1
|
Xu Y, Ye Z, Wang Y, Ma Y, Chen X, Wang S, Zhang B, Xia C. Alleviating osteoarthritis-induced damage through extracellular vesicles derived from inflammatory chondrocytes. Int Immunopharmacol 2025; 146:113829. [PMID: 39675196 DOI: 10.1016/j.intimp.2024.113829] [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: 05/22/2024] [Revised: 11/19/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
The role of extracellular vesicles (EVs) derived from inflammatory chondrocytes in EV-based therapy for osteoarthritis (OA) has received little attention. We examined the effects of EVs derived from both normal rat chondrocytes (nEVs) and IL-1β-treated rat chondrocytes (iEVs) on IL-1β-treated rat chondrocytes, macrophages, and osteoblasts, alongside mRNA-seq and miRNA-seq analyses of both them. Additionally, nEVs and iEVs were administered intra-articularly in the joints of rat models subjected to anterior cruciate ligament transection (ACLT), and the morphological alterations across the joints were assessed. These findings indicated that iEVs, compared with nEVs, significantly enhanced collagen II synthesis in IL-1β-treated chondrocytes, accompanied by marked increases in ER stress and autophagy. In comparison to nEVs, iEVs exhibited a greater effect on facilitating M2-type macrophage polarization while simultaneously diminishing M1-type polarization, a process likely mediated by the downregulation of chemotactic cytokines such as Cxcl10, Ccl5, Cxcl9, Cxcl1, and Cxcl11. iEVs exerted a more pronounced influence on the phenotypic characteristics of IL-1β-treated osteoblasts than nEVs. In the ACLT-rat model, iEVs, akin to nEVs, effectively mitigated articular cartilage degradation. However, there was no significant difference in OARSI Scores between the two groups, despite iEVs exerting a greater effect on increasing hyaline cartilage thickness and proteoglycan content. iEVs were superior to nEVs in attenuating synovium inflammation and promoting trabecula formation in the femur subchondral bone. Consequently, iEVs, akin to nEVs, significantly alleviated OA-induced damage. Moreover, iEVs outperformed nEVs in certain aspects, notably in augmenting hyaline cartilage, reducing synovium inflammation, and promoting trabecular formation in the subchondral bone during the early stage of OA.
Collapse
Affiliation(s)
- Yang Xu
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Zesen Ye
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Yue Wang
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Yongkang Ma
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Xiaolei Chen
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Shaojie Wang
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China.
| | - Bing Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Chun Xia
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China.
| |
Collapse
|
2
|
Salehi S, Brambilla S, Rasponi M, Lopa S, Moretti M. Development of a Microfluidic Vascularized Osteochondral Model as a Drug Testing Platform for Osteoarthritis. Adv Healthc Mater 2024; 13:e2402350. [PMID: 39370575 DOI: 10.1002/adhm.202402350] [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: 06/28/2024] [Revised: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by changes in cartilage and subchondral bone. To date, there are no available drugs that can counteract the progression of OA, partly due to the inadequacy of current models to recapitulate the relevant cellular complexity. In this study, an osteochondral microfluidic model is developed using human primary cells to mimic an OA-like microenvironment and this study validates it as a drug testing platform. In the model, the cartilage compartment is created by embedding articular chondrocytes in fibrin hydrogel while the bone compartment is obtained by embedding osteoblasts, osteoclasts, endothelial cells, and mesenchymal stem cells in a fibrin hydrogel enriched with calcium phosphate nanoparticles. After developing and characterizing the model, Interleukin-1β is applied to induce OA-like conditions. Subsequently, the model potential is evaluated as a drug testing platform by assessing the effect of two anti-inflammatory drugs (Interleukin-1 Receptor antagonist and Celecoxib) on the regulation of inflammation- and matrix degradation-related markers. The model responded to inflammation and demonstrated differences in drug efficacy. Finally, it compares the behavior of the "Cartilage" and "Cartilage+Bone" models, emphasizing the necessity of incorporating both cartilage and bone compartments to capture the complex pathophysiology of OA.
Collapse
Affiliation(s)
- Shima Salehi
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Stefania Brambilla
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Via Chiesa 5, Bellinzona, 6500, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Via Tesserete 46, Lugano, 6900, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Via Buffi 13, Lugano, 6900, Switzerland
| |
Collapse
|
3
|
Liu X, Chen R, Cui G, Feng R, Liu K. Exosomes derived from platelet-rich plasma present a novel potential in repairing knee articular cartilage defect combined with cyclic peptide-modified β-TCP scaffold. J Orthop Surg Res 2024; 19:718. [PMID: 39497084 PMCID: PMC11533314 DOI: 10.1186/s13018-024-05202-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND The aim of this study was to investigate the therapeutic effects and mechanisms of PRP-exos combined with cyclic peptide-modified β-TCP scaffold in the treatment of rabbit knee cartilage defect. METHODS PRP-exos were extracted and characterized by TEM, NTA and WB. The therapeutic effects were evaluated by ICRS score, HE staining, Immunohistochemistry, qRT-PCR and ELISA. The repair mechanism of PRP-exos was estimated and predicted by miRNA sequencing analysis and protein-protein interaction network analysis. RESULTS The results showed that PRP-exos had a reasonable size distribution and exhibited typical exosome morphology. The combination of PRP-exos and cyclic peptide-modified β-TCP scaffold improved ICRS score and the expression level of COL-2, RUNX2, and SOX9. Moreover, this combination therapy reduced the level of MMP-3, TNF-α, IL-1β, and IL-6, while increasing the level of TIMP-1. In PRP-exos miRNA sequencing analysis, the total number of known miRNAs aligned across all samples was 252, and a total of 91 differentially expressed miRNAs were detected. The results of KEGG enrichment analysis and the protein-protein interaction network analysis indicated that the PI3K/AKT signaling pathway could impact the function of chondrocytes by regulating key transcription factors to repair cartilage defect. CONCLUSION PRP-exos combined with cyclic peptide-modified β-TCP scaffold effectively promoted cartilage repair and improved chondrocyte function in rabbit knee cartilage defect. Based on the analysis and prediction of PRP-exos miRNAs sequencing, PI3K/AKT signaling pathway may contribute to the therapeutic effect. These findings provide experimental evidence for the application of PRP-exos in the treatment of cartilage defect.
Collapse
Affiliation(s)
- Xuchang Liu
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan, 250103, Shandong, China
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan, 250103, Shandong, China
| | - Rudong Chen
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Guanzheng Cui
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Rongjie Feng
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan, 250103, Shandong, China.
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Jinan, 250103, Shandong, China.
| |
Collapse
|
4
|
Anker-Hansen C, Pirouzifard M, Memon A, Sundquist J, Sundquist K, Zöller B. Mitochondria-DNA copy-number in osteoporosis and osteoarthritis among middle-aged women - A population-based cohort study. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100501. [PMID: 39101051 PMCID: PMC11295846 DOI: 10.1016/j.ocarto.2024.100501] [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: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024] Open
Abstract
Background Mitochondrial DNA copy number (mtDNA-CN) is associated with aging. A relationship between mtDNA-CN and degenerative disorders, e.g. osteoarthritis (OA) and osteoporosis (OP), has been suggested. We aimed to investigate the relationship of mtDNA-CN and incident OA and OP. Materials and methods MtDNA-CN was studied in relationship to incident OA and OP in a population-based cohort study of 6916 middle-aged women (52-63 years). Totally 2521 women with sufficient quality of mtDNA were analyzed. After exclusions, 1978 women remained in the study population. Four different endpoints obtained from the National Patient register were studied: 1) OA, 2) OP 3) OA surgery, and 4) OP fracture. In the multivariate model adjustments were made for potential OA and OP risk factors. Results Women with low mtDNA-CN were older and had more activity at work. 125 women (6.32%) were affected by incident OP and 254 women (12.84%) had an OP fracture. Incident OA affected 451 women (22.80%) and 175 women (8.85%) had OA surgery. There were no associations between mtDNA-CN and incident risk of OA (Hazard ratio = 1.00, 95% confidence interval 0.83-1.20), OA surgery (0.79, 0.58-1.07), OP (0.89, 0.62-1.27), or OP fracture (1.00, 0.78-1.29). However, incident OP was significantly associated with T-score (bone density), smoking, diabetes mellitus, and chronic obstructive bronchitis (COPD). OA was associated with body mass index and COPD. Conclusions The present study suggests that mtDNA-CN, reflecting mitochondrial dysfunction, is not a major predictor for incident OA or OP. However, due to the limited study size minor associations cannot be excluded.
Collapse
Affiliation(s)
| | - MirNabi Pirouzifard
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Ashfaque Memon
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Bengt Zöller
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| |
Collapse
|
5
|
Meng F, Zhu P, Ren X, Wang L, Ding D, Yan J, Zhang Y, Yang SY, Ning B. Cardamonin inhibits osteogenic differentiation by downregulating Wnt/beta-catenin signaling and alleviates subchondral osteosclerosis in osteoarthritic mice. J Orthop Res 2024; 42:1933-1942. [PMID: 38520666 DOI: 10.1002/jor.25842] [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: 12/18/2023] [Revised: 02/27/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024]
Abstract
Osteoarthritis (OA) is a common degenerative joint disease, and subchondral osteosclerosis is an important pathological change that occurs in its late stages. Cardamonin (CD) is a natural flavonoid isolated from Alpinia katsumadai that has anti-inflammatory activity. The objectives of this study were to investigate the therapeutic effects and potential mechanism of CD in regulating OA subchondral osteosclerosis at in vivo and in vitro settings. Eight-week-old male C57BL/6J mice were randomly divided into four groups: sham operation, anterior cruciate ligament transection (ACLT)-induced OA model, low-dose and high-dose CD treated ACLT-OA model groups. Histological assessment and immunohistochemical examinations for chondrocyte metabolism-related markers metalloproteinase-13, ADAMTS-4, Col II, and Sox-9 were performed. Microcomputed tomography was used to assess the sclerosis indicators in subchondral bone. Further, MC3T3-E1 (a mouse calvarial preosteoblast cell line) cells were treated with various concentrations of CD to reveal the influence and potential molecular pathways of CD in osteogenic differentiations. Animal studies suggested that CD alleviated the pathological changes in OA mice such as maintaining integrity and increasing the thickness of hyaline cartilage, decreasing the thickness of calcified cartilage, decreasing the Osteoarthritis Research Society International score, regulating articular cartilage metabolism, and inhibiting subchondral osteosclerosis. In vitro investigation indicated that CD inhibited alkaline phosphatase expression and production of calcium nodules during osteogenic differentiation of MC3T3-E1 cells. In addition, CD inhibited the expression of osteogenic differentiation-related indicators and Wnt/β-catenin pathway-related proteins. In conclusion, CD inhibits osteogenic differentiation by downregulating Wnt/β-catenin signaling and alleviating subchondral osteosclerosis in a mouse model of OA.
Collapse
Affiliation(s)
- Fanding Meng
- Department of Hand and Foot Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Pengchong Zhu
- Department of Orthopedic Surgery, Jinan Central Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoli Ren
- Department of Orthopedic Surgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Limei Wang
- Faculty of Preclinical Medicine, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Dong Ding
- Department of Orthopedic Surgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Jiangbo Yan
- The 3rd Orthopedic Ward, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ying Zhang
- Department of Orthopedic Surgery, Jinan Central Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shang-You Yang
- Department of Orthopaedic Surgery, University of Kansas School of Medicine Wichita, Wichita, Kansas, USA
| | - Bin Ning
- Department of Orthopedic Surgery, Jinan Central Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
6
|
Epanomeritakis IE, Khan WS. Adipose-derived regenerative therapies for the treatment of knee osteoarthritis. World J Stem Cells 2024; 16:324-333. [PMID: 38690511 PMCID: PMC11056639 DOI: 10.4252/wjsc.v16.i4.324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 04/25/2024] Open
Abstract
Knee osteoarthritis is a degenerative condition with a significant disease burden and no disease-modifying therapy. Definitive treatment ultimately requires joint replacement. Therapies capable of regenerating cartilage could significantly reduce financial and clinical costs. The regenerative potential of mesenchymal stromal cells (MSCs) has been extensively studied in the context of knee osteoarthritis. This has yielded promising results in human studies, and is likely a product of immunomodulatory and chondroprotective biomolecules produced by MSCs in response to inflammation. Adipose-derived MSCs (ASCs) are becoming increasingly popular owing to their relative ease of isolation and high proliferative capacity. Stromal vascular fraction (SVF) and micro-fragmented adipose tissue (MFAT) are produced by the enzymatic and mechanical disruption of adipose tissue, respectively. This avoids expansion of isolated ASCs ex vivo and their composition of heterogeneous cell populations, including immune cells, may potentiate the reparative function of ASCs. In this editorial, we comment on a multicenter randomized trial regarding the efficacy of MFAT in treating knee osteoarthritis. We discuss the study's findings in the context of emerging evidence regarding adipose-derived regenerative therapies. An underlying mechanism of action of ASCs is proposed while drawing important distinctions between the properties of isolated ASCs, SVF, and MFAT.
Collapse
Affiliation(s)
- Ilias E Epanomeritakis
- Division of Trauma and Orthopaedic Surgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Wasim S Khan
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, United Kingdom.
| |
Collapse
|
7
|
Ho TJ, Tsai WT, Wu JR, Chen HP. Biological Activities of Deer Antler-Derived Peptides on Human Chondrocyte and Bone Metabolism. Pharmaceuticals (Basel) 2024; 17:434. [PMID: 38675396 PMCID: PMC11053545 DOI: 10.3390/ph17040434] [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: 02/01/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Orally administered "tortoiseshell and deer antler gelatin" is a common traditional medicine for patients with osteoporosis or osteoarthritis. From the pepsin-digested gelatin, we previously isolated and identified the osteoblast-stimulating pentapeptide, TSKYR. Its trypsin digestion products include the dipeptide YR, enhancing calcium ion uptake, and tripeptide TSK, resulting in remarkable 30- and 50-fold increases in mineralized nodule area and density in human osteoblast cells. These peptides were chemically synthesized in this study. The composition of deer antler preparations comprises not only proteins and peptides but also a significant quantity of metal ion salts. By analyzing osteoblast growth in the presence of peptide YR and various metal ions, we observed a synergistic effect of calcium and strontium on the effects of YR. Those peptides could also stimulate the growth of C2C12 skeletal muscle cells and human chondrocytes, increasing collagen and glycosaminoglycan content in a three-dimensional environment. The maintenance of bone homeostasis relies on a balance between osteoclasts and osteoblasts. Deer antler peptides were observed to inhibit osteoclast differentiation, as evidenced by ROS generation, tartrate-resistant acid phosphatase (TRACP) activity assays, and gene expression in RAW264.7 cells. In summary, our findings provide a deep understanding of the efficacy of this folk medicine.
Collapse
Affiliation(s)
- Tsung-Jung Ho
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan; (T.-J.H.); (W.-T.T.)
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970473, Taiwan
| | - Wan-Ting Tsai
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan; (T.-J.H.); (W.-T.T.)
| | - Jia-Ru Wu
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan; (T.-J.H.); (W.-T.T.)
| | - Hao-Ping Chen
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan; (T.-J.H.); (W.-T.T.)
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| |
Collapse
|
8
|
Yang D, Xu K, Xu X, Xu P. Revisiting prostaglandin E2: A promising therapeutic target for osteoarthritis. Clin Immunol 2024; 260:109904. [PMID: 38262526 DOI: 10.1016/j.clim.2024.109904] [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: 12/01/2023] [Revised: 01/08/2024] [Accepted: 01/14/2024] [Indexed: 01/25/2024]
Abstract
Osteoarthritis (OA) is a complex disease characterized by cartilage degeneration and persistent pain. Prostaglandin E2 (PGE2) plays a significant role in OA inflammation and pain. Recent studies have revealed the significant role of PGE2-mediated skeletal interoception in the progression of OA, providing new insights into the pathogenesis and treatment of OA. This aspect also deserves special attention in this review. Additionally, PGE2 is directly involved in pathologic processes including aberrant subchondral bone remodeling, cartilage degeneration, and synovial inflammation. Therefore, celecoxib, a commonly used drug to alleviate inflammatory pain through inhibiting PGE2, serves not only as an analgesic for OA but also as a potential disease-modifying drug. This review provides a comprehensive overview of the discovery history, synthesis and release pathways, and common physiological roles of PGE2. We discuss the roles of PGE2 and celecoxib in OA and pain from skeletal interoception and multiple perspectives. The purpose of this review is to highlight PGE2-mediated skeletal interoception and refresh our understanding of celecoxib in the pathogenesis and treatment of OA.
Collapse
Affiliation(s)
- Dinglong Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Ke Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Xin Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China.
| |
Collapse
|
9
|
Costa FR, Santos MDS, Martins RA, Costa CB, Hamdan PC, Da Silva MB, Azzini GOM, Pires L, Menegassi Z, Santos GS, Lana JF. The Synergistic Effects of Hyaluronic Acid and Platelet-Rich Plasma for Patellar Chondropathy. Biomedicines 2023; 12:6. [PMID: 38275367 PMCID: PMC10813186 DOI: 10.3390/biomedicines12010006] [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: 11/01/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/27/2024] Open
Abstract
Musculoskeletal disorders are increasingly prevalent worldwide, causing significant socioeconomic burdens and diminished quality of life. Notably, patellar chondropathy (PC) is among the most widespread conditions affecting joint structures, resulting in profound pain and disability. Hyaluronic acid (HA) and platelet-rich plasma (PRP) have emerged as reliable, effective, and minimally invasive alternatives. Continuous research spanning from laboratory settings to clinical applications demonstrates the numerous advantages of both products. These encompass lubrication, anti-inflammation, and stimulation of cellular behaviors linked to proliferation, differentiation, migration, and the release of essential growth factors. Cumulatively, these benefits support the rejuvenation of bone and cartilaginous tissues, which are otherwise compromised due to the prevailing degenerative and inflammatory responses characteristic of tissue damage. While existing literature delves into the physical, mechanical, and biological facets of these products, as well as their commercial variants and distinct clinical uses, there is limited discussion on their interconnected roles. We explore basic science concepts, product variations, and clinical strategies. This comprehensive examination provides physicians with an alternative insight into the pathophysiology of PC as well as biological mechanisms stimulated by both HA and PRP that contribute to tissue restoration.
Collapse
Affiliation(s)
- Fábio Ramos Costa
- Department of Orthopedics, FC Sports Traumatology Clinic, Salvador 40296-210, Brazil; (F.R.C.); (C.B.C.)
| | | | | | - Cláudia Bruno Costa
- Department of Orthopedics, FC Sports Traumatology Clinic, Salvador 40296-210, Brazil; (F.R.C.); (C.B.C.)
| | - Paulo César Hamdan
- Department of Orthopedics, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-630, Brazil; (P.C.H.); (M.B.D.S.); (Z.M.)
| | - Marcos Britto Da Silva
- Department of Orthopedics, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-630, Brazil; (P.C.H.); (M.B.D.S.); (Z.M.)
| | - Gabriel Ohana Marques Azzini
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (G.O.M.A.); (L.P.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil
| | - Luyddy Pires
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (G.O.M.A.); (L.P.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil
| | - Zartur Menegassi
- Department of Orthopedics, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-630, Brazil; (P.C.H.); (M.B.D.S.); (Z.M.)
| | - Gabriel Silva Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (G.O.M.A.); (L.P.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil
| | - José Fábio Lana
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, Brazil; (G.O.M.A.); (L.P.); (J.F.L.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, Brazil
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, Brazil
- Clinical Research, Anna Vitória Lana Institute (IAVL), Indaiatuba 13334-170, Brazil
| |
Collapse
|
10
|
Riggs KC, Sankar U. Inflammatory mechanisms in post-traumatic osteoarthritis: a role for CaMKK2. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00031. [PMID: 37849987 PMCID: PMC10578519 DOI: 10.1097/in9.0000000000000031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/23/2023] [Indexed: 10/19/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) is a multifactorial disease of the cartilage, synovium, and subchondral bone resulting from direct joint trauma and altered joint mechanics after traumatic injury. There are no current disease-modifying therapies for PTOA, and early surgical interventions focused on stabilizing the joint do not halt disease progression. Chronic pain and functional disability negatively affect the quality of life and take an economic toll on affected patients. While multiple mechanisms are at play in disease progression, joint inflammation is a key contributor. Impact-induced mitochondrial dysfunction and cell death or altered joint mechanics after trauma culminate in inflammatory cytokine release from synoviocytes and chondrocytes, cartilage catabolism, suppression of cartilage anabolism, synovitis, and subchondral bone disease, highlighting the complexity of the disease. Current understanding of the cellular and molecular mechanisms underlying the disease pathology has allowed for the investigation of a variety of therapeutic strategies that target unique apoptotic and/or inflammatory processes in the joint. This review provides a concise overview of the inflammatory and apoptotic mechanisms underlying PTOA pathogenesis and identifies potential therapeutic targets to mitigate disease progression. We highlight Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2), a serine/threonine protein kinase that was recently identified to play a role in murine and human osteoarthritis pathogenesis by coordinating chondrocyte inflammatory responses and apoptosis. Given its additional effects in regulating macrophage inflammatory signaling and bone remodeling, CaMKK2 emerges as a promising disease-modifying therapeutic target against PTOA.
Collapse
Affiliation(s)
- Keegan C. Riggs
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Uma Sankar
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
11
|
Zhao T, Li X, Li H, Deng H, Li J, Yang Z, He S, Jiang S, Sui X, Guo Q, Liu S. Advancing drug delivery to articular cartilage: From single to multiple strategies. Acta Pharm Sin B 2023; 13:4127-4148. [PMID: 37799383 PMCID: PMC10547919 DOI: 10.1016/j.apsb.2022.11.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022] Open
Abstract
Articular cartilage (AC) injuries often lead to cartilage degeneration and may ultimately result in osteoarthritis (OA) due to the limited self-repair ability. To date, numerous intra-articular delivery systems carrying various therapeutic agents have been developed to improve therapeutic localization and retention, optimize controlled drug release profiles and target different pathological processes. Due to the complex and multifactorial characteristics of cartilage injury pathology and heterogeneity of the cartilage structure deposited within a dense matrix, delivery systems loaded with a single therapeutic agent are hindered from reaching multiple targets in a spatiotemporal matched manner and thus fail to mimic the natural processes of biosynthesis, compromising the goal of full cartilage regeneration. Emerging evidence highlights the importance of sequential delivery strategies targeting multiple pathological processes. In this review, we first summarize the current status and progress achieved in single-drug delivery strategies for the treatment of AC diseases. Subsequently, we focus mainly on advances in multiple drug delivery applications, including sequential release formulations targeting various pathological processes, synergistic targeting of the same pathological process, the spatial distribution in multiple tissues, and heterogeneous regeneration. We hope that this review will inspire the rational design of intra-articular drug delivery systems (DDSs) in the future.
Collapse
Affiliation(s)
- Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Haoyuan Deng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianwei Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhen Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuangpeng Jiang
- Department of Joint Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
12
|
Paz-González R, Lourido L, Calamia V, Fernández-Puente P, Quaranta P, Picchi F, Blanco FJ, Ruiz-Romero C. An Atlas of the Knee Joint Proteins and Their Role in Osteoarthritis Defined by Literature Mining. Mol Cell Proteomics 2023; 22:100606. [PMID: 37356495 PMCID: PMC10393810 DOI: 10.1016/j.mcpro.2023.100606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/14/2023] [Accepted: 06/18/2023] [Indexed: 06/27/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent rheumatic pathology. However, OA is not simply a process of wear and tear affecting articular cartilage but rather a disease of the entire joint. One of the most common locations of OA is the knee. Knee tissues have been studied using molecular strategies, generating a large amount of complex data. As one of the goals of the Rheumatic and Autoimmune Diseases initiative of the Human Proteome Project, we applied a text-mining strategy to publicly available literature to collect relevant information and generate a systematically organized overview of the proteins most closely related to the different knee components. To this end, the PubPular literature-mining software was employed to identify protein-topic relationships and extract the most frequently cited proteins associated with the different knee joint components and OA. The text-mining approach searched over eight million articles in PubMed up to November 2022. Proteins associated with the six most representative knee components (articular cartilage, subchondral bone, synovial membrane, synovial fluid, meniscus, and cruciate ligament) were retrieved and ranked by their relevance to the tissue and OA. Gene ontology analyses showed the biological functions of these proteins. This study provided a systematic and prioritized description of knee-component proteins most frequently cited as associated with OA. The study also explored the relationship of these proteins to OA and identified the processes most relevant to proper knee function and OA pathophysiology.
Collapse
Affiliation(s)
- Rocío Paz-González
- Grupo de Investigación de Reumatología (GIR) - Unidad de Proteómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Sergas, Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Lucía Lourido
- Grupo de Investigación de Reumatología (GIR) - Unidad de Proteómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Sergas, Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Valentina Calamia
- Grupo de Investigación de Reumatología (GIR) - Unidad de Proteómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Sergas, Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Patricia Fernández-Puente
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Grupo de Investigación de Reumatología y Salud (GIR-S), Centro Interdisciplinar de Química e Bioloxía (CICA), Universidade da Coruña (UDC), A Coruña, Spain
| | - Patricia Quaranta
- Grupo de Investigación de Reumatología (GIR) - Unidad de Proteómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Sergas, Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Florencia Picchi
- Grupo de Investigación de Reumatología (GIR) - Unidad de Proteómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Sergas, Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Francisco J Blanco
- Grupo de Investigación de Reumatología (GIR) - Unidad de Proteómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Sergas, Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain; Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Grupo de Investigación de Reumatología y Salud (GIR-S), Centro Interdisciplinar de Química e Bioloxía (CICA), Universidade da Coruña (UDC), A Coruña, Spain.
| | - Cristina Ruiz-Romero
- Grupo de Investigación de Reumatología (GIR) - Unidad de Proteómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Sergas, Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
13
|
Zhang S, Li T, Feng Y, Zhang K, Zou J, Weng X, Yuan Y, Zhang L. Exercise improves subchondral bone microenvironment through regulating bone-cartilage crosstalk. Front Endocrinol (Lausanne) 2023; 14:1159393. [PMID: 37288291 PMCID: PMC10242115 DOI: 10.3389/fendo.2023.1159393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Articular cartilage degeneration has been proved to cause a variety of joint diseases, among which osteoarthritis is the most typical. Osteoarthritis is characterized by articular cartilage degeneration and persistent pain, which affects the quality of life of patients as well as brings a heavy burden to society. The occurrence and development of osteoarthritis is related to the disorder of the subchondral bone microenvironment. Appropriate exercise can improve the subchondral bone microenvironment, thus playing an essential role in preventing and treating osteoarthritis. However, the exact mechanism whereby exercise improves the subchondral bone microenvironment remains unclear. There is biomechanical interaction as well as biochemical crosstalk between bone and cartilage. And the crosstalk between bone and cartilage is the key to bone-cartilage homeostasis maintenance. From the perspective of biomechanical and biochemical crosstalk between bone and cartilage, this paper reviews the effects of exercise-mediated bone-cartilage crosstalk on the subchondral bone microenvironment, aiming to provide a theoretical basis for the prevention and treatment of degenerative bone diseases.
Collapse
Affiliation(s)
- Shihua Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Tingting Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yao Feng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Keping Zhang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiquan Weng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Lan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- College of Sports and Health, Shandong Sport University, Jinan, China
| |
Collapse
|
14
|
Saito H, Yayama T, Mori K, Kumagai K, Fujikawa H, Chosei Y, Imai S. Increased Cellular Expression of Interleukin-6 in Patients With Ossification of the Posterior Longitudinal Ligament. Spine (Phila Pa 1976) 2023; 48:E78-E86. [PMID: 36729990 DOI: 10.1097/brs.0000000000004557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/24/2022] [Indexed: 02/03/2023]
Abstract
STUDY DESIGN We performed histologic, immunohistochemical, immunoblot examination and suspension array analyses of cytokine expression in cultured cells derived from human cervical ossification of the posterior longitudinal ligament (OPLL). OBJECTIVE To determine the roles of interleukin-6 (IL-6) during the maturation of osteoblasts and chondrocytes associated with the development of OPLL. SUMMARY OF BACKGROUND DATA Ectopic OPLL affects ~3% of the general population, with a higher incidence in Asian ethnic groups. Alterations in cytokine profiles may influence osteoblast differentiation, but the mechanisms and signaling pathways associated with the ossification process remain unclear. METHODS Samples were collected from 14 patients with OPLL who had undergone spinal surgery and seven with cervical spondylotic myelopathy without OPLL. Tissue sections were used for histologic and immunohistochemical studies, and primary cells from ligamentum samples were used for cytokine array and immunoblotting. A suspension array was used to measure the concentrations of 27 inflammatory cytokines or growth factors. RESULTS Suspension array and immunoblot analyses revealed significantly elevated levels of IL-6 in OPLL patients. Alterations in IL-6 concentrations were found to alter the expression of the genes Sox9 , Runx2 , and SIRT1 . In addition, immunohistochemical analysis revealed that these factors are present in mesenchymal cells within the degenerative portion of the ligament matrix that is adjacent to the ossification front. CONCLUSIONS IL-6 plays a profound role in the osteoblast differentiation process along with the induction of chondrocyte hypertrophy and cell apoptosis in the early stages of ossification in OPLL. These changes in cytokine profiles are essential factors for regulation of the ectopic ossified plaque in OPLL.
Collapse
Affiliation(s)
- Hideki Saito
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Shiga, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Ho TJ, Lin JH, Lin SZ, Tsai WT, Wu JR, Chen HP. Isolation, Identification, and Characterization of Bioactive Peptides in Human Bone Cells from Tortoiseshell and Deer Antler Gelatin. Int J Mol Sci 2023; 24:ijms24021759. [PMID: 36675272 PMCID: PMC9861678 DOI: 10.3390/ijms24021759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Tortoiseshell and deer antler gelatin has been used to treat bone diseases in Chinese society. A pepsin-digested gelatin peptide with osteoblast-proliferation-stimulating properties was identified via LC-MS/MS. The resulting pentapeptide, TSKYR, was presumably subjected to further degradation into TSKY, TSK, and YR fragments in the small intestine. The above four peptides were chemically synthesized. Treatment of tripeptide TSK can lead to a significant 30- and 50-fold increase in the mineralized nodule area and density in osteoblast cells and a 47.5% increase in the number of chondrocyte cells. The calcium content in tortoiseshell was relatively higher than in human soft tissue. The synergistic effects of calcium ions and the peptides were observed for changes in osteoblast proliferation and differentiation. Moreover, these peptides can enhance the expression of RUNX2, OCN, FGFR2, and FRFR3 genes in osteoblasts, and aggrecan and collagen type II in chondrocyte (patent pending).
Collapse
Affiliation(s)
- Tsung-Jung Ho
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970473, Taiwan
| | - Jung-Hsing Lin
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Shinn Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
| | - Wan-Ting Tsai
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
| | - Jia-Ru Wu
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
- Correspondence: (J.-R.W.); (H.-P.C.)
| | - Hao-Ping Chen
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 970473, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
- Correspondence: (J.-R.W.); (H.-P.C.)
| |
Collapse
|
16
|
Dilley JE, Bello MA, Roman N, McKinley T, Sankar U. Post-traumatic osteoarthritis: A review of pathogenic mechanisms and novel targets for mitigation. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
17
|
Jiang A, Xu P, Yang Z, Zhao Z, Tan Q, Li W, Song C, Dai H, Leng H. Increased Sparc release from subchondral osteoblasts promotes articular chondrocyte degeneration under estrogen withdrawal. Osteoarthritis Cartilage 2023; 31:26-38. [PMID: 36241137 DOI: 10.1016/j.joca.2022.08.020] [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: 04/04/2022] [Revised: 07/08/2022] [Accepted: 08/04/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The incidence of osteoarthritis (OA) in menopausal women is significantly higher than in same-aged men. Investigating the role of subchondral osteoblasts in estrogen deficiency-induced OA may help elucidate the pathological mechanism, providing new insights for the diagnosis and treatment of menopausal OA. METHODS A classical ovariectomy-induced OA (OVX-OA) rat model was utilized to isolate primary articular chondrocytes and subchondral osteoblasts, which were identified and then cocultured in Transwell. The expression of chondrocyte anabolic and catabolic indicators was evaluated. The differentially expressed proteins in the conditioned medium (CM) of osteoblasts were identified by Liquid Chromatograph-Mass Spectrometer (LC-MS/MS). Normal chondrocytes were treated with osteoblast CM, and then RNA sequencing was performed on the treated chondrocytes. KEGG was used to identify significant enrichment of signaling pathways, and Simple Western was used to verify the expression of related proteins in the signaling pathways. RESULTS Coculture of OVX-OA subchondral osteoblasts with chondrocytes significantly downregulated the expression of the anabolic indicators and upregulated the expression of the catabolic indicators in chondrocytes. 1,601 proteins were identified in both normal and OVX osteoblast culture supernatants. Protein-protein interaction network analysis revealed that Sparc was one of the hub proteins. The AMPK/Foxo3a signaling pathway of chondrocytes was downregulated by OVX-OA osteoblasts CM. AICAR, the AMPK agonist, partially reversed the catabolic effect of OVX-OA osteoblasts on chondrocytes. CONCLUSIONS Sparc secreted by OVX-OA subchondral osteoblasts can downregulate the AMPK/Foxo3a signaling pathway of chondrocytes, thereby promoting chondrocyte degeneration.
Collapse
Affiliation(s)
- A Jiang
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; Department of General Surgery, Beijing Pinggu Hospital, Beijing 101299, China
| | - P Xu
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Z Yang
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China
| | - Z Zhao
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China
| | - Q Tan
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China
| | - W Li
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; Engineering Research Center of Bone and Joint Precision Medicine, Beijing 100191, China
| | - C Song
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; Beijing Key Lab of Spine Diseases, Beijing 100191, China
| | - H Dai
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - H Leng
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
18
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
19
|
Yuan W, Wu Y, Huang M, Zhou X, Liu J, Yi Y, Wang J, Liu J. A new frontier in temporomandibular joint osteoarthritis treatment: Exosome-based therapeutic strategy. Front Bioeng Biotechnol 2022; 10:1074536. [PMID: 36507254 PMCID: PMC9732036 DOI: 10.3389/fbioe.2022.1074536] [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: 10/19/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a debilitating degenerative disease with high incidence, deteriorating quality of patient life. Currently, due to ambiguous etiology, the traditional clinical strategies of TMJOA emphasize on symptomatic treatments such as pain relief and inflammation alleviation, which are unable to halt or reverse the destruction of cartilage or subchondral bone. A number of studies have suggested the potential application prospect of mesenchymal stem cells (MSCs)-based therapy in TMJOA and other cartilage injury. Worthy of note, exosomes are increasingly being considered the principal efficacious agent of MSC secretions for TMJOA management. The extensive study of exosomes (derived from MSCs, synoviocytes, chondrocytes or adipose tissue et al.) on arthritis recently, has indicated exosomes and their specific miRNA components to be potential therapeutic agents for TMJOA. In this review, we aim to systematically summarize therapeutic properties and underlying mechanisms of MSCs and exosomes from different sources in TMJOA, also analyze and discuss the approaches to optimization, challenges, and prospects of exosome-based therapeutic strategy.
Collapse
Affiliation(s)
- Wenxiu Yuan
- Lab for Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yange Wu
- Lab for Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Maotuan Huang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xueman Zhou
- Lab for Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiaqi Liu
- Lab for Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yating Yi
- Lab for Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China,*Correspondence: Jin Liu, ; Jun Wang,
| | - Jin Liu
- Lab for Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Jin Liu, ; Jun Wang,
| |
Collapse
|
20
|
Biglycan neo-epitope (BGN 262), a novel biomarker for screening early changes in equine osteoarthritic subchondral bone. Osteoarthritis Cartilage 2022; 30:1328-1336. [PMID: 35870736 DOI: 10.1016/j.joca.2022.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Native biglycan (BGN), which can undergo proteolytic cleavage in pathological conditions, is well known to be involved in bone formation and mineralization. This study aimed to delineate the specific cleavage fragment, a neo-epitope for BGN (BGN262), in synovial fluid (SF) from young racehorses in training, osteoarthritic (OA) joints with subchondral bone sclerosis (SCBS), and chip fracture joints. DESIGN A custom-made inhibition ELISA was developed to quantify BGN262 in SF. Cohort 1: A longitudinal study comprising 10 racehorses undergoing long-term training. Cohort 2: A cross-sectional study comprising joints from horses (N = 69) with different stages of OA and radiographically classified SCBS. Cohort 3: A cross-sectional study comprising horses (N = 9) with chip fractures. Receiver operating characteristic (ROC) curve analysis was performed (healthy joints vs chip joints) to evaluate BGN262 robustness. RESULTS Cohort 1: SF BGN262 levels from racehorses showed a statistical increase during the first 6 months of the training period. Cohort 2: BGN262 levels were significantly higher in the SF from severe SCBS joints. Cohort 3: SF BGN262 levels in chip fracture joints showed a significant increase compared to normal joints. The ROC analysis showed an AUC of 0.957 (95% C.I 0.868-1.046), indicating good separation between the groups. CONCLUSIONS The data presented show that BGN262 levels increase in SF in correlation with the initiation of training, severity of SCBS, and presence of chip fractures. This suggests that BGN262 is a potential predictor and a novel biomarker for early changes in subchondral bone (SCB), aiming to prevent catastrophic injuries in racehorses.
Collapse
|
21
|
Wu Y, Li J, Zeng Y, Pu W, Mu X, Sun K, Peng Y, Shen B. Exosomes rewire the cartilage microenvironment in osteoarthritis: from intercellular communication to therapeutic strategies. Int J Oral Sci 2022; 14:40. [PMID: 35927232 PMCID: PMC9352673 DOI: 10.1038/s41368-022-00187-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage loss and accounts for a major source of pain and disability worldwide. However, effective strategies for cartilage repair are lacking, and patients with advanced OA usually need joint replacement. Better comprehending OA pathogenesis may lead to transformative therapeutics. Recently studies have reported that exosomes act as a new means of cell-to-cell communication by delivering multiple bioactive molecules to create a particular microenvironment that tunes cartilage behavior. Specifically, exosome cargos, such as noncoding RNAs (ncRNAs) and proteins, play a crucial role in OA progression by regulating the proliferation, apoptosis, autophagy, and inflammatory response of joint cells, rendering them promising candidates for OA monitoring and treatment. This review systematically summarizes the current insight regarding the biogenesis and function of exosomes and their potential as therapeutic tools targeting cell-to-cell communication in OA, suggesting new realms to improve OA management.
Collapse
Affiliation(s)
- Yuangang Wu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Li
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zeng
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenchen Pu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Mu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kaibo Sun
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Bin Shen
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
22
|
Bao Z, Chen M, Li C, Shan Q, Wang Y, Yang W. Monosodium iodoacetate-induced subchondral bone microstructure and inflammatory changes in an animal model of osteoarthritis. Open Life Sci 2022; 17:781-793. [PMID: 35903181 PMCID: PMC9287849 DOI: 10.1515/biol-2022-0079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/02/2022] [Accepted: 04/21/2022] [Indexed: 11/15/2022] Open
Abstract
The monosodium iodoacetate (MIA)-induced osteoarthritis (OA) may lead to cartilage degeneration and histopathological lesions. However, the correlation between inflammatory reaction and subchondral bone remodeling in a rodent osteoarthritic model is ambiguous. In this study, intra-articular injection of MIA was performed in 36 four-week-old specific pathogen-free male Wistar rats to induce OA. After 4 weeks of intervention, changes in intrinsic structural properties of the subchondral bones were measured, and the histological evaluation, as well as biochemical analysis, was conducted. We found that intra-articular injection of MIA increased chondrocyte apoptosis and promoted cartilage matrix degradation, such as cartilage surface defects and shallow or disappearing staining. MIA also induced inflammation, improved the expression of IL-1β, TNF-α, and matrix metalloproteinase, and decreased the expression of cartilage-specific proteins with the extension of modeling time. Meanwhile, the MIA also significantly accelerated the subchondral bone remodeling, as shown by the decreased subchondral bone density, thinning of trabeculae, disordered cartilage structure, and morphology. In conclusion, we have shown that MIA-induced rodent osteoarthritic model would cause decreased subchondral bone density, sparse trabecular bone, and other manifestations of osteoporosis accompanied by an inflammatory response, which would worsen with the progression of modeling time. Our results suggest that different phases of MIA-induced OA are associated with the changes in subchondral bone microstructure and the progression of local inflammation.
Collapse
Affiliation(s)
- Zheming Bao
- Department of Pharmacy, Medical Supplies Centre of PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
- Orthopedics Department, 960th Hospital of PLA Joint Service Support Force, Jinan, China
| | - Mengli Chen
- Department of Pharmacy, Medical Supplies Centre of PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Chen Li
- Department of Pharmacy, Medical Supplies Centre of PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Qing Shan
- Department of Pharmacy, Medical Supplies Centre of PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Yichen Wang
- Department of Pharmacy, Medical Supplies Centre of PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| | - Wenshan Yang
- Department of Pharmacy, Medical Supplies Centre of PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
23
|
Osteoblasts induce glucose-derived ATP perturbations in chondrocytes through noncontact communication. Acta Biochim Biophys Sin (Shanghai) 2022; 54:625-636. [PMID: 35593470 PMCID: PMC9828329 DOI: 10.3724/abbs.2022042] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cartilage and subchondral bone communicate with each other through material and signal exchanges. However, direct evidence provided by experimental studies on their interactions is insufficient. In the present study, we establish a noncontact co-culture model with a transwell chamber to explore the energetic perturbations in chondrocytes influenced by osteoblasts. Our results indicate that osteoblasts induce more ATP generation in chondrocytes through an energetic shift characterized by enhanced glycolysis and impaired mitochondrial tricarboxylic acid cycle. Enhanced glycolysis is shown by an increase of secreted lactate and the upregulation of glycolytic enzymes, including glucose-6-phosphate isomerase (Gpi), liver type ATP-dependent 6-phosphofructokinase (Pfkl), fructose-bisphosphate aldolase C (Aldoc), glyceraldehyde-3-phosphate dehydrogenase (Gapdh), triosephosphate isomerase (Tpi1), and phosphoglycerate kinase 1 (Pgk1). Impaired mitochondrial tricarboxylic acid cycle is characterized by the downregulation of cytoplasmic aspartate aminotransferase (Got1) and mitochondrial citrate synthase (Cs). Osteoblasts induce the activation of Akt and P38 signaling to mediate ATP perturbations in chondrocytes. This study may deepen our understanding of the maintenance of metabolic homeostasis in the bone-cartilage unit.
Collapse
|
24
|
Chen Y, Chen Q, Zhong M, Xu C, Wu Y, Chen R. miR-637 Inhibits Osteogenic Differentiation of Human Intervertebral Disc Cartilage Endplate Stem Cells by Targeting WNT5A. J INVEST SURG 2022; 35:1313-1321. [PMID: 35296211 DOI: 10.1080/08941939.2022.2050857] [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: 10/18/2022]
Abstract
Background: Degenerative disk disease (DDD) remains the leading incentive of severe lumbago. DDD is mainly caused by degeneration of cartilage endplate (CEP). Cartilage endplate stem cells (CESCs) are essential in chondrogenesis and osteogenesis of CEP. This study investigated the mechanism of miR-637 inhibiting osteogenic differentiation of human CESC by regulating WNT5A.Methods: The degenerative CEP (N = 10) and non-degenerative CEP (N = 6) were obtained from patients undergoing disk fusion surgery. CESCs were examined for surface stem cell markers, alkaline phosphatase (ALP) levels, osteogenic differentiation, osteogenic genes (Runx2, COL1), and chondrogenic gene (COL2). The miR-637 expression in CESCs was detected. The targeting relationship of miR-637 and WNT5A was confirmed. After miR-637 overexpression/WNT5A down-regulation, the action of miR-637/WNT5A on osteogenic differentiation of CESCs was evaluated. After simultaneous overexpression of miR-637/WNT5A, the effect of miR-637 on osteogenic differentiation of CESCs was assessed.Results: miR-637 was down-expressed in degenerative CESCs (D-CESCs), and miR-637 overexpression inhibited the osteogenic differentiation of D-CESCs, while inhibition of miR-637 promoted the osteogenic differentiation ability of D-CESCs. miR-637 targeted WNT5A and down-regulation of WNT5A inhibited the osteogenic differentiation of D-CESCs. Up-regulated WNT5A partially annulled the inhibitory action of miR-637 overexpression on osteogenic differentiation of D-CESCs.Conclusion: miR-637 inhibited osteogenic differentiation of D-CESCs via targeting WNT5A.
Collapse
Affiliation(s)
- Yunsheng Chen
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Qin Chen
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Mingliang Zhong
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Canhua Xu
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Yaohong Wu
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Rongchun Chen
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
25
|
Muenzebrock KA, Kersten V, Alblas J, Garcia JP, Creemers LB. The Added Value of the “Co” in Co-Culture Systems in Research on Osteoarthritis Pathology and Treatment Development. Front Bioeng Biotechnol 2022; 10:843056. [PMID: 35309991 PMCID: PMC8927651 DOI: 10.3389/fbioe.2022.843056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent disease and a major health burden. Its development and progression are influenced by factors such as age, obesity or joint overuse. As a whole organ disease OA affects not only cartilage, bone and synovium but also ligaments, fatty or nervous tissue surrounding the joint. These joint tissues interact with each other and understanding this interaction is important in developing novel treatments. To incorporate and study these interactions in OA research, several co-culture models have evolved. They combine two or more cell types or tissues and investigate the influence of amongst others inflammatory or degenerative stimuli seen in OA. This review focuses on co-cultures and the differential processes occurring in a given tissue or cell as a consequence of being combined with another joint cell type or tissue, and/or the extent to which a co-culture mimics the in vivo processes. Most co-culture models depart from synovial lining and cartilage culture, but also fat pad and bone have been included. Not all of the models appear to reflect the postulated in vivo OA pathophysiology, although some of the discrepancies may indicate current assumptions on this process are not entirely valid. Systematic analysis of the mutual influence the separate compartments in a given model exert on each other and validation against in vivo or ex vivo observation is still largely lacking and would increase their added value as in vitro OA models.
Collapse
|
26
|
The Role of Synovial Membrane in the Development of a Potential In Vitro Model of Osteoarthritis. Int J Mol Sci 2022; 23:ijms23052475. [PMID: 35269618 PMCID: PMC8910122 DOI: 10.3390/ijms23052475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 01/15/2023] Open
Abstract
There is a lack of in vitro models able to plausibly represent the inflammation microenvironment of knee osteoarthritis (OA). We analyzed the molecules released from OA tissues (synovial membrane, cartilage, infrapatellar fat pad) and investigated whether the stimulation of human synovial fibroblasts (SFs), with synthetic cytokines (IL-1β and TNF-α or IFN-γ) or conditioned media (CM) from OA tissues, influence the SFs’ response, in the sense of pro-inflammatory cytokines, chemokines, growth factors, and degradative enzymes modulation. Human SFs were obtained from OA synovial membranes. SFs and their CM were analyzed for biomarkers, proliferation rate, protein profile and gene expression, before and after stimulation. Real-time PCR and multiplex assays quantified OA-related gene expression and biomolecule production. Unlike other activators, CM from OA synovial membrane (CM-SM), significantly up-regulated all genes of interest (IL-6, IL-8, MMP-1, MMP-3, RANTES, MCP-1, TSG-6, YKL-40) in SFs. Multiplex immunoassay analysis showed that levels of OA-related cytokines (IL-6, IL-8, MCP 1, IL-1Ra), chemokine (RANTES) and growth factor (VEGF), produced by CM-SM stimulated SFs, increased significantly compared to non-stimulated SFs. Molecules released from the SM from OA patients induces OA-like changes in vitro, in specific OA synovial populations (SFs). These findings promote the use and establish a compelling in vitro model that simulates the versatility and complexity of the OA disease. This model, in the future, will allow us to study new cell therapies or test drugs by reducing or avoiding animal models.
Collapse
|
27
|
Lin C, Chen Z, Guo D, Zhou L, Lin S, Li C, Li S, Wang X, Lin B, Ding Y. Increased expression of osteopontin in subchondral bone promotes bone turnover and remodeling, and accelerates the progression of OA in a mouse model. Aging (Albany NY) 2022; 14:253-271. [PMID: 34982732 PMCID: PMC8791213 DOI: 10.18632/aging.203707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/28/2021] [Indexed: 02/05/2023]
Abstract
Osteopontin (OPN) has been proved to be closely related to the pathogenesis of osteoarthritis (OA), but the role of OPN in the pathogenesis of OA has not been fully clarified. Current studies on OPN in OA mostly focus on articular cartilage, synovial membrane and articular fluid, while ignoring its role in OA subchondral bone turnover and remodeling. In this study, we used a destabilization OA mouse model to investigate the role of OPN in OA subchondral bone changes. Our results indicate that increased expression of OPN accelerates the turnover and remodeling of OA subchondral bone, promotes the formation of h-type vessels in subchondral bone, and mediates articular cartilage degeneration induced by subchondral bone metabolism. In addition, our results confirmed that inhibition of PI3K/AKT signaling pathway inhibits OPN-mediated OA subchondral bone remodeling and cartilage degeneration. This study revealed the role and mechanism of OPN in OA subchondral bone, which is of great significance for exploring specific biological indicators for early diagnosis of OA and monitoring disease progression, as well as for developing drugs to regulate the metabolism and turnover of subchondral bone and alleviate the subchondral bone sclerosis of OA.
Collapse
Affiliation(s)
- Chuangxin Lin
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
- Department of Orthopedic Surgery, Shantou Central Hospital, Shantou 515000, P.R. China
| | - Zhong Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Dong Guo
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, P.R China
| | - Laixi Zhou
- Department of Orthopedic Surgery, Shantou Central Hospital, Shantou 515000, P.R. China
| | - Sipeng Lin
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Changchuan Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Shixun Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Xinjia Wang
- Department of Orthopedic, Affiliated Cancer Hospital, Shantou University Medical College, Shantou 515041, P.R. China
| | - Bendan Lin
- Department of Orthopedic Surgery, Shantou Central Hospital, Shantou 515000, P.R. China
| | - Yue Ding
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| |
Collapse
|
28
|
Wang Y, Zhang T, Xu Y, Chen R, Qu N, Zhang B, Xia C. Suppressing phosphoinositide-specific phospholipases Cγ1 promotes mineralization of osteoarthritic subchondral bone osteoblasts via increasing autophagy, thereby ameliorating articular cartilage degeneration. Bone 2022; 154:116262. [PMID: 34813965 DOI: 10.1016/j.bone.2021.116262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/11/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Phosphoinositide-specific phospholipases C-γ1 (PLC-γ1) signaling has been shown to modulate osteoarthritis (OA) chondrocyte metabolism. However, the role of PLC-γ1 in OA osteoblasts remains unclear. Herein, whether and how PLC-γ1 was involved in mineralization in OA subchondral bone osteoblasts were investigated. METHODS Primary non-OA and OA osteoblasts of human and rat isolated from the subchondral bone or the calvaria were cultured in vitro, as well as mouse pre-osteoblastic cell line MC3T3-E1 cells. Rat knee OA model was induced by anterior cruciate ligament transection (ACLT), in which bone canal was carried out from the surface of lateral epicondyle of femur using micro-electric drill. Morphological characteristics of subchondral bone structure and articular cartilage were assessed using CT, micro-CT, and Safranin O/Fast green staining, respectively. Mineralization was measured by alizarin red staining. The expression and production of genes involved in osteoblastic phenotype and mineralization were evaluated by qPCR, western blotting, and immunohistochemistry assays, respectively. The inhibitions were performed using inhibitors and ShRNAs. RESULTS The decreased relative bone density and thickness in the early stage of OA and the increased one in the late stage of OA were observed in subchondral bone of ACLT-rat model. Decreased ALP and OCN levels and absorbance values of ARS content were observed in in vitro osteoblasts isolated from 2 w post-ACLT rat model, as well as IL-1β-treated (for maintaining and mimicking inflammatory status) human OA and rat osteoblasts. Decreased Atg7 level and LC3BII/I ratio in combination with an increase in the P62 level, was concomitant with decreased ALP and OCN mRNA levels and absorbance values of ARS content in OA or IL-1β-treated osteoblasts. Specific inhibition of PLC-γ1 by ShRNAs or inhibitor (U73122) elevated ALP and OCN mRNA levels and absorbance values of ARS content accompanied with increased Atg7 level and LC3BII/I ratio in combination with a decrease in the P62 level in OA osteoblasts. Furthermore, the promoting effect of PLC-γ1 inhibition on ALP and OCN mRNA levels and absorbance values of ARS content was reversed by endoplasmic reticulum (ER) stress activator HA15, as well as autophagic inhibitors CQ and 3MA. Injection with PLC-γ1 inhibitor U73122 from the surface of lateral epicondyle of femur reduced aberrant subchondral bone formation and attenuated articular cartilage degeneration in ACLT-rat. CONCLUSION Aberrant changes of OA subchondral bone structure were concomitant with altered osteoblastic phenotype and mineralization. Impaired autophagy contributed to decreased osteoblastic mineralization in the early stage of OA. PLC-γ1 inhibition promoted osteoblastic mineralization through increasing autophagy in OA osteoblasts, which was partially attributed to suppression of ER stress. Targeting PLC-γ1 in subchondral bone osteoblasts could be more efficacious for OA therapy through treating the bone and cartilage at the same time. In summary, we hypothesize that suppressing PLCγ1 promotes mineralization of osteoarthritic subchondral bone osteoblasts via increasing autophagy, thereby ameliorating articular cartilage degeneration.
Collapse
Affiliation(s)
- Yue Wang
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Tongen Zhang
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Yang Xu
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Rui Chen
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ning Qu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Bing Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Chun Xia
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
29
|
Larrañaga-Vera A, Marco-Bonilla M, Largo R, Herrero-Beaumont G, Mediero A, Cronstein B. ATP transporters in the joints. Purinergic Signal 2021; 17:591-605. [PMID: 34392490 PMCID: PMC8677878 DOI: 10.1007/s11302-021-09810-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular adenosine triphosphate (ATP) plays a central role in a wide variety of joint diseases. ATP is generated intracellularly, and the concentration of the extracellular ATP pool is determined by the regulation of its transport out of the cell. A variety of ATP transporters have been described, with connexins and pannexins the most commonly cited. Both form intercellular channels, known as gap junctions, that facilitate the transport of various small molecules between cells and mediate cell-cell communication. Connexins and pannexins also form pores, or hemichannels, that are permeable to certain molecules, including ATP. All joint tissues express one or more connexins and pannexins, and their expression is altered in some pathological conditions, such as osteoarthritis (OA) and rheumatoid arthritis (RA), indicating that they may be involved in the onset and progression of these pathologies. The aging of the global population, along with increases in the prevalence of obesity and metabolic dysfunction, is associated with a rising frequency of joint diseases along with the increased costs and burden of related illness. The modulation of connexins and pannexins represents an attractive therapeutic target in joint disease, but their complex regulation, their combination of gap-junction-dependent and -independent functions, and their interplay between gap junction and hemichannel formation are not yet fully elucidated. In this review, we try to shed light on the regulation of these proteins and their roles in ATP transport to the extracellular space in the context of joint disease, and specifically OA and RA.
Collapse
Affiliation(s)
- Ane Larrañaga-Vera
- Department of Medicine, Division of Translational Medicine, NYU Langone Health, New York, NY, USA
| | - Miguel Marco-Bonilla
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain
| | - Raquel Largo
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain
| | | | - Aránzazu Mediero
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, 28040, Madrid, Spain.
| | - Bruce Cronstein
- Department of Medicine, Division of Translational Medicine, NYU Langone Health, New York, NY, USA
| |
Collapse
|
30
|
Jiang A, Xu P, Sun S, Zhao Z, Tan Q, Li W, Song C, Leng H. Cellular alterations and crosstalk in the osteochondral joint in osteoarthritis and promising therapeutic strategies. Connect Tissue Res 2021; 62:709-719. [PMID: 33397157 DOI: 10.1080/03008207.2020.1870969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/28/2020] [Indexed: 02/03/2023]
Abstract
Osteoarthritis (OA) is a joint disorder involving cartilage degeneration and subchondral bone sclerosis. The bone-cartilage interface is implicated in OA pathogenesis due to its susceptibility to mechanical and biological factors. The crosstalk between cartilage and the underlying subchondral bone is elevated in OA due to multiple factors, such as increased vascularization, porosity, microcracks and fissures. Changes in the osteochondral joint are traceable to alterations in chondrocytes and bone cells (osteoblasts, osteocytes and osteoclasts). The phenotypes of these cells can change with the progression of OA. Aberrant intercellular communications among bone cell-bone cell and bone cell-chondrocyte are of great importance and might be the factors promoting OA development. An appreciation of cellular phenotypic changes in OA and the mechanisms by which these cells communicate would be expected to lead to the development of targeted drugs with fewer side effects.
Collapse
Affiliation(s)
- Ai Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Peng Xu
- State Key Laboratory of Systematic and Evolutionary Botany, Institute of Botany, Chinese Academy of Sciences, Beijing, China
| | - Shang Sun
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Zhenda Zhao
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Qizhao Tan
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education Lisbon Portugal
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Lab of Spine Diseases, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
31
|
Characterization and miRNA Profiling of Extracellular Vesicles from Human Osteoarthritic Subchondral Bone Multipotential Stromal Cells (MSCs). Stem Cells Int 2021; 2021:7232773. [PMID: 34667479 PMCID: PMC8520657 DOI: 10.1155/2021/7232773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/26/2021] [Accepted: 08/19/2021] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is a heterogeneous disease in which the cross-talk between the cells from different tissues within the joint is affected as the disease progresses. Extracellular vesicles (EVs) are known to have a crucial role in cell-cell communication by means of cargo transfer. Subchondral bone (SB) resident cells and its microenvironment are increasingly recognised to have a major role in OA pathogenesis. The aim of this study was to investigate the EV production from OA SB mesenchymal stromal cells (MSCs) and their possible influence on OA chondrocytes. Small EVs were isolated from OA-MSCs, characterized and cocultured with chondrocytes for viability and gene expression analysis, and compared to small EVs from MSCs of healthy donors (H-EVs). OA-EVs enhanced viability of chondrocytes and the expression of chondrogenesis-related genes, although the effect was marginally lower compared to that of the H-EVs. miRNA profiling followed by unsupervised hierarchical clustering analysis revealed distinct microRNA sets in OA-EVs as compared to their parental MSCs or H-EVs. Pathway analysis of OA-EV miRNAs showed the enrichment of miRNAs implicated in chondrogenesis, stem cells, or other pathways related to cartilage and OA. In conclusion, OA SB MSCs were capable of producing EVs that could support chondrocyte viability and chondrogenic gene expression and contained microRNAs implicated in chondrogenesis support. These EVs could therefore mediate the cross-talk between the SB and cartilage in OA potentially modulating chondrocyte viability and endogenous cartilage regeneration.
Collapse
|
32
|
Singh YP, Moses JC, Bhardwaj N, Mandal BB. Overcoming the Dependence on Animal Models for Osteoarthritis Therapeutics - The Promises and Prospects of In Vitro Models. Adv Healthc Mater 2021; 10:e2100961. [PMID: 34302436 DOI: 10.1002/adhm.202100961] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/10/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis (OA) is a musculoskeletal disease characterized by progressive degeneration of osteochondral tissues. Current treatment is restricted to the reduction of pain and loss of function of the joint. To better comprehend the OA pathophysiological conditions, several models are employed, however; there is no consensus on a suitable model. In this review, different in vitro models being developed for possible therapeutic intervention of OA are outlined. Herein, various in vitro OA models starting from 2D model, co-culture model, 3D models, dynamic culture model to advanced technologies-based models such as 3D bioprinting, bioassembly, organoids, and organ-on-chip-based models are discussed with their advantages and disadvantages. Besides, different growth factors, cytokines, and chemicals being utilized for induction of OA condition are reviewed in detail. Furthermore, there is focus on scrutinizing different molecular and possible therapeutic targets for better understanding the mechanisms and OA therapeutics. Finally, the underlying challenges associated with in vitro models are discussed followed by future prospective. Taken together, a comprehensive overview of in vitro OA models, factors to induce OA-like conditions, and intricate molecular targets with the potential to develop personalized osteoarthritis therapeutics in the future with clinical translation is provided.
Collapse
Affiliation(s)
- Yogendra Pratap Singh
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Joseph Christakiran Moses
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Nandana Bhardwaj
- Department of Science and Mathematics Indian Institute of Information Technology Guwahati Bongora Guwahati Assam 781015 India
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Sciences and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
33
|
Ding X, Xiang W, Meng D, Chao W, Fei H, Wang W. Osteoblasts Regulate the Expression of ADAMTS and MMPs in Chondrocytes through ERK Signaling Pathway. ZEITSCHRIFT FUR ORTHOPADIE UND UNFALLCHIRURGIE 2021; 161:201-210. [PMID: 34500490 DOI: 10.1055/a-1527-7900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Degradative enzymes such as matrix metalloproteinase (MMP) and disintegrin metalloproteinase with platelet thrombin-sensitive protein-like motifs (ADAMTS) play a key role in the development of osteoarthritis (OA). We aimed to investigate the effects of OA subchondral osteoblasts on the expression of ADAMTS4, ADAMTS5, MMP-3, MMP-9, and MMP-13 in chondrocytes and the regulation of mitogen-activated protein kinase (MAPK) signaling pathway. METHODS A rat knee OA model was constructed by cutting the anterior cruciate ligament of the knee joints, and normal rat articular cartilage chondrocytes (N-ACC), OA rat articular cartilage chondrocytes (O-ACC), normal subchondral bone osteoblasts (N-SBO), and OA subchondral bone osteoblasts (O-SBO) were isolated and extracted. The expressions of O-ACC and O-SBO COL1 and COL2 were detected respectively. Chondrocytes were identified by immunofluorescence of COL2 and toluidine blue staining, and osteoblasts were identified by COL1 immunofluorescence, alkaline phosphatase (ALP), and Alizarin Red staining. Gene expression of COL1, COL2, and aggrecan in normal chondrocytes and OA chondrocytes, and gene expression of osteoblast ALP and osteocalcin (OCN) were detected by RT-PCR to identify the two chondrocytes and the two osteoblast phenotypes. The constructing N-ACC group, O-ACC group, N-ACC + N-SBO group, N-ACC + O-SBO group, O-ACC + N-SBO group, O-ACC + O-SBO group, I + N-ACC + O-SBO group, and I + O-ACC + O-SBO group cell cultures, and the expression of ERK, ADAMTS4, ADAMTS5, MMP-3, MMP-9, and MMP-13 genes in chondrocytes cultured for 0, 24, 48, and 72 h were detected by RT-PCR. The protein expressions of pERK, ADAMTS4, ADAMTS5, MMP-3, MMP-9, and MMP-13 were detected by Western blot. RESULTS · The X-ray showed that the knee joint space of the affected limb became narrow.. · The results of RT-PCR of COL2 and aggrecan gene in OA and normal chondrocytes suggest that the relative expression of COL2 in OA articular chondrocytes (0.24 ± 0.07) is significantly lower than that in normal cartilage (0.61 ± 0.07) (p < 0.05). The relative expression of AGG (0.37 ± 0.16) in OA chondrocytes was significantly lower than that of normal chondrocytes AGG (1.30 ± 0.25) (p < 0.05). The expression of COL1 was very low, and was not statistically significant.. · The results of RT-PCR of the osteoblast ALP and OCN gene indicated that gene expression of ALP (12.30 ± 1.17) and OCN (20.47 ± 4.19)was upregulated when compared with the relative expression of ALP (4.66 ± 0.71) (p < 0.05) and OCN (12.17 ± 2.76) (p < 0.05) in normal osteoblasts, indicating that osteoblasts of OA have greater osteogenic potential than normal osteoblasts.. · The expressions of ADAMTS4, ADAMTS5, MMP-3, MMP-9, and MMP-13 genes and proteins in OA chondrocytes or normal chondrocytes were basically unchanged when they were cocultured with normal osteoblasts. Indirect coculture of OA osteoblasts and chondrocytes could promote the expression of ADAMTS4, ADAMTS5, MMP-3, MMP-9, and MMP-13 genes and proteins in chondrocytes. Overexpression of ADAMTS and MMP in coculture systems can be reversed by MAPK-ERK inhibitors.. CONCLUSIONS · OA subchondral bone osteoblasts can promote the overexpression of ADAMTS and MMPs in chondrocytes.. · The ERK signaling pathway may be involved in the regulation of the effect of subchondral bone osteoblasts on chondrocytes..
Collapse
Affiliation(s)
- Xiao Ding
- Department of Orthopaedics, The First Affiliated Hospital of the Medical Colleges, Shihezi University, China
| | - Wei Xiang
- Department of Orthopaedics, The First Affiliated Hospital of the Medical Colleges, Shihezi University, China
| | - Defeng Meng
- Department of Orthopaedics, The First Affiliated Hospital of the Medical Colleges, Shihezi University, China
| | - Wang Chao
- Department of Orthopaedics, The First Affiliated Hospital of the Medical Colleges, Shihezi University, China
| | - Han Fei
- Department of Orthopaedics, The First Affiliated Hospital of the Medical Colleges, Shihezi University, China
| | - Weishan Wang
- Department of Orthopaedics, The First Affiliated Hospital of the Medical Colleges, Shihezi University, China
| |
Collapse
|
34
|
Song CX, Liu SY, Zhu WT, Xu SY, Ni GX. Excessive mechanical stretch‑mediated osteoblasts promote the catabolism and apoptosis of chondrocytes via the Wnt/β‑catenin signaling pathway. Mol Med Rep 2021; 24:593. [PMID: 34165157 PMCID: PMC8222797 DOI: 10.3892/mmr.2021.12232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 05/05/2021] [Indexed: 02/05/2023] Open
Abstract
Excessive biomechanical loading is considered an important cause of osteoarthritis. Although the mechanical responses of chondrocytes and osteoblasts have been investigated, their communication during mechanical loading and the underlying molecular mechanisms are not yet fully known. The present study investigated the effects of excessive mechanically stretched osteoblasts on the metabolism and apoptosis of chondrocytes, and also assessed the involvement of the Wnt/β‑catenin signaling pathway. In the present study, rat chondrocytes and osteoblasts were subjected to mechanical tensile strain, and an indirect chondrocyte‑osteoblast co‑culture model was established. Reverse transcription‑quantitative PCR and western blotting were performed to determine the expression levels of genes and proteins of interest. An ELISA was performed to investigate the levels of cytokines, including matrix metalloproteinase (MMP) 13, MMP 3, interleukin‑6 (IL‑6) and prostaglandin E2 (PG E2), released from osteoblasts. Flow cytometry was performed to detect the apoptosis of chondrocytes exposed to stretched osteoblast conditioned culture medium. The levels of MMP 13, IL‑6 and PG E2 increased significantly in the supernatants of stretched osteoblasts compared with the un‑stretched group. By contrast, the mRNA expression levels of Collagen 1a and alkaline phosphatase were significantly decreased in osteoblasts subjected to mechanical stretch compared with the un‑stretched group. The mRNA expression level of Collagen 2a was significantly decreased, whereas the expression levels of MMP 13 and a disintegrin and metalloproteinase with thrombospondin‑like motifs 5 were significantly increased in chondrocytes subjected to mechanical stretch compared with the un‑stretched group. In the co‑culture model, the results indicated that excessive mechanically stretched osteoblasts induced the catabolism and apoptosis of chondrocytes, which was partly inhibited by Wnt inhibitor XAV‑939. The results of the present study demonstrated that excessive mechanical stretch led to chondrocyte degradation and inhibited osteoblast osteogenic differentiation; furthermore, excessive mechanically stretched osteoblasts induced the catabolism and apoptosis of chondrocytes via the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Cheng-Xian Song
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Sheng-Yao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Wen-Ting Zhu
- Department of Pharmacy, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Shao-Yong Xu
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Guo-Xin Ni
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, P.R. China
- Correspondence to: Professor Guo-Xin Ni, School of Sport Medicine and Rehabilitation, Beijing Sport University, 48 Xinxi Road, Haidian, Beijing 100084, P.R. China, E-mail:
| |
Collapse
|
35
|
Hopkins T, Wright KT, Kuiper NJ, Roberts S, Jermin P, Gallacher P, Kuiper JH. An In Vitro System to Study the Effect of Subchondral Bone Health on Articular Cartilage Repair in Humans. Cells 2021; 10:1903. [PMID: 34440671 PMCID: PMC8392168 DOI: 10.3390/cells10081903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
Chondrocyte-based cartilage repair strategies, such as articular chondrocyte implantation, are widely used, but few studies addressed the communication between native subchondral bone cells and the transplanted chondrocytes. An indirect co-culture model was developed, representing a chondrocyte/scaffold-construct repair of a cartilage defect adjoining bone, where the bone could have varying degrees of degeneration. Human BM-MSCs were isolated from two areas of subchondral bone in each of five osteochondral tissue specimens from five patients undergoing knee arthroplasty. These two areas underlaid the macroscopically and histologically best and worst cartilage, representing early and late-stage OA, respectively. BM-MSCs were co-cultured with normal chondrocytes suspended in agarose, with the two cell types separated by a porous membrane. After 0, 7, 14 and 21 days, chondrocyte-agarose scaffolds were assessed by gene expression and biochemical analyses, and the abundance of selected proteins in conditioned media was assessed by ELISA. Co-culture with late-OA BM-MSCs resulted in a reduction in GAG deposition and a decreased expression of genes encoding matrix-specific proteins (COL2A1 and ACAN), compared to culturing with early OA BM-MSCs. The concentration of TGF-β1 was significantly higher in the early OA conditioned media. The results of this study have clinical implications for cartilage repair, suggesting that the health of the subchondral bone may influence the outcomes of chondrocyte-based repair strategies.
Collapse
Affiliation(s)
- Timothy Hopkins
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Karina T. Wright
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Nicola J. Kuiper
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Sally Roberts
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Paul Jermin
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Peter Gallacher
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
| | - Jan Herman Kuiper
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| |
Collapse
|
36
|
Baek A, Jung SH, Pyo S, Kim SY, Jo S, Kim L, Lee EY, Kim SH, Cho SR. 3'-Sialyllactose Protects SW1353 Chondrocytic Cells From Interleukin-1β-Induced Oxidative Stress and Inflammation. Front Pharmacol 2021; 12:609817. [PMID: 33912037 PMCID: PMC8072478 DOI: 10.3389/fphar.2021.609817] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/19/2021] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is a major degenerative joint disease. Oxidative stress and inflammation play key roles in the pathogenesis of OA. 3'-Sialyllactose (3'-SL) is derived from human milk and is known to regulate a variety of biological functions related to immune homeostasis. This study aimed to investigate the therapeutic mechanisms of 3'-SL in interleukin-1β (IL-1β)-treated SW1353 chondrocytic cells. 3'-SL potently suppressed IL-1β-induced oxidative stress by increasing the levels of enzymatic antioxidants. 3'-SL significantly reversed the IL-1β mediated expression levels of reactive oxygen species in IL-1β-stimulated chondrocytic cells. In addition, 3'-SL could reverse the increased levels of inflammatory markers such as nitrite, prostaglandin E2, inducible nitric oxide synthase, cyclooxygenase-2, IL-1β, and IL-6 in IL-1β-stimulated chondrocytic cells. Moreover, 3'-SL significantly inhibited the apoptotic process, as indicated by the downregulation of the pro-apoptotic protein Bax, upregulation of the anti-apoptotic protein Bcl-2 expression, and significant reduction in the number of TUNEL-positive cells in the IL-1β-treated chondrocytic cells. Furthermore, 3'-SL reversed cartilage destruction by decreasing the release of matrix metalloproteinases (MMP), such as MMP1, MMP3, and MMP13. In contrast, 3'-SL significantly increased the expression levels of matrix synthesis proteins, such as collagen II and aggrecan, in IL-1β-treated chondrocytic cells. 3'-SL dramatically suppressed the activation of mitogen-activated protein kinases (MAPK) and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways, which are related to the pathogenesis of OA. Taken together, our data suggest that 3'-SL alleviates IL-1β-induced OA pathogenesis via inhibition of activated MAPK and PI3K/AKT/NF-κB signaling cascades with the downregulation of oxidative stress and inflammation. Therefore, 3'-SL has the potential to be used as a natural compound for OA therapy owing to its ability to activate the antioxidant defense system and suppress inflammatory responses.
Collapse
Affiliation(s)
- Ahreum Baek
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - So Hee Jung
- Department of Rehabilitation Medicine, The Graduate School Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Soonil Pyo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Yeon Kim
- Department of Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Seongmoon Jo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | | | - Eun Young Lee
- Department of Rehabilitation Medicine, The Graduate School Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sung Hoon Kim
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Graduate Program of Nano Science and Technology, Yonsei University College of Medicine, Seoul, Korea.,Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Kleuskens MWA, van Donkelaar CC, Kock LM, Janssen RPA, Ito K. An ex vivo human osteochondral culture model. J Orthop Res 2021; 39:871-879. [PMID: 32592503 PMCID: PMC8048497 DOI: 10.1002/jor.24789] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 02/04/2023]
Abstract
To reduce animal experimentation and to overcome translational issues in cartilage tissue engineering, there is a need to develop an ex vivo human tissue-based approach. This study aims to demonstrate that a human osteochondral explant at different stages of osteoarthritis (OA) can be kept in long-term culture while preserving its viability and composition. Osteochondral explants with either a smooth or fibrillated cartilage surface, representing different OA stages, were harvested from fresh human tibial plateaus. Explants were cultured for 2 or 4 weeks in a double-chamber culture platform. The biochemical content of the cartilage of cultured explants did not significantly change over a period of 4 weeks and these findings were supported by histology. Chondrocytes mostly preserved their metabolic activity during culture and active bone and marrow were found in the periphery of the explants, while metabolic activity was decreased in the bone core in cultured explants compared to fresh explants. In fibrillated explants, chondrocyte viability decreased in the periphery of the sample in cultured groups compared to fresh explants (fresh, 94 ± 6%; cultured, 64% ± 17%, 2 weeks, and 69% ± 17%, 4 weeks; P < .05). Although biochemical and histological results did not show changes within the cartilage tissue, the viability of the explants should be carefully controlled for each specific use. This system provides an alternative to explore drug treatment and implant performance under more controlled experimental conditions than possible in vivo, in combination with clinically relevant human osteochondral tissue.
Collapse
Affiliation(s)
- Meike W. A. Kleuskens
- Orthopaedic Biomechanics, Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| | - Corrinus C. van Donkelaar
- Orthopaedic Biomechanics, Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| | - Linda M. Kock
- Orthopaedic Biomechanics, Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands,LifeTec Group BVEindhovenThe Netherlands
| | - Rob P. A. Janssen
- Orthopaedic Biomechanics, Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands,Orthopaedic Center MáximaMáxima Medical Center Eindhoven/VeldhovenEindhovenThe Netherlands,Value‐Based Health Care, Faculty of Paramedical SciencesFontys University of Applied SciencesEindhovenThe Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| |
Collapse
|
38
|
Pirosa A, Gottardi R, Alexander PG, Puppi D, Chiellini F, Tuan RS. An in vitro chondro-osteo-vascular triphasic model of the osteochondral complex. Biomaterials 2021; 272:120773. [PMID: 33798958 DOI: 10.1016/j.biomaterials.2021.120773] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 01/06/2023]
Abstract
The generation of engineered models of the osteochondral complex to study its pathologies and develop possible treatments is hindered by the distinctly different properties of articular cartilage and subchondral bone, with the latter characterized by vascularization. In vitro models of the osteochondral complex have been mainly engineered as biphasic constructs containing just cartilage and bone cells, a condition very dissimilar from the in vivo environment. The different cellular components of the osteochondral complex are governed by interacting biochemical signaling; hence, to study the crosstalk among chondrocytes, osteoblasts, and endothelial cells, we have developed a novel triphasic model of the osteochondral tissue interface. Wet-spun poly(ε-caprolactone) (PCL) and PCL/hydroxyapatite (HA) scaffolds in combination with a methacrylated gelatin (gelMA) hydrogel were used as the polymeric backbone of the constructs. The scaffold components were engineered with human bone marrow derived mesenchymal stem cells (hMSCs) and human umbilical vein endothelial cells (HUVECs), and differentiated using a dual chamber microphysiological system (MPS) bioreactor that allows the simultaneous, separate flow of media of different compositions for induced differentiation of each compartment towards a cartilaginous or osseous lineage. Within the engineered Microphysiological Vascularized Osteochondral System, hMSCs showed spatially distinct chondrogenic and osteogenic markers in terms of histology and gene expression. HUVECs formed a stable capillary-like network in the engineered bone compartment and enhanced both chondrogenic and osteogenic differentiation of hMSCs, resulting in the generation of an in vitro system that mimics a vascularized osteochondral interface tissue.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Ri.MED Foundation, Palermo, Italy
| | - Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dario Puppi
- BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Federica Chiellini
- BIOlab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
39
|
Subchondral bone microenvironment in osteoarthritis and pain. Bone Res 2021; 9:20. [PMID: 33731688 PMCID: PMC7969608 DOI: 10.1038/s41413-021-00147-z] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
Osteoarthritis comprises several joint disorders characterized by articular cartilage degeneration and persistent pain, causing disability and economic burden. The incidence of osteoarthritis is rapidly increasing worldwide due to aging and obesity trends. Basic and clinical research on osteoarthritis has been carried out for decades, but many questions remain unanswered. The exact role of subchondral bone during the initiation and progression osteoarthritis remains unclear. Accumulating evidence shows that subchondral bone lesions, including bone marrow edema and angiogenesis, develop earlier than cartilage degeneration. Clinical interventions targeting subchondral bone have shown therapeutic potential, while others targeting cartilage have yielded disappointing results. Abnormal subchondral bone remodeling, angiogenesis and sensory nerve innervation contribute directly or indirectly to cartilage destruction and pain. This review is about bone-cartilage crosstalk, the subchondral microenvironment and the critical role of both in osteoarthritis progression. It also provides an update on the pathogenesis of and interventions for osteoarthritis and future research targeting subchondral bone.
Collapse
|
40
|
Zhang X, He J, Wang W. Progress in the use of mesenchymal stromal cells for osteoarthritis treatment. Cytotherapy 2021; 23:459-470. [PMID: 33736933 DOI: 10.1016/j.jcyt.2021.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/20/2020] [Accepted: 01/29/2021] [Indexed: 12/26/2022]
Abstract
LITERATURE REVIEW OF MSCS IN THE TREATMENT OF OSTEOARTHRITIS IN THE PAST FIVE YEARS: Osteoarthritis (OA) is one of the most common chronic joint diseases, with prominent symptoms caused by many factors. However, current medical interventions for OA have resulted in poor clinical outcomes, demonstrating that there are huge unmet medical needs in this area. Cell therapy has opened new avenues of OA treatment. Different sources of mesenchymal stromal cells (MSCs) may have different phenotypes and cellular functions. Pre-clinical and clinical studies have demonstrated the feasibility, safety and efficacy of MSC therapy. Mitogen-activated protein kinase, Wnt and Notch signaling pathways are involved in the chondrogenesis of MSC-mediated treatments. MSCs may also exert effective immunoregulatory and paracrine effects to stimulate tissue repair. Therapy with extracellular vesicles containing cytokines, which are secreted by MSCs, might be a potential treatment for OA.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiyin He
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wen Wang
- Clinical Development, IASO Biotherapeutics Co., Ltd., Shanghai, China.
| |
Collapse
|
41
|
Xiao H, Chen J, Duan L, Li S. Role of emerging vitamin K‑dependent proteins: Growth arrest‑specific protein 6, Gla‑rich protein and periostin (Review). Int J Mol Med 2021; 47:2. [PMID: 33448308 PMCID: PMC7834955 DOI: 10.3892/ijmm.2020.4835] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/21/2020] [Indexed: 01/27/2023] Open
Abstract
Vitamin K‑dependent proteins (VKDPs) are a group of proteins that need vitamin K to conduct carboxylation. Thus far, scholars have identified a total of 17 VKDPs in the human body. In this review, we summarize three important emerging VKDPs: Growth arrest‑specific protein 6 (Gas 6), Gla‑rich protein (GRP) and periostin in terms of their functions in physiological and pathological conditions. As examples, carboxylated Gas 6 and GRP effectively protect blood vessels from calcification, Gas 6 protects from acute kidney injury and is involved in chronic kidney disease, GRP contributes to bone homeostasis and delays the progression of osteoarthritis, and periostin is involved in all phases of fracture healing and assists myocardial regeneration in the early stages of myocardial infarction. However, periostin participates in the progression of cardiac fibrosis, idiopathic pulmonary fibrosis and airway remodeling of asthma. In addition, we discuss the relationship between vitamin K, VKDPs and cancer, and particularly the carboxylation state of VKDPs in cancer.
Collapse
Affiliation(s)
- Huiyu Xiao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| | - Jiepeng Chen
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Lili Duan
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| |
Collapse
|
42
|
Culibrk RA, Hahn MS. The Role of Chronic Inflammatory Bone and Joint Disorders in the Pathogenesis and Progression of Alzheimer's Disease. Front Aging Neurosci 2020; 12:583884. [PMID: 33364931 PMCID: PMC7750365 DOI: 10.3389/fnagi.2020.583884] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Late-onset Alzheimer's Disease (LOAD) is a devastating neurodegenerative disorder that causes significant cognitive debilitation in tens of millions of patients worldwide. Throughout disease progression, abnormal secretase activity results in the aberrant cleavage and subsequent aggregation of neurotoxic Aβ plaques in the cerebral extracellular space and hyperphosphorylation and destabilization of structural tau proteins surrounding neuronal microtubules. Both pathologies ultimately incite the propagation of a disease-associated subset of microglia-the principle immune cells of the brain-characterized by preferentially pro-inflammatory cytokine secretion and inhibited AD substrate uptake capacity, which further contribute to neuronal degeneration. For decades, chronic neuroinflammation has been identified as one of the cardinal pathophysiological driving features of AD; however, despite a number of works postulating the underlying mechanisms of inflammation-mediated neurodegeneration, its pathogenesis and relation to the inception of cognitive impairment remain obscure. Moreover, the limited clinical success of treatments targeting specific pathological features in the central nervous system (CNS) illustrates the need to investigate alternative, more holistic approaches for ameliorating AD outcomes. Accumulating evidence suggests significant interplay between peripheral immune activity and blood-brain barrier permeability, microglial activation and proliferation, and AD-related cognitive decline. In this work, we review a narrow but significant subset of chronic peripheral inflammatory conditions, describe how these pathologies are associated with the preponderance of neuroinflammation, and posit that we may exploit peripheral immune processes to design interventional, preventative therapies for LOAD. We then provide a comprehensive overview of notable treatment paradigms that have demonstrated considerable merit toward treating these disorders.
Collapse
Affiliation(s)
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
43
|
Azzini GOM, Santos GS, Visoni SBC, Azzini VOM, Santos RGD, Huber SC, Lana JF. Metabolic syndrome and subchondral bone alterations: The rise of osteoarthritis - A review. J Clin Orthop Trauma 2020; 11:S849-S855. [PMID: 32999567 PMCID: PMC7503158 DOI: 10.1016/j.jcot.2020.06.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/22/2022] Open
Abstract
Metabolic syndrome (MS) has become one of the top major health burdens for over three decades not only due to its effects on cardiovascular health but also its implications in orthopedics. Extensive research has shown that MS is tightly linked to osteoarthritis and inflammation, a process which appears to primarily occur in the subchondral bone via the incidence of bone-marrow lesions (BMLs). Numerous studies identify obesity, dyslipidemia, insulin resistance and hypertension as the top metabolic risk factors, the so-called "deadly quartet". These factors are responsible for the disruptive physiological processes that culminate in detrimental alterations within the subchondral bone, cartilage damage and, overall, the predominant pro-inflammatory joint microenvironment. Although it has long been thought that osteoarthritis was limited to the cartilage component of the joint, other studies indicate that the disease may originate from the harmful alterations that occur primarily in the subchondral bone, especially via means of vascular pathology. Since metabolic risk factors are manageable to a certain extent, it is therefore possible to decelerate the progression of OA and mitigate its devastating effects on the subchondral bone and subsequent articular cartilage damage. METHODS Literature was reviewed using PubMed and Google Scholar in order to find a correlation between metabolic syndrome and osteoarthritic progression. The investigation included a combination of nomenclature such as: "metabolic syndrome", "obesity", "insulin resistance", "hypertension", "dyslipidemia", "low-grade systemic inflammation", "osteoarthritis", "subchondral bone", "cartilage" and "inflammatory biomarkers". CONCLUSION Based on several studies, there seems to be a significant association between The Deadly Quartet (metabolic syndrome), dysregulation of both pro- and anti-inflammatory biomarkers, and osteoarthritic progression arising from unbridled systemic inflammation.
Collapse
Affiliation(s)
- Gabriel Ohana Marques Azzini
- Orthopedics, Sports Medicine, Pain Physician, IOC, Instituto do Osso e da Cartilagem, The Bone and Cartilage Institute, Presidente Kennedy Avenue, 1386, 2nd Floor, Room #29, Cidade Nova I, Indaiatuba, SP, Brazil
| | - Gabriel Silva Santos
- Biomedical Scientist, IOC, Instituto do Osso e da Cartilagem, The Bone and Cartilage Institute, Presidente Kennedy Avenue, 1386, 2nd Floor, Room #29, Cidade Nova I, Indaiatuba, SP, Brazil
| | - Silvia Beatriz Coutinho Visoni
- Biologist, IOC, Instituto do Osso e da Cartilagem, The Bone and Cartilage Institute, Presidente Kennedy Avenue, 1386, 2nd Floor, Room #29, Cidade Nova I, Indaiatuba, SP, Brazil
| | - Vitor Ohana Marques Azzini
- Orthopedics, Sports Medicine, Pain Physician, IOC, Instituto do Osso e da Cartilagem, The Bone and Cartilage Institute, Presidente Kennedy Avenue, 1386, 2nd Floor, Room #29, Cidade Nova I, Indaiatuba, SP, Brazil
| | - Rafael Gonzales dos Santos
- Orthopedics, Sports Medicine, Pain Physician, IOC, Instituto do Osso e da Cartilagem, The Bone and Cartilage Institute, Presidente Kennedy Avenue, 1386, 2nd Floor, Room #29, Cidade Nova I, Indaiatuba, SP, Brazil
| | - Stephany Cares Huber
- Biomedical Scientist, Universidade Estadual de Campinas (UNICAMP), The University of Campinas, Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - José Fábio Lana
- Orthopedics, Sports Medicine, Pain Physician, IOC, Instituto do Osso e da Cartilagem, The Bone and Cartilage Institute, Presidente Kennedy Avenue, 1386, 2nd Floor, Room #29, Cidade Nova I, Indaiatuba, SP, Brazil
| |
Collapse
|
44
|
Ayan B, Wu Y, Karuppagounder V, Kamal F, Ozbolat IT. Aspiration-assisted bioprinting of the osteochondral interface. Sci Rep 2020; 10:13148. [PMID: 32753630 PMCID: PMC7403300 DOI: 10.1038/s41598-020-69960-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/22/2020] [Indexed: 11/09/2022] Open
Abstract
Osteochondral defects contain damage to both the articular cartilage and underlying subchon- dral bone, which remains a significant challenge in orthopedic surgery. Layered structure of bone, cartilage and the bone-cartilage interface must be taken into account in the case of biofabrication of the osteochondral (OC) interface. In this study, a dual layered OC interface was bioprinted using a newly developed aspiration-assisted bioprinting (AAB) technique, which has been the first time that scaffold-free bioprinting was applied to OC interface engineering. Tissue spheroids, made of human adipose-derived stem cells (ADSCs), were differentiated in three dimensions (3D) into chondrogenic and osteogenic spheroids, which were confirmed by immunostaining and histology qualitatively, and biochemistry assays and gene expression, quantitatively. Remarkably, the OC interface was bioprinted by accurate positioning of a layer of osteogenic spheroids onto a sacrificial alginate support followed by another layer of chondrogenic spheroids overlaid by the same support. Spheroids in individual zones fused and the maintenance of phenotypes in both zones confirmed the successful biofabrication of the histomorphologically-relevant OC interface. The biofabrication of OC tissue model without the use of polymeric scaffolds unveils great potential not only in regenerative medicine but also in drug testing and disease modeling for osteoarthritis.
Collapse
Affiliation(s)
- Bugra Ayan
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
| | - Yang Wu
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Vengadeshprabhu Karuppagounder
- Center for Orthopedic Research and Translational Sciences, Department of Orthopedics and Re-Habilitation, Penn State University, Hershey, PA, 17033, USA
| | - Fadia Kamal
- Center for Orthopedic Research and Translational Sciences, Department of Orthopedics and Re-Habilitation, Penn State University, Hershey, PA, 17033, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA.
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA.
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, USA.
- Materials Research Institute, Penn State University, University Park, PA, 16802, USA.
- Department of Neurosurgery, Penn State University, Hershey, PA, 17033, USA.
| |
Collapse
|
45
|
Robust phenotypic maintenance of limb cells during heterogeneous culture in a physiologically relevant polymeric-based constructed graft system. Sci Rep 2020; 10:11739. [PMID: 32678185 PMCID: PMC7367281 DOI: 10.1038/s41598-020-68658-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/29/2020] [Indexed: 11/08/2022] Open
Abstract
A major challenge during the simultaneous regeneration of multiple tissues is the ability to maintain the phenotypic characteristics of distinct cell populations on one construct, especially in the presence of different exogenous soluble cues such as growth factors. Therefore, in this study, we questioned whether phenotypic maintenance over a distinct population of cells can be achieved by providing biomimetic structural cues relevant to each cell phenotype into the construct's design and controlling the presentation of growth factors in a region-specific manner. To address this question, we developed a polymeric-based constructed graft system (CGS) as a physiologically relevant model that consists of three combined regions with distinct microstructures and growth factor types. Regions A and B of the CGS exhibited similar microstructures to the skin and soft tissues and contained rhPDGF-BB and rhIGF-I, while region C exhibited a similar microstructure to the bone tissue and contained rhBMP-2. Primary rat skin fibroblasts, soft tissue fibroblasts, and osteoblasts were then cultured on regions A, B, and C of the CGS, respectively and their phenotypic characteristics were evaluated in this heterogenous environment. In the absence of growth factors, we found that the structural cues presented in every region played a key role in maintaining the region-specific cell functions and heterogeneity during a heterogeneous culture. In the presence of growth factors, we found that spatially localizing the growth factors at their respective regions resulted in enhanced region-specific cell functions and maintained region-specific cell heterogeneity compared to supplementation, which resulted in a significant reduction of cell growth and loss of phenotype. Our data suggest that providing biomimetic structural cues relevant to each cell phenotype and controlling the presentation of growth factors play a crucial role in ensuring heterogeneity maintenance of distinct cell populations during a heterogeneous culture. The presented CGS herein provides a reliable platform for investigating different cells responses to heterogeneous culture in a physiologically relevant microenvironment. In addition, the model provides a unique platform for evaluating the feasibility and efficacy of different approaches for simultaneously delivering multiple growth factors or molecules from a single construct to achieve enhanced cell response while maintaining cellular heterogeneity during a heterogenous culture.
Collapse
|
46
|
Zhang Y, Luo G, Yu X. Cellular Communication in Bone Homeostasis and the Related Anti-osteoporotic Drug Development. Curr Med Chem 2020; 27:1151-1169. [PMID: 30068268 DOI: 10.2174/0929867325666180801145614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/28/2018] [Accepted: 07/19/2018] [Indexed: 02/08/2023]
Abstract
Background:Intercellular crosstalk among osteoblast, osteoclast, osteocyte and chondrocyte is involved in the precise control of bone homeostasis. Disruption of this cellular and molecular signaling would lead to metabolic bone diseases such as osteoporosis. Currently a number of anti-osteoporosis interventions are restricted by side effects, complications and long-term intolerance. This review aims to summarize the bone cellular communication involved in bone remodeling and its usage to develop new drugs for osteoporosis. Methods:We searched PubMed for publications from 1 January 1980 to 1 January 2018 to identify relevant and latest literatures, evaluation and prospect of osteoporosis medication were summarized. Detailed search terms were 'osteoporosis', 'osteocyte', 'osteoblast', 'osteoclast', 'bone remodeling', 'chondrocyte', 'osteoporosis treatment', 'osteoporosis therapy', 'bisphosphonates', 'denosumab', 'Selective Estrogen Receptor Modulator (SERM)', 'PTH', 'romosozumab', 'dkk-1 antagonist', 'strontium ranelate'. Results:A total of 170 papers were included in the review. About 80 papers described bone cell interactions involved in bone remodeling. The remaining papers were focused on the novel advanced and new horizons in osteoporosis therapies. Conclusion:There exists a complex signal network among bone cells involved in bone remodeling. The disorder of cell-cell communications may be the underlying mechanism of osteoporosis. Current anti-osteoporosis therapies are effective but accompanied by certain drawbacks simultaneously. Restoring the abnormal signal network and individualized therapy are critical for ideal drug development.
Collapse
Affiliation(s)
- Yi Zhang
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guojing Luo
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
47
|
Zhou X, Cao H, Yuan Y, Wu W. Biochemical Signals Mediate the Crosstalk between Cartilage and Bone in Osteoarthritis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5720360. [PMID: 32337258 PMCID: PMC7165323 DOI: 10.1155/2020/5720360] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/30/2019] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Osteochondral junction is a functional unit comprising the articular cartilage, calcified cartilage, and subchondral bone. Alteration in any component of this composite unit can disrupt the joint integrity and function directly or indirectly. Biochemical signals mediate the crosstalk between tissues and play an essential role in the initiation and progression of osteoarthritis. As osteoarthritis progresses, abnormal subchondral bone remodelling leads to increased angiogenesis and porosity of the subchondral bone plate, which further triggers biochemical signals to mediate the crosstalk between cartilage and bone, contributing to the progression of osteoarthritis. Notably, common biochemical signals include the TGF-β/Smad, Wnt/β-catenin, RANK/RANKL/OPG, and MAPK pathways. This biomarker crosstalk network is the basis of osteoarthritis pathogenesis, and some of their key regulators may be potential therapeutic targets for osteoarthritis drug therapy. This review summarised the biochemical crosstalk between cartilage and bone in the pathogenesis of osteoarthritis, which may provide the basis for the discovery of osteoarthritis treatment targets.
Collapse
Affiliation(s)
- Xuchang Zhou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Hong Cao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yu Yuan
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Wei Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
48
|
Li L, Yang L, Zhang K, Zhu L, Wang X, Jiang Q. Three-dimensional finite-element analysis of aggravating medial meniscus tears on knee osteoarthritis. J Orthop Translat 2020; 20:47-55. [PMID: 31908933 PMCID: PMC6939112 DOI: 10.1016/j.jot.2019.06.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/11/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The biomechanical change during the medial meniscus damage in the process of knee osteoarthritis has not been explored. The purpose of this study was to determine the effect of aggravating medial meniscus degenerative tear on the progress of knee osteoarthritis through the finite-element simulation method. METHODS The three-dimensional digital model of a total-knee joint was obtained using a combination of magnetic resonance imaging and computed tomography images. Four types of medial meniscus tears were created to represent the aggravating degenerative meniscus lesions. Meniscectomy of each meniscal tear was also utilized in the simulation. The compression and shear stress of bony tissue, cartilage, and meniscus were evaluated, and meniscus extrusion of the healthy knee, postinjured knee, and postmeniscectomy knee were investigated under the posture of balanced standing. RESULTS Based on the results of finite-element simulation, the peak shear principal stress, peak compression principal stress, and meniscus extrusion increased gradually as the meniscus tears' region enlarged progressively (from 7.333 MPa to 15.14 MPa on medial femur and from 6 MPa to 20.94 MPa on medial tibia). The higher stress and larger meniscus extrusion displacement in all tests were observed in the flap and complex tears. The oblique tears also had a biomechanical variation of stress and meniscus extrusion in the knee joint, but their level was milder. Both the peak value of the stress and meniscus displacement increased after the meniscectomy. CONCLUSION In contrast to the damaged hemijoint, the stress applied on the healthy lateral hemijoint increased. The change of biomechanics was more obvious with the aggravation of meniscus injury. The advanced degenerative damage resulted in increasing stress that was more likely to cause symptomatic clinical manifestation in the knee joint and accelerate the progress of osteoarthritis. Moreover, we found that the meniscus injury caused higher stress concentration on the contralateral side of the joint. We also discovered that the meniscectomy can lead to more serious biomechanical changes, and although this technique can relieve pain over a period of time, it increased the risk of osteoarthritis (OA) occurrence. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE It is clear that the meniscal lesions can cause osteoarthritic knee, but the biomechanical change during the meniscus damage period has not been explored. We have evaluated the variation of stress during the aggravating medial degenerative meniscus tears and the relationship in the process of knee OA through finite-element simulation. This study does favour to obtain a better understanding on the symptoms and pathological changes of OA. It also may provide some potential directions for the prophylaxis and treatment of OA.
Collapse
Affiliation(s)
- Lan Li
- School of Mechanical Engineering, Southeast University, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, China
| | - Longfei Yang
- School of Mechanical Engineering, Southeast University, China
| | - Kaijia Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, China
| | - Liya Zhu
- School of Electrical and Automation Engineering, Nanjing Normal University, China
| | - Xingsong Wang
- School of Mechanical Engineering, Southeast University, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital Affiliated to Medical School of Nanjing University, China
| |
Collapse
|
49
|
He S, Ruan D, Chen Y, Ran J, Chen X, Yin Z, Tang C, Huang J, Heng BC, Chen J, Chen W, Shen W, Ouyang H. Characterization and Comparison of Postnatal Rat Meniscus Stem Cells at Different Developmental Stages. Stem Cells Transl Med 2019; 8:1318-1329. [PMID: 31638337 PMCID: PMC6877772 DOI: 10.1002/sctm.19-0125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/24/2019] [Indexed: 11/25/2022] Open
Abstract
Meniscus‐derived stem cells (MeSCs) are a potential cell source for meniscus tissue engineering. The stark morphological and structural changes of meniscus tissue during development indicate the complexity of MeSCs at different tissue regions and stages of development. In this study, we characterized and compared postnatal rat meniscus tissue and MeSCs at different tissue regions and stages of development. We observed that the rat meniscus tissue exhibited marked changes in tissue morphology during development, with day 7 being the most representative time point of different developmental stages. All rat MeSCs displayed typical stem cell characteristics. Rat MeSCs derived from day 7 inner meniscus tissue exhibited the highest self‐renewal capacity, cell proliferation, differentiation potential toward various mesenchymal lineage and the highest expression levels of chondrogenic genes and proteins. Transplantation of rat MeSCs derived from day 7 inner meniscus tissue promoted neo‐tissue formation and effectively protected joint surface cartilage in vivo. Our results demonstrated for the first time that rat MeSCs are not necessarily better at earlier developmental stages, and that rat MeSCs derived from day 7 inner meniscus tissue may be a superior cell source for effective meniscus regeneration and articular cartilage protection. This information could make a significant contribution to human meniscus tissue engineering in the future. stem cells translational medicine2019;8:1318&1329 (A): Meniscus tissue at different tissue regions and stages of development. (B): MeSCs at different tissue regions and stages of development. (C): Intra‐articular injection of MeSCs for meniscus regeneration and OA suppression. *Significant difference between two groups at p < .05. **Significant difference between two groups at p < .01. ***Significant difference between two groups at p < .001. ****Significant difference between two groups at p < .0001. N.S., No significant difference between two groups at p ≥ .05.![]()
Collapse
Affiliation(s)
- Shaoqi He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Orthopedic Surgery, Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yangwu Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jisheng Ran
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xiao Chen
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zi Yin
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jiayun Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Boon Chin Heng
- Peking University School of Stomatology, Beijing, People's Republic of China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Orthopedics, Research Institute of Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| | - Hongwei Ouyang
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| |
Collapse
|
50
|
Synovium-derived stromal cell-induced osteoclastogenesis: a potential osteoarthritis trigger. Clin Sci (Lond) 2019; 133:1813-1824. [DOI: 10.1042/cs20190169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/06/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022]
Abstract
Abstract
Purpose: To shed light on the idea that mesenchymal stem/stromal cells (MSCs) recruited in synovium (SM) (i.e. Synovium-Derived Stromal Cells, SDSCs) could be involved in Osteoarthritis (OA) pathophysiology. Attention was also paid to a further stromal cell type with a peculiar ultrastructure called telocytes (TCs), whose role is far from clarified. Methods: In the present in vitro study, we compared SDSCs isolated from healthy and OA subjects in terms of phenotype, morphology and differentiation potential as well as in their capability to activate normal Peripheral Blood Mononuclear Cells (PBMCs). Histological, immunohistochemical and ultrastructural analyses were integrated by qRT-PCR and functional resorbing assays. Results: Our data demonstrated that both SDSC populations stimulated the formation of osteoclasts from PBMCs: the osteoclast-like cells generated by healthy-SDSCs via transwell co-cultures were inactive, while OA-derived SDSCs have a much greater effectiveness. Moreover, the presence of TCs was more evident in cultures obtained from OA subjects and suggests a possible involvement of these cells in OA. Conclusions: Osteoclastogenic differentiation capability of PBMCs from OA subjects, also induced by B synoviocytes has been already documented. Here we hypothesized that SDSCs, generally considered for their regenerative potential in cartilage lesions, have also a role in the onset/maintenance of OA. Clinical relevance: Our observations may represent an interesting opportunity for the development of a holistic approach for OA treatment, that considers the multifaceted capability of MSCs in relation to the environment.
Collapse
|