1
|
Chen W, Wang Q, Tao H, Lu L, Zhou J, Wang Q, Huang W, Yang X. Subchondral osteoclasts and osteoarthritis: new insights and potential therapeutic avenues. Acta Biochim Biophys Sin (Shanghai) 2024; 56:499-512. [PMID: 38439665 DOI: 10.3724/abbs.2024017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, and good therapeutic results are often difficult to obtain due to its complex pathogenesis and diverse causative factors. After decades of research and exploration of OA, it has been progressively found that subchondral bone is essential for its pathogenesis, and pathological changes in subchondral bone can be observed even before cartilage lesions develop. Osteoclasts, the main cells regulating bone resorption, play a crucial role in the pathogenesis of subchondral bone. Subchondral osteoclasts regulate the homeostasis of subchondral bone through the secretion of degradative enzymes, immunomodulation, and cell signaling pathways. In OA, osteoclasts are overactivated by autophagy, ncRNAs, and Rankl/Rank/OPG signaling pathways. Excessive bone resorption disrupts the balance of bone remodeling, leading to increased subchondral bone loss, decreased bone mineral density and consequent structural damage to articular cartilage and joint pain. With increased understanding of bone biology and targeted therapies, researchers have found that the activity and function of subchondral osteoclasts are affected by multiple pathways. In this review, we summarize the roles and mechanisms of subchondral osteoclasts in OA, enumerate the latest advances in subchondral osteoclast-targeted therapy for OA, and look forward to the future trends of subchondral osteoclast-targeted therapies in clinical applications to fill the gaps in the current knowledge of OA treatment and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Wenlong Chen
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiufei Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiang Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Wei Huang
- Department of Orthopaedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| |
Collapse
|
2
|
Zhang L, Lin Y, Lu AX, Liu JX, Li J, Yan CH. Metabolomics insights into the effects of pre-pregnancy lead exposure on bone metabolism in pregnant rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122468. [PMID: 37652228 DOI: 10.1016/j.envpol.2023.122468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/07/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Today's women of childbearing age with a history of high lead (Pb) exposure in childhood have large Pb body burdens, which increases Pb release during pregnancy by promoting bone Pb mobilisation. The purpose of this study was to investigate the metabolic mechanisms underlying bone Pb mobilisation and explore the bone metabolism-related pathways during pregnancy. Drinking water containing 0.05% sodium acetate or Pb acetate was provided to weaned female rats for 4 weeks followed by a 4-week washout period, and then rats were co-caged with healthy males of the same age until pregnancy. Blood and bone tissues of the female rats were collected at gestational day (GD) 3 (early pregnancy), GD 10 (middle pregnancy), and GD 17 (late pregnancy), respectively. Pb and calcium concentrations, biomarkers for bone turnover, bone microstructure, serum metabolomics, and metabolic indicators were intensively analyzed. The results demonstrated that pre-pregnancy Pb exposure elevated blood lead levels (BLLs) at GD17, accompanied by a negative correlation between BLLs and trabecular bone Pb levels. Meanwhile, Pb-exposed rats had low bone mass and aberrant bone architecture with a larger number of mature osteoclasts (OCs) compared to the control group. Moreover, the metabolomics uncovered that Pb exposure caused mitochondrial dysfunction, such as enhanced oxidative stress and inflammatory response, and suppressed energy metabolism. Additionally, the levels of ROS, MDA, IL-1β, and IL-18 involved in redox and inflammatory pathways of bone tissues were significantly increased in the Pb-exposed group, while antioxidant SOD and energy metabolism-related indicators including ATP levels, Na+-K+-ATPase, and Ca2+-Mg2+-ATPase activities were significantly decreased. In conclusion, pre-pregnancy Pb exposure promotes bone Pb mobilisation and affects bone microstructure in the third trimester of pregnancy, which may be attributed to OC activation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Lin Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yin Lin
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - An-Xin Lu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jun-Xia Liu
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jing Li
- School of Public Health, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Chong-Huai Yan
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW To assess the present status of gene therapy for osteoarthritis (OA). RECENT FINDINGS An expanding list of cDNAs show therapeutic activity when introduced into the joints of animals with experimental models of OA. In vivo delivery with adenovirus or adeno-associated virus is most commonly used for this purpose. The list of encoded products includes cytokines, cytokine antagonists, enzymes, enzyme inhibitors, growth factors and noncoding RNA. Elements of CRISPR-Cas have also been delivered to mouse knees to ablate key genes. Several human trials have been initiated, using transgenes encoding transforming growth factor-β1, interleukin-1 receptor antagonist, interferon-β, the NKX3.2 transcription factor or variant interleukin-10. The first of these, using ex vivo delivery with allogeneic chondrocytes, gained approval in Korea which was subsequently retracted. However, it is undergoing Phase III clinical trials in the United States. The other trials are in Phase I or II. No gene therapy for OA has current marketing approval in any jurisdiction. SUMMARY Extensive preclinical data support the use of intra-articular gene therapy for treating OA. Translation is beginning to accelerate and six gene therapeutics are in clinical trials. Importantly, venture capital has begun to flow and at least seven companies are developing products. Significant progress in the future can be expected.
