recent investigations have delved into its underlying mechanism, yielding some headway. Consequently, this comprehensive analysis seeks to encapsulate the clinical application and molecular mechanism of traditional Chinese non-pharmacological therapy in KOA treatment. The review reveals that various therapies, such as acupuncture, electroacupuncture, warm needle acupuncture, tuina, and acupotomy, primarily target localized knee components like cartilage, subchondral bone, and synovium. Moreover, their impact extends to the central nervous system and intestinal flora. More perfect experimental design and more comprehensive research remain a promising avenue in the future.
Collapse
Affiliation(s)
- Zhi-Feng Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yang Zhang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Jing Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yu-Yan Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Mo Chen
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Er-Yang Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Jun-Ming Guo
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yan-Hua Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Zhi-Wen Weng
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Chang-Xin Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Chang-He Yu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Xi-You Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| |
Collapse
|
14
|
He L, Xu Z, Niu X, Li R, Wang F, You Y, Gao J, Zhao L, Shah KM, Fan J, Liu M, Luo J. GPRC5B protects osteoarthritis by regulation of autophagy signaling. Acta Pharm Sin B 2023; 13:2976-2989. [PMID: 37521864 PMCID: PMC10372909 DOI: 10.1016/j.apsb.2023.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/17/2023] [Accepted: 03/14/2023] [Indexed: 08/01/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common chronic diseases in the world. However, current treatment modalities mainly relieve pain and inhibit cartilage degradation, but do not promote cartilage regeneration. In this study, we show that G protein-coupled receptor class C group 5 member B (GPRC5B), an orphan G-protein-couple receptor, not only inhibits cartilage degradation, but also increases cartilage regeneration and thereby is protective against OA. We observed that Gprc5b deficient chondrocytes had an upregulation of cartilage catabolic gene expression, along with downregulation of anabolic genes in vitro. Furthermore, mice deficient in Gprc5b displayed a more severe OA phenotype in the destabilization of the medial meniscus (DMM) induced OA mouse model, with upregulation of cartilage catabolic factors and downregulation of anabolic factors, consistent with our in vitro findings. Overexpression of Gprc5b by lentiviral vectors alleviated the cartilage degeneration in DMM-induced OA mouse model by inhibiting cartilage degradation and promoting regeneration. We also assessed the molecular mechanisms downstream of Gprc5b that may mediate these observed effects and identify the role of protein kinase B (AKT)-mammalian target of rapamycin (mTOR)-autophagy signaling pathway. Thus, we demonstrate an integral role of GPRC5B in OA pathogenesis, and activation of GPRC5B has the potential in preventing the progression of OA.
Collapse
Affiliation(s)
- Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Ziwei Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xin Niu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Rong Li
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Fanhua Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Yu You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jingduo Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lei Zhao
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| | - Karan M. Shah
- Department of Oncology and Metabolism, the Medical School, the University of Sheffield, Sheffield S10 2TN, UK
| | - Jian Fan
- Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai 201613, China
| |
Collapse
|
15
|
Richard D, Capellini TD, Diekman BO. Epigenetics as a mediator of genetic risk in osteoarthritis: role during development, homeostasis, aging, and disease progression. Am J Physiol Cell Physiol 2023; 324:C1078-C1088. [PMID: 36971423 PMCID: PMC10191130 DOI: 10.1152/ajpcell.00574.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
The identification of genomic loci that are associated with osteoarthritis (OA) has provided a starting point for understanding how genetic variation activates catabolic processes in the joint. However, genetic variants can only alter gene expression and cellular function when the epigenetic environment is permissive to these effects. In this review, we provide examples of how epigenetic shifts at distinct life stages can alter the risk for OA, which we posit is critical for the proper interpretation of genome-wide association studies (GWAS). During development, intensive work on the growth and differentiation factor 5 (GDF5) locus has revealed the importance of tissue-specific enhancer activity in controlling both joint development and the subsequent risk for OA. During homeostasis in adults, underlying genetic risk factors may help establish beneficial or catabolic "set points" that dictate tissue function, with a strong cumulative effect on OA risk. During aging, methylation changes and the reorganization of chromatin can "unmask" the effects of genetic variants. The destructive function of variants that alter aging would only mediate effects after reproductive competence and thus avoid any evolutionary selection pressure, as consistent with larger frameworks of biological aging and its relationship to disease. A similar "unmasking" may occur during OA progression, which is supported by the finding of distinct expression quantitative trait loci (eQTLs) in chondrocytes depending on the degree of tissue degradation. Finally, we propose that massively parallel reporter assays (MPRAs) will be a valuable tool to test the function of putative OA GWAS variants in chondrocytes from different life stages.
Collapse
Affiliation(s)
- Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, United States
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
16
|
Deng Y, Han Y, Gao S, Dong W, Yu Y. The Physiological Functions and Polymorphisms of Type II Deiodinase. Endocrinol Metab (Seoul) 2023; 38:190-202. [PMID: 37150515 PMCID: PMC10164501 DOI: 10.3803/enm.2022.1599] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/21/2023] [Indexed: 05/09/2023] Open
Abstract
Type II deiodinase (DIO2) is thought to provide triiodothyronine (T3) to the nucleus to meet intracellular needs by deiodinating the prohormone thyroxine. DIO2 is expressed widely in many tissues and plays an important role in a variety of physiological processes, such as controlling T3 content in developing tissues (e.g., bone, muscles, and skin) and the adult brain, and regulating adaptive thermogenesis in brown adipose tissue (BAT). However, the identification and cloning of DIO2 have been challenging. In recent years, several clinical investigations have focused on the Thr92Ala polymorphism, which is closely correlated with clinical syndromes such as type 2 diabetes, obesity, hypertension, and osteoarthritis. Thr92Ala-DIO2 was also found to be related to bone and neurodegenerative diseases and tumors. However, relatively few reviews have synthesized research on individual deiodinases, especially DIO2, in the past 5 years. This review summarizes current knowledge regarding the physiological functions of DIO2 in thyroid hormone signaling and adaptive thermogenesis in BAT and the brain, as well as the associations between Thr92Ala-DIO2 and bone and neurodegenerative diseases and tumors. This discussion is expected to provide insights into the physiological functions of DIO2 and the clinical syndromes associated with Thr92Ala-DIO2.
Collapse
Affiliation(s)
- Yan Deng
- Department of Histology and Embryology, School of Basic Medical Sciences, Southwest Medical University, China
| | - Yi Han
- Department of Histology and Embryology, School of Basic Medical Sciences, Southwest Medical University, China
| | - Sheng Gao
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yang Yu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southwest Medical University, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
17
|
Huang H, Dong X, Mao K, Pan W, Nie B, Jiang L. Identification of key candidate genes and pathways in rheumatoid arthritis and osteoarthritis by integrated bioinformatical analysis. Front Genet 2023; 14:1083615. [PMID: 36861127 PMCID: PMC9968929 DOI: 10.3389/fgene.2023.1083615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are the most common joint disorders. Although they have shown analogous clinical manifestations, the pathogenesis of RA and OA are different. In this study, we used the online Gene Expression Omnibus (GEO) microarray expression profiling dataset GSE153015 to identify gene signatures between RA and OA joints. The relevant data on 8 subjects obtained from large joints of RA patients (RA-LJ), 8 subjects obtained from small joints of RA patients (RA-SJ), and 4 subjects with OA were investigated. Differentially expressed genes (DEGs) were screened. Functional enrichment analysis of DEGs including the Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified, which were mainly associated with T cell activation or chemokine activity. Besides, protein-protein interaction (PPI) network analysis was performed, and key modules were identified. Hub genes of RA-LJ and OA groups were screened, they were CD8A, GZMB, CCL5, CD2, and CXCL9, whereas CD8A, CD2, IL7R, CD27, and GZMB were hub genes of RA-SJ and OA group. The novel DEGs and functional pathways between RA and OA identified in this study may provide new insight into the underlying molecular mechanisms and therapeutic strategies of RA and OA.
Collapse
Affiliation(s)
- Huijing Huang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyi Dong
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kaimin Mao
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wanwan Pan
- Yankuang New Journey General Hospital, Jingning, Shandong, China
| | - Bin’en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lindi Jiang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China,*Correspondence: Lindi Jiang,
| |
Collapse
|
18
|
Cassidy A, Onal M, Pelletier S. Novel methods for the generation of genetically engineered animal models. Bone 2023; 167:116612. [PMID: 36379415 PMCID: PMC9936561 DOI: 10.1016/j.bone.2022.116612] [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: 09/06/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/15/2022]
Abstract
Genetically modified mouse models have shaped our understanding of biological systems in both physiological and pathological conditions. For decades, mouse genome engineering has relied on transgenesis and spontaneous gene replacement in embryonic stem (ES) cells. While these technologies provided a wealth of knowledge, they remain imprecise and expensive to use. Recent advances in genome editing technologies such as the development of targetable nucleases, the improvement of delivery systems, and the simplification of targeting strategies now allow for the rapid, precise manipulation of the mouse genome. In this review article, we discuss novel methods and targeting strategies for the generation of mouse models for the study of bone and skeletal muscle biology.
Collapse
Affiliation(s)
- Annelise Cassidy
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Melda Onal
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephane Pelletier
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
19
|
Miotla-Zarebska J, Parisi I, Batchelor V, Vincent TL, Potter PK. Modeling and Assessing Osteoarthritis in Mice by Destabilization of the Medial Meniscus (DMM). Methods Mol Biol 2023; 2598:357-373. [PMID: 36355305 DOI: 10.1007/978-1-0716-2839-3_26] [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] [Indexed: 06/16/2023]
Abstract
In this chapter, we describe an induced model of osteoarthritis in mice, frequently employed in the study of this disease. We outline in detail the surgical induction of disease and preparation of samples for histological assessment of disease.
Collapse
Affiliation(s)
- Jadwiga Miotla-Zarebska
- Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Ida Parisi
- Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Vicky Batchelor
- Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Paul K Potter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK.
| |
Collapse
|
20
|
Chang B, Hu Z, Chen L, Jin Z, Yang Y. Development and validation of cuproptosis-related genes in synovitis during osteoarthritis progress. Front Immunol 2023; 14:1090596. [PMID: 36817415 PMCID: PMC9932029 DOI: 10.3389/fimmu.2023.1090596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common refractory degenerative joint diseases worldwide. Synovitis is believed to drive joint cartilage destruction during OA pathogenesis. Cuproptosis is a novel form of copper-induced cell death. However, few studies have examined the correlations between cuproptosis-related genes (CRGs), immune infiltration, and synovitis. Therefore, we analyzed CRGs in synovitis during OA. Microarray datasets (GSE55235, GSE55457, GSE12021, GSE82107 and GSE176308) were downloaded from the Gene Expression Omnibus database. Next, we conducted differential and subtype analyses of CRGs across synovitis. Immune infiltration and correlation analyses were performed to explore the association between CRGs and immune cell abundance in synovitis. Finally, single-cell RNA-seq profiling was performed using the GSE176308 dataset to investigate the expression of CRGs in the various cell clusters. We found that the expression of five CRGs (FDX1, LIPT1, PDHA1, PDHB, and CDKN2A) was significantly increased in the OA synovium. Moreover, abundant and various types of immune cells infiltrated the synovium during OA, which was correlated with the expression of CRGs. Additionally, single-cell RNA-seq profiling revealed that the cellular composition of the synovium was complex and that their proportions varied greatly as OA progressed. The expression of CRGs differed across various cell types in the OA synovium. The current study predicted that cuproptosis may be involved in the pathogenesis of synovitis. The five screened CRGs (FDX1, LIPT1, PDHA1, PDHB, and CDKN2A) could be explored as candidate biomarkers or therapeutic targets for OA synovitis.
Collapse
Affiliation(s)
- Bohan Chang
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhehan Hu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liang Chen
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhuangzhuang Jin
- Department of Emergence Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Yang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
21
|
Beier F. The impact of omics research on our understanding of osteoarthritis and future treatments. Curr Opin Rheumatol 2023; 35:55-60. [PMID: 36350386 DOI: 10.1097/bor.0000000000000919] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE OF REVIEW To review recent studies using 'Omics' approaches (genomics, proteomics, metabolomics, single cell analyses) in patient populations and animal models of osteoarthritis (OA), with the goal of identifying disease-modifying mechanisms that could serve as therapeutic and diagnostic targets. RECENT FINDINGS The number of genes, pathways and molecules with potential roles in OA pathogenesis has grown substantially over the last 18 months. Studies have expanded from their traditional focus on cartilage and gene expression to other joint tissues, proteins and metabolites. Single cell approaches provide unprecedented resolution and exciting insights into the heterogeneity of cellular activities in OA. Functional validation and investigation of underlying mechanisms in animal models of OA, in particular genetically engineered mice, link Omics findings to pathophysiology and potential therapeutic applications. SUMMARY Although great progress has been made in the use of Omics approaches to OA, in both animal models and patient samples, much work remains to be done. In addition to filling gaps in data sets not yet existing, integration of data from the various approaches, mechanistic investigations, and linkage of Omics data to patient stratification remain significant challenges.
Collapse
Affiliation(s)
- Frank Beier
- Department of Physiology and Pharmacology, Western University Bone and Joint Institute, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
22
|
Zhang Z, Liu W, Xiong J, Chen T, Jiang L, Liu M. Candidate Marker Genes for Diagnosis of Osteoarthritis and Prediction of Their Regulatory Mechanisms. Folia Biol (Praha) 2023; 69:22-33. [PMID: 37962028 DOI: 10.14712/fb2023069010022] [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] [Indexed: 11/15/2023]
Abstract
We have screened candidate marker genes for the diagnosis of osteoarthritis and predicted their regulatory mechanisms. Six expression chips of tissue samples and one expression chip of peripheral blood mononuclear cell (PMBC) samples were obtained from the GEO database. Differential analysis, GSEA, and WGCNA were performed on the integra-ted tissue sample data with batch correction. Can-didate genes were obtained from the intersection of the genes significantly related to osteoarthritis in the WGCNA and the differentially expressed genes. ROC analysis was performed on the candidate genes in the tissue and PMBC samples. Genes with AUC values greater than 0.6 were retained as final candidates, and their upstream regulatory miRNAs were predicted. A total of 106 genes with differential expression were found in osteoarthritis tissue samples, which were mainly enriched in cell cycle and p53 signalling pathways. WGCNA selected a gene module significantly correlated with the occurrence of osteoarthritis. Fourteen candidate genes were obtained from the intersection of the genes in the module and the differentially expressed genes. ROC analysis showed that among these 14 candidate genes, only ADM, CX3CR1 and GADD45A had AUC values greater than 0.6 in both tissue and PMBC samples. The AUC values of the gene set of these three genes were greater than 0.7. Multiple miRNAs were predicted to be regulators of these three genes. ADM, CX3CR1 and GADD45A have potential as diagnostic marker genes for osteoarthritis and may be regulated by multiple miRNAs.
Collapse
Affiliation(s)
- Zuyang Zhang
- Department of Orthopedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410004, China
| | - Wei Liu
- Department of Orthopedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410004, China
| | - Jiepeng Xiong
- Department of Orthopedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410004, China
| | - Tianhua Chen
- Department of Orthopedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410004, China
| | - Liangdong Jiang
- Department of Orthopedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410004, China.
| | - Mingjiang Liu
- Department of Orthopedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410004, China.
| |
Collapse
|
23
|
Wang Y, Zhou W, Chen Y, He D, Qin Z, Wang Z, Liu S, Zhou L, Su J, Zhang C. Identification of susceptibility modules and hub genes of osteoarthritis by WGCNA analysis. Front Genet 2022; 13:1036156. [DOI: 10.3389/fgene.2022.1036156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/19/2022] [Indexed: 11/19/2022] Open
Abstract
Osteoarthritis (OA) is a major cause of pain, disability, and social burden in the elderly throughout the world. Although many studies focused on the molecular mechanism of OA, its etiology remains unclear. Therefore, more biomarkers need to be explored to help early diagnosis, clinical outcome measurement, and new therapeutic target development. Our study aimed to retrieve the potential hub genes of osteoarthritis (OA) by weighted gene co-expression network analysis (WGCNA) and assess their clinical utility for predicting OA. Here, we integrated WGCNA to identify novel OA susceptibility modules and hub genes. In this study, we first selected 477 and 834 DEGs in the GSE1919 and the GSE55235 databases, respectively, from the Gene Expression Omnibus (GEO) website. Genes with p-value<0.05 and | log2FC | > 1 were included in our analysis. Then, WGCNA was conducted to build a gene co-expression network, which filtered out the most relevant modules and screened out 23 overlapping WGCNA-derived hub genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses elucidated that these hub genes were associated with cell adhesion molecules pathway, leukocyte activation, and inflammatory response. In addition, we conducted the protein–protein interaction (PPI) network in 23 hub genes, and the top four upregulated hub genes were sorted out (CD4, SELL, ITGB2, and CD52). Moreover, our nomogram model showed good performance in predicting the risk of OA (C-index = 0.76), and this model proved to be efficient in diagnosis by ROC curves (AUC = 0.789). After that, a single-sample gene set enrichment (ssGSEA) analysis was performed to discover immune cell infiltration in OA. Finally, human primary synoviocytes and immunohistochemistry study of synovial tissues confirmed that those candidate genes were significantly upregulated in the OA groups compared with normal groups. We successfully constructed a co-expression network based on WGCNA and found out that OA-associated susceptibility modules and hub genes, which may provide further insight into the development of pre-symptomatic diagnosis, may contribute to understanding the molecular mechanism study of OA risk genes.
Collapse
|
24
|
Theaflavin-3,3 -Digallate Protects Cartilage from Degradation by Modulating Inflammation and Antioxidant Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3047425. [PMID: 35847580 PMCID: PMC9286955 DOI: 10.1155/2022/3047425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/15/2022] [Indexed: 12/25/2022]
Abstract
Background Osteoarthritis (OA) is a common degenerative joint disease that may be closely linked to inflammation and oxidative stress destroying the balance of cartilage matrix. Theaflavin-3,3′-digallate (TFDG), a natural substance derived from black tea, has been reported to restrict the activity of inflammatory cytokines and effectively eliminate reactive oxygen species (ROS) in various diseases. However, it is not clear whether TFDG can improve OA. Methods Chondrocytes were treated with or without IL-1β and 20 μM and 40 μM TFDG. The effect of TFDG on the proliferation of chondrocytes was detected by CCK8. RT-qPCR was used to detect the gene expression of inflammatory factors, extracellular matrix synthesis, and degradation genes. Western blot and immunofluorescence assays were used to detect the protein expression. The fluorescence intensity of reactive oxygen species labeled by DCFH-DA was detected by flow cytometry. We established an OA rat model by performing destabilized medial meniscus (DMM) surgery to observe whether TFDG can protect chondrocytes under arthritis in vivo. Results TFDG was found to inhibit proinflammatory factors (IL-6, TNF-α, iNOS, and PGE) and matrix-degrading enzymes (MMP13, MMP3, and ADAMTS5) expression and protected extracellular matrix components of chondrocytes (ACAN, COL2, and SOX9). TFDG accelerated the scavenging of ROS caused by IL-1β according to the Nrf2 signaling pathway activation. At the same time, TFDG suppressed the PI3K/AKT/NF-κB and MAPK signaling pathways to delay the inflammatory process. The cartilage of DMM rats receiving TFDG showed lower Osteoarthritis Research Society International (OARSI) scores and expressed higher levels of COL2 and Nrf2 compared with those of rats in the DMM group. Conclusion TFDG could protect cartilage from degradation and alleviate osteoarthritis in rats, which suggests that TFDG has potential as a drug candidate for OA therapy.
Collapse
|
25
|
Tong J, Chen Z, Sun G, Zhou J, Zeng Y, Zhong P, Deng C, Chen X, Liu L, Wang S, Chen J, Liao Y. The Efficacy of Pulsed Electromagnetic Fields on Pain, Stiffness, and Physical Function in Osteoarthritis: A Systematic Review and Meta-Analysis. Pain Res Manag 2022; 2022:9939891. [PMID: 35586276 PMCID: PMC9110240 DOI: 10.1155/2022/9939891] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/02/2022] [Indexed: 11/17/2022]
Abstract
Background Although there are many pharmacological interventions for adults with osteoarthritis (OA) who do not meet the indications for surgery, side effects and adverse effects cannot be ignored. Physical interventions are known for their effectiveness and safety, and pulsed electromagnetic fields (PEMFs) have already been applied to skeletal diseases such as osteoporosis. Objective In this systematic review and meta-analysis, we aimed to assess the efficacy of PEMF on the major symptoms of patients with OA compared with efficacy of other interventions. Methods Randomized controlled trials (RCTs) investigating OA patients treated with PEMF and with pain, stiffness, and physical function impairment since 2009 were included. The Visual Analog Scale (VAS) and Western Ontario McMaster Universities Osteoarthritis Index (WOMAC) scores were used for assessment. All extracted data were analyzed using RevMan V.5.3. Results Eleven RCTs consisting of 614 patients were enrolled in this meta-analysis, of which 10 trials comprised knee OA and one comprised hand OA. Compared with the control groups, the PEMF treatment yielded a more favorable output. PEMF alleviated pain (standardized mean differences [SMD] = 0.71, 95% confidence interval [CI]: 0.08-1.34, p = 0.03), improved stiffness (SMD = 1.34, 95% CI: 0.45-2.23,p=0.003), and restored physical function (SMD = 1.52, 95% CI: 0.49-2.55,p=0.004). Conclusions PEMF therapy ameliorates OA symptoms such as pain, stiffness, and physical function in patients compared to other conservative treatments. There is an urgent need to search for different types of OA in multiple locations.
Collapse
Affiliation(s)
- Jie Tong
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhengyu Chen
- Department of Spine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guanghua Sun
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jun Zhou
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ye Zeng
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Peirui Zhong
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Chengyuan Deng
- Department of Anatomy, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 421001, China
| | - Xiaocui Chen
- Department of Rehabilitation, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Liu Liu
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shiyong Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jiaqian Chen
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ying Liao
- Rehabilitation Medicine Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
26
|
Aubourg G, Rice SJ, Bruce-Wootton P, Loughlin J. Genetics of osteoarthritis. Osteoarthritis Cartilage 2022; 30:636-649. [PMID: 33722698 PMCID: PMC9067452 DOI: 10.1016/j.joca.2021.03.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/17/2021] [Accepted: 03/06/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis genetics has been transformed in the past decade through the application of large-scale genome-wide association scans. So far, over 100 polymorphic DNA variants have been associated with this common and complex disease. These genetic risk variants account for over 20% of osteoarthritis heritability and the vast majority map to non-protein coding regions of the genome where they are presumed to act by regulating the expression of target genes. Statistical fine mapping, in silico analyses of genomics data, and laboratory-based functional studies have enabled the identification of some of these targets, which encode proteins with diverse roles, including extracellular signaling molecules, intracellular enzymes, transcription factors, and cytoskeletal proteins. A large number of the risk variants correlate with epigenetic factors, in particular cartilage DNA methylation changes in cis, implying that epigenetics may be a conduit through which genetic effects on gene expression are mediated. Some of the variants also appear to have been selected as humans adapted to bipedalism, suggesting that a proportion of osteoarthritis genetic susceptibility results from antagonistic pleiotropy, with risk variants having a positive role in joint formation but a negative role in the long-term health of the joint. Although data from an osteoarthritis genetic study has not yet directly led to a novel treatment, some of the osteoarthritis associated genes code for proteins that have available therapeutics. Genetic investigations are therefore revealing fascinating fundamental insights into osteoarthritis and can expose options for translational intervention.
Collapse
Affiliation(s)
- G Aubourg
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - S J Rice
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - P Bruce-Wootton
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - J Loughlin
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
27
|
Zhong Y, Xu Y, Xue S, Zhu L, Lu H, Wang C, Chen H, Sang W, Ma J. Nangibotide attenuates osteoarthritis by inhibiting osteoblast apoptosis and TGF-β activity in subchondral bone. Inflammopharmacology 2022; 30:1107-1117. [PMID: 35391646 DOI: 10.1007/s10787-022-00984-2] [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: 02/26/2022] [Accepted: 03/25/2022] [Indexed: 11/26/2022]
Abstract
Osteoarthritis (OA) is a chronic joint disorder that causes cartilage degradation and subchondral bone abnormalities. Nangibotide, also known as LR12, is a dodecapeptide with considerable anti-inflammatory properties, but its significance in OA is uncertain. The aim of the study was to determine whether nangibotide could attenuate the progression of OA, and elucidate the underlying mechanism. In vitro experiments showed that nangibotide strongly inhibited TNF-α-induced osteogenic reduction, significantly enhanced osteoblast proliferation and prevented apoptosis in MC3T3-E1 cells. Male C57BL/6 J mice aged 2 months were randomly allocated to three groups: sham, ACLT, and ACLT with nangibotide therapy. Nangibotide suppressed ACLT-induced cartilage degradation and MMP-13 expression. MicroCT analysis revealed that nangibotide attenuated in vivo subchondral bone loss induced by ACLT. Histomorphometry results showed that nangibotide attenuated ACLT-induced osteoblast inhibition; TUNEL assays and immunohistochemical staining of cleaved-caspase3 further confirmed the in vivo anti-apoptotic effect of nangibotide on osteoblasts. Furthermore, we found that nangibotide exerted protective effects by suppressing TGF-β signaling mediated by Smad2/3 to restore coupled bone remodeling in the subchondral bone. In conclusion, the findings suggest that nangibotide might exert a protective effect on the bone-cartilage unit and maybe an alternative treatment option for OA.
Collapse
Affiliation(s)
- Yiming Zhong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Yiming Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Song Xue
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Libo Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Haiming Lu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Cong Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Hongjie Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Weilin Sang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China.
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China.
| |
Collapse
|
28
|
Xian Bo S, Chen W, Chang L, Hao Ran Y, Hui Hui G, Ya Kun Z, Wu Kun X, Hai Tao F, Wen Dan C. The Research Progress of Exosomes in Osteoarthritis, With Particular Emphasis on the Therapeutic Effect. Front Pharmacol 2022; 13:731756. [PMID: 35308214 PMCID: PMC8924513 DOI: 10.3389/fphar.2022.731756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Exosomes participate in many physiological and pathological processes by regulating cell-to-cell communication. This affects the etiology and development of diseases, such as osteoarthritis (OA). Although exosomes in the OA tissue microenvironment are involved in the progression of OA, exosomes derived from therapeutic cells represent a new therapeutic strategy for OA treatment. Recent studies have shown that exosomes participate in OA treatment by regulating the proliferation, apoptosis, inflammation, and extracellular matrix synthesis of chondrocytes. However, studies in this field are scant. This review summarizes the therapeutic properties of exosomes on chondrocytes in OA and their underlying molecular mechanisms. We also discuss the challenges and prospects of exosome-based OA treatment.
Collapse
Affiliation(s)
- Shang Xian Bo
- Anhui Medical University, Hefei, China.,Second Hospital of Anhui Medical University, Hefei, China
| | - Wang Chen
- Anhui Medical University, Hefei, China.,Second Hospital of Anhui Medical University, Hefei, China
| | - Liu Chang
- Armed Police Corps Hospital of Anhui Province, Hefei, China
| | - Yu Hao Ran
- Second Hospital of Anhui Medical University, Hefei, China
| | - Guo Hui Hui
- Anhui Medical University, Hefei, China.,Second Hospital of Anhui Medical University, Hefei, China
| | - Zhu Ya Kun
- Fuyang Hospital of Anhui Medical University, Anhui, China
| | - Xie Wu Kun
- Second Hospital of Anhui Medical University, Hefei, China
| | - Fan Hai Tao
- Fuyang Hospital of Anhui Medical University, Anhui, China
| | - Cheng Wen Dan
- Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
29
|
Loughlin J. Translating osteoarthritis genetics research: challenging times ahead. Trends Mol Med 2022; 28:176-182. [PMID: 35033441 DOI: 10.1016/j.molmed.2021.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022]
Abstract
The ultimate goal of molecular genetic studies of human diseases is to translate the discoveries for patient benefit. For diseases that lack licensed disease-modifying therapeutics, such as osteoarthritis (OA), the need is acute. OA is polygenic and affects older individuals, with a recent genome-wide study of over 800 000 individuals adding 52 novel association signals to those already reported on for this common arthritis. Many of the predicted effector genes of these signals encode proteins that are targets of drugs for other indications, highlighting repurposing opportunities. Here, the potential for OA genetic data to translate is discussed, including whether the developmental origin of OA will limit the application of genetic risk data for disease-modification purposes.
Collapse
Affiliation(s)
- John Loughlin
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK.
| |
Collapse
|
30
|
Young DA, Barter MJ, Soul J. Osteoarthritis year in review: genetics, genomics, epigenetics. Osteoarthritis Cartilage 2022; 30:216-225. [PMID: 34774787 PMCID: PMC8811265 DOI: 10.1016/j.joca.2021.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In this review, we have highlighted the advances over the past year in genetics, genomics and epigenetics in the field of osteoarthritis (OA). METHODS A literature search of PubMed was performed using the criteria: "osteoarthritis" and one of the following terms "genetic(s), genomic(s), epigenetic(s), polymorphism, noncoding ribonucleic acid (RNA), microRNA, long noncoding RNA, lncRNA, circular RNA, RNA sequencing (RNA-seq), single cell sequencing, transcriptomics, or deoxyribonucleic acid (DNA) methylation between April 01, 2020 and April 30, 2021. RESULTS In total we identified 765 unique publications, which eventually reduced to 380 of relevance to the field as judged by two assessors. Many of these studies included multiple search terms. We summarised advances relating to genetics, functional genetics, genomics and epigenetics, focusing on our personal key papers during the year. CONCLUSIONS This year few studies have identified new genetic variants contributing to OA susceptibility, but a focus has been on refining risk loci or their functional validation. The use of new technologies together with investigating the cross-talk between multiple tissue types, greater sample sizes and/or better patient classification (OA subtypes) will continue to increase our knowledge of disease mechanisms and progress towards understanding and treating OA.
Collapse
Affiliation(s)
- D A Young
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK.
| | - M J Barter
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - J Soul
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
31
|
Tan Q, Cai Z, Li J, Li J, Xiang H, Li B, Cai G. Imaging Study on Acupuncture Inhibiting Inflammation and Bone Destruction in Knee Osteoarthritis Induced by Monosodium Iodoacetate in Rat Model. J Pain Res 2022; 15:93-103. [PMID: 35046719 PMCID: PMC8760981 DOI: 10.2147/jpr.s346242] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/18/2021] [Indexed: 11/26/2022] Open
Abstract
Objective We aim to explore whether acupuncture inhibits inflammation and bone destruction in rat model monosodium iodoacetate (MIA)-induced knee osteoarthritis (KOA) by 18F-fluorodeoxyglucose (18F-FDG) small-animal positron emission tomography (PET) and micro-computed tomography (CT) imaging. Methods KOA was induced in rats by intra-articular injection MIA (2 mg/50 μL) through the right knee of the rats. Forty male Sprague Dawley rats weighing 280 to 340 g (12 weeks old) were randomly divided into four groups including Control group, KOA group, KOA plus manual acupuncture group (KOA+MA), KOA plus sham acupuncture group (KOA+SA). The acupuncture treatment lasted for three weeks (one-day rest after six days of treatment). Paw withdrawal threshold test and open-field test were used to assess mechanical allodynia and locomotor activity respectively for once a week. Hematoxylin and eosin (H&E) staining was used to assess the damage of the cartilage, synovium and infrapatellar fat pad (IFP). 18F-FDG PET was performed to quantify joint inflammation. The influence on the subchondral bone in these rats was confirmed by micro-CT. Results Mechanical hyperalgesia, joint inflammation, and obvious bone destruction were observed in the KOA group. H&E staining of the knee joint found that manual acupuncture played a protective effect in cartilage, synovium and IFP destruction. However, compared with KOA group, the results in sham acupuncture had no significant difference. After manual acupuncture treatment in KOA rats, inflammation was significantly suppressed shown by 18F-FDG PET imaging. Micro-CT analysis of the knee joint revealed that manual acupuncture protected bone by inhibiting osteophyte development and subchondral bone remodeling. Conclusion The results of 18F-FDG PET and micro-CT showed that manual acupuncture inhibited inflammation and bone destruction, which provides reliable evidence for the effectiveness of acupuncture in hindering development of KOA, and provides reliable evidence for clinical application of acupuncture.
Collapse
Affiliation(s)
- Qian Tan
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhengkun Cai
- Department of Public Administration, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jia Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Jing Li
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Hongchun Xiang
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bocun Li
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Guowei Cai
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Correspondence: Guowei Cai Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China Email
| |
Collapse
|
32
|
Li M, Li H, Ran X, Yin H, Luo X, Chen Z. Effects of adenovirus-mediated knockdown of IRAK4 on synovitis in the osteoarthritis rabbit model. Arthritis Res Ther 2021; 23:294. [PMID: 34863246 PMCID: PMC8643028 DOI: 10.1186/s13075-021-02684-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/20/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The use of interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor as a treatment for the inflammatory joint disease is a promising method. However, its underlying mechanism in osteoarthritis (OA) remains unclear. The purpose of this study is to look into the effects of adenovirus-mediated knockdown of IRAK4 on synovitis in the OA rabbit model. METHODS Ad-shIRAK4 was injected two weeks after anterior cruciate ligament resection. Six weeks later, the rabbits were killed. The expression of IRAK4, TNFR-associated factor 6(TRAF6), TGF-activated kinase 1(TAK1), p-IKB kinase (p-IKK), p-nuclear factor kappa-B (p-NFκB), p38, and p-p38 in the synovial membrane was detected by western blot, qRT-PCR, and immunohistochemistry analysis. Immunohistochemistry was to detect the expression of IRAK4 proteins in articular cartilage. H&E staining was to assess the pathological changes of synovium and cartilage. The levels of interleukin (IL)-1β, tumor necrosis factor-α(TNF-α), and MMP-13 in the synovial fluid were measured by ELISA. X-ray and micro-computerized tomography (μCT) scans were used to assess knee joint conditions and microstructure of subchondral bone. RESULTS IRAK4 expression levels in synovial tissues of the OA model group exhibited a significant upward trend. Ad-shIRAK4 significantly reduced IRAK4 mRNA expression in synovium tissues. Notably, Ad-shIRAK4 suppressed the Toll-like receptor/interleukin-1 receptor (TLR/IL-1R) signaling. In addition, in the Ad-shIRAK4 treatment group, we can see less inflammatory cell infiltration and reduced hyperplasia and angiogenesis. The levels of IL-1β, TNF-α, and MMP-13 in the synovial fluid in the OA model group were significantly higher than that in the control group, which were reduced by Ad-shIRAK4 treatment. Finally, Results of HE stains, immunohistochemistry, and μCT showed that Ad-shIRAK4 treatment has a protective effect on cartilage damage. CONCLUSIONS IRAK4 is significantly upregulated in the synovium from the osteoarthritis rabbit model. In addition, Ad-shIRAK4 reduced the expression of IRAK4 and suppressed TLR/IL-1R signaling in the synovium from the osteoarthritis rabbit model. Ad-shIRAK4 could alleviate synovitis and cartilage degradation in the osteoarthritis rabbit model, and thus alleviate the symptoms of OA and prevent the progression of OA.
Collapse
Affiliation(s)
- Muzhe Li
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Huiyun Li
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Xun Ran
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Han Yin
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xuling Luo
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China.
| | - Zhiwei Chen
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China.
| |
Collapse
|
33
|
Abstract
This review describes results obtained with tissue from prior studies of equine and human osteoarthritis (OA). The main methods considered are scanning electron microscopy, novel methods in light microscopy and X-ray Micro-tomography. The same samples have been re-utilised in several ways. The tissues described are hyaline articular cartilage (HAC; or substitutes), with its deep layer, articular calcified cartilage (ACC), whose deep surface is resorbed in cutting cone events to allow the deposition of subchondral bone (SCB). Multiple tidemarks are normal. Turnover at the osteochondral (ACC-HAC-SCB) junction is downregulated by overload exercise, conversely, during rest periods. Consequent lack of support predisposes to microfracture of the ACC-SCB plate, in the resorption-related repair phase of which the plate is further undermined to form sink holes. The following characteristics contribute to the OA scenario: penetrating resorption canals and local loss of ACC; cracking of ACC and SCB; sealing of cracks with High-Density Mineral Infill (HDMI); extrusion of HDMI into HAC to form High-Density Mineral Protrusions (HDMP) in HAC which may fragment and contribute to its destruction; SCB marrow space infilling and densification with (at first) woven bone; disruption, fibrillation and loss of HAC; eburnation; repair with abnormal tissues including fibrocartilage and woven bone; attachment of Sharpey fibres to SCB trabeculae and adipocyte-moulded extensions to trabeculae (excrescences).
Collapse
Affiliation(s)
- Alan Boyde
- Dental Physical Sciences Imaging Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Mile End Campus, London, E1 4NS, UK.
| |
Collapse
|
34
|
Destouni A, Tsolis KC, Economou A, Papathanasiou I, Balis C, Mourmoura E, Tsezou A. Chondrocyte protein co-synthesis network analysis links ECM mechanosensing to metabolic adaptation in osteoarthritis. Expert Rev Proteomics 2021; 18:623-635. [PMID: 34348542 DOI: 10.1080/14789450.2021.1962299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Knee osteoarthritis (OA) is one of the most common structural OA disorders globally. Incomplete understanding of the fundamental biological aspects of osteoarthritis underlies the current lack of effective treatment or disease modifying drugs. RESEARCH DESIGN AND METHODS We implemented a systems approach by making use of the statistical network concepts in Weighted Gene Co-expression Analysis to reconstruct the organization of the core proteome network in chondrocytes obtained from OA patients and healthy individuals. Protein modules reflect groups of tightly co-ordinated changes in protein abundance across healthy and OA chondrocytes. RESULTS The unbiased systems analysis identified extracellular matrix (ECM) mechanosensing and glycolysis as two modules that are most highly correlated with ΟΑ. The ECM module was enriched in the OA genetic risk factors tenascin-C (TNC) and collagen 11A1 (COL11A1), as well as in cartilage oligomeric matrix protein (COMP), a biomarker associated with cartilage integrity. Mapping proteins that are unique to OA or healthy chondrocytes onto the core interactome, which connects microenvironment sensing and regulation of glycolysis, identified differences in metabolic and anti-inflammatory adaptation. CONCLUSION The interconnection between cartilage ECM remodeling and metabolism is indicative of the dynamic chondrocyte states and their significance in osteoarthritis.
Collapse
Affiliation(s)
- Aspasia Destouni
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Konstantinos C Tsolis
- KULeuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, Leuven, Belgium
| | - Anastassios Economou
- KULeuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, Leuven, Belgium
| | - Ioanna Papathanasiou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece.,Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Charalampos Balis
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Evanthia Mourmoura
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece.,Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| |
Collapse
|
35
|
Control of the Autophagy Pathway in Osteoarthritis: Key Regulators, Therapeutic Targets and Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms22052700. [PMID: 33800062 PMCID: PMC7962119 DOI: 10.3390/ijms22052700] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/17/2022] Open
Abstract
Autophagy is involved in different degenerative diseases and it may control epigenetic modifications, metabolic processes, stem cells differentiation as well as apoptosis. Autophagy plays a key role in maintaining the homeostasis of cartilage, the tissue produced by chondrocytes; its impairment has been associated to cartilage dysfunctions such as osteoarthritis (OA). Due to their location in a reduced oxygen context, both differentiating and mature chondrocytes are at risk of premature apoptosis, which can be prevented by autophagy. AutophagomiRNAs, which regulate the autophagic process, have been found differentially expressed in OA. AutophagomiRNAs, as well as other regulatory molecules, may also be useful as therapeutic targets. In this review, we describe and discuss the role of autophagy in OA, focusing mainly on the control of autophagomiRNAs in OA pathogenesis and their potential therapeutic applications.
Collapse
|