Collapse
|
4
|
Lu B, Liu Z, Kong F, An H, Hou X, Fan G, Dong W. A bibliometrics analysis and visualization of osteoimmunology on osteoarthritis studies. Am J Transl Res 2022; 14:4260-4277. [PMID: 35836890 PMCID: PMC9274596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Osteoarthritis (OA), the most prevalent form of arthritis, which affects up to 15% of the adult population. The work presented in this paper focuses on the analysis of the publications on osteoimmunology of OA. The purpose of this paper is to provide inspiration for future research on osteoimmunology of OA. METHODS We extracted all of the English publications relevant to osteoimmunology of OA published from 1991-2020 from the Web of Science. SPSS, GraphPad Prism, Citespace, and VOSviewer were utilized to collect and analyze the publication trends in osteoimmunology of OA. RESULTS We identified a total number of 1,004 publications with a total number of citations of 35,675 by October 31, 2020. Most publications came from China (26.8%). The United States ranked second, but its average number of citations and H index ranked best (10,130 citations and 55 H-index). Shandong University is the main center of institutional cooperation. Most papers related to osteoimmunology of OA were published in the journal of Osteoarthritis and Cartilage. In this field, TAK PP published the most papers (15), while Kotake, S's article was cited the most frequently (1,195). As presented in Figure 6, the 70 keywords, defined as terms that appeared more than 50 times in all papers, were classified into four clusters molecular research, human research, animal research and cell research. Research on cytokines and pathways is the new trend in Molecular research. CONCLUSION The current status and global trend of osteoimmunology of OA revealed by this study indicates that there is a strong possibility that the number of papers will increase in the coming year, and the research on cytokines as well as pathways would be the next hot topic.
Collapse
Affiliation(s)
- Bo Lu
- Department of Orthopaedics, Hebei Petrochina Central Hospital Langfang 065000, Hebei, China
| | - Zhenteng Liu
- Department of Orthopaedics, Hebei Petrochina Central Hospital Langfang 065000, Hebei, China
| | - Fanlin Kong
- Department of Orthopaedics, Hebei Petrochina Central Hospital Langfang 065000, Hebei, China
| | - Hepeng An
- Department of Orthopaedics, Hebei Petrochina Central Hospital Langfang 065000, Hebei, China
| | - Xueling Hou
- Department of Orthopaedics, Hebei Petrochina Central Hospital Langfang 065000, Hebei, China
| | - Guofeng Fan
- Department of Orthopaedics, Hebei Petrochina Central Hospital Langfang 065000, Hebei, China
| | - Wei Dong
- Department of Orthopaedics, Hebei Petrochina Central Hospital Langfang 065000, Hebei, China
| |
Collapse
|
5
|
Wang R, Xu B. TGF-β1-modified MSC-derived exosomal miR-135b attenuates cartilage injury via promoting M2 synovial macrophage polarization by targeting MAPK6. Cell Tissue Res 2021; 384:113-127. [PMID: 33404840 DOI: 10.1007/s00441-020-03319-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022]
Abstract
Osteoarthritis (OA) is the most common joint disease with an unsatisfactory therapy outcome and characterized by the degradation of articular cartilage and synovial inflammation. Here, we isolated bone marrow mesenchymal stem cells (BMSCs) from rat's bone marrow and BMSC-derived exosome (BMSCs-Exo) from BMSCs successfully. MiR-135b was proved to be highly expressed in TGF-β1-stimulated BMSC-derived exosomes (BMSCs-ExoTGF-β1). Then, our results demonstrated that BMSCs-ExoTGF-β1 reduced OA-induced upregulation of pro-inflammatory factors in rat's serum and damage in cartilage tissues, which was then reversed by miR-135b decreasing. Subsequently, we found that the OA-resulted M1 polarization of synovial macrophages (SMs) was repressed by BMSCs-ExoTGF-β1, this effect of BMSCs-ExoTGF-β1 was limited by miR-135b decreasing. We also proved that M2 polarization of SMs can be induced by miR-135b mimics. Furthermore, we found that the promotory effect of miR-135b and BMSCs-ExoTGF-β1 on M2 SMs polarization was reversed by increasing of MAPK6. Overall, our data showed that BMSCs-ExoTGF-β1 attenuated cartilage damage in OA rats through carrying highly expressed miR-135b. Mechanistically, miR-135b promoted M2 polarization of SMs through targeting MAPK6, thus improving cartilage damage. Our study provided a novel regulatory mechanism of BMSCs-Exo in OA development and revealed a new potential treatment target of OA.
Collapse
Affiliation(s)
- Rui Wang
- Department of Sports Trauma & Arthroscopy, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui, 230022, People's Republic of China
| | - Bin Xu
- Department of Sports Trauma & Arthroscopy, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
6
|
Abstract
Chemokines are a family of small proteins, subdivided by their conserved cysteine residues and common structural features. Chemokines interact with their cognate G-protein-coupled receptors to elicit downstream signals that result in cell migration, proliferation, and survival. This review presents evidence for how the various CXC and CC subfamily chemokines influence bone hemostasis by acting on osteoclasts, osteoblasts, and progenitor cells. Also discussed are the ways in which chemokines contribute to bone loss as a result of inflammatory diseases such as rheumatoid arthritis, HIV infection, and periodontal infection. Both positive and negative effects of chemokines on bone formation and bone loss are presented. In addition, the role of chemokines in altering the bone microenvironment through effects on angiogenesis and tumor invasion is discussed. Very few therapeutic agents that influence bone formation by targeting chemokines or chemokine receptors are available, although a few are currently being evaluated.
Collapse
Affiliation(s)
- Annette Gilchrist
- Department of Pharmaceutical Sciences, Midwestern University, Downers Grove, IL, USA.
| |
Collapse
|
7
|
Scanzello CR. Chemokines and inflammation in osteoarthritis: Insights from patients and animal models. J Orthop Res 2017; 35:735-739. [PMID: 27808445 PMCID: PMC5912941 DOI: 10.1002/jor.23471] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/28/2016] [Indexed: 02/04/2023]
Abstract
Evidence has been building that the pathologic drive for development of osteoarthritis (OA) involves more than simple mechanical "wear and tear." Inflammatory mechanisms play an important role in the tissue response to joint injury, and are involved in development of post-traumatic OA. Inflammation also appears integral to the progression of OA, whether post-traumatic or spontaneous, contributing to the evolution of joint tissue degradation and remodeling as well as joint pain. Both patient-based studies and in vivo models of disease have shed light on a number of inflammatory pathways and mediators that impact various aspects of this disease, both structurally and symptomatically. Recent work in this field has implicated inflammatory chemokines in osteoarthritis pathogenesis. Expression of multiple chemokines and their receptors is modulated during disease in both patients and animal models. Although best known for their effects on leukocyte migration and trafficking within the immune system, chemokines can have a wide variety of effects on both motile and non-motile cell types, impacting proliferation, differentiation, and activation of cellular responses. Their role in OA models has also demonstrated diverse effects on disease that exemplify their wide-ranging effects. Understanding how these important mediators of inflammation impact joint disease, and whether they can be targeted therapeutically, is actively being investigated by many groups in this field. This narrative review focuses on evidence published within the last 5 years highlighting chemokine-mediated pathways with mechanistic involvement in osteoarthritis and joint tissue repair. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:735-739, 2017.
Collapse
Affiliation(s)
- Carla R. Scanzello
- Corporal Michael J. Cresenz VA Medical Center, Division of Rheumatology and Translational Musculoskeletal Research Center, and University of Pennsylvania Perelman School of Medicine, Division of Rheumatology, 3900 Woodland Ave. Bldg. 21, Rm A213, Philadelphia, Pennsylvania 19104
| |
Collapse
|
8
|
Blaker CL, Clarke EC, Little CB. Using mouse models to investigate the pathophysiology, treatment, and prevention of post-traumatic osteoarthritis. J Orthop Res 2017; 35:424-439. [PMID: 27312470 DOI: 10.1002/jor.23343] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/14/2016] [Indexed: 02/04/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) is defined by its development after joint injury. Factors contributing to the risk of PTOA occurring, the rate of progression, and degree of associated disability in any individual, remain incompletely understood. What constitutes an "OA-inducing injury" is not defined. In line with advances in the traumatic brain injury field, we propose the scope of PTOA-inducing injuries be expanded to include not only those causing immediate structural damage and instability (Type I), but also those without initial instability/damage from moderate (Type II) or minor (Type III) loading severity. A review of the literature revealed this full spectrum of potential PTOA subtypes can be modeled in mice, with 27 Type I, 6 Type II, and 4 Type III models identified. Despite limitations due to cartilage anatomy, joint size, and bio-fluid availability, mice offer advantages as preclinical models to study PTOA, particularly genetically modified strains. Histopathology was the most common disease outcome, cartilage more frequently studied than bone or synovium, and meniscus and ligaments rarely evaluated. Other methods used to examine PTOA included gene expression, protein analysis, and imaging. Despite the major issues reported by patients being pain and biomechanical dysfunction, these were the least commonly measured outcomes in mouse models. Informative correlations of simultaneously measured disease outcomes in individual animals, was rarely done in any mouse PTOA model. This review has identified knowledge gaps that need to be addressed to increase understanding and improve prevention and management of PTOA. Preclinical mouse models play a critical role in these endeavors. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:424-439, 2017.
Collapse
Affiliation(s)
- Carina L Blaker
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Level 10, Kolling Institute B6, Northern Sydney Local Health District, Sydney Medical School Northern, University of Sydney, The Royal North Shore Hospital, St. Leonards, New South Wales, 2065, Australia.,Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Sydney Medical School Northern, University of Sydney, St. Leonards, New South Wales, 2065, Australia
| | - Elizabeth C Clarke
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Level 10, Kolling Institute B6, Northern Sydney Local Health District, Sydney Medical School Northern, University of Sydney, The Royal North Shore Hospital, St. Leonards, New South Wales, 2065, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Sydney Medical School Northern, University of Sydney, St. Leonards, New South Wales, 2065, Australia
| |
Collapse
|
9
|
Yan S, Wang M, Zhao J, Zhang H, Zhou C, Jin L, Zhang Y, Qiu X, Ma B, Fan Q. MicroRNA-34a affects chondrocyte apoptosis and proliferation by targeting the SIRT1/p53 signaling pathway during the pathogenesis of osteoarthritis. Int J Mol Med 2016; 38:201-9. [PMID: 27247228 PMCID: PMC4899008 DOI: 10.3892/ijmm.2016.2618] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 05/16/2016] [Indexed: 01/07/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease with multifactorial etiology caused by risk factors such as ageing, obesity and trauma. Previously, it was reported that the inhibition of microRNA-34a (miR-34a) may reduce rat chondrocyte apoptosis induced by IL-1β, whereas the molecular mechanism and the role of miR-34a in human chondrocyte as well as in OA progression remains to be determined. In the current study, using MTT, luciferase reporter assays and western blot analysis we identified that miR-34a was upregulated while silent information regulator 1 (SIRT1) was inhibited in chondrocytes from 12 OA patients compared with healthy chondrocytes from 10 trauma amputees. Overexpression of miR-34a promoted apoptosis and inhibited cell proliferation in human chondrocytes. Transfection with miR-34a mimic inhibited SIRT1 expression, which attenuated the deacetylation of p53, leading to the upregulation of Bax and downregulation of Bcl-2. Furthermore, results from the western blot analysis and luciferase reporter assay demonstrated that SIRT1 was directly regulated by miR-34a in human chondrocytes. A rat model of OA was induced through anterior cruciate ligament transection and medial meniscus resection (ACLT+MMx). The results showed that the intra-articular injection of lentiviral vector encoding anti-miR-34a sequence effectively ameliorated the progression of OA. The results suggest that miR-34a has a crucial role in the pathogenesis of OA through direct regulation of the SIRT1/p53 signaling pathway and serves as a potential therapeutic target of OA.
Collapse
Affiliation(s)
- Shiju Yan
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Meng Wang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Jian Zhao
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Hongtao Zhang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Chengpei Zhou
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Lei Jin
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yinglong Zhang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xiuchun Qiu
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Baoan Ma
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Qingyu Fan
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
10
|
The Estrogen Receptor-β Expression in De Quervain's Disease. Int J Mol Sci 2015; 16:26452-62. [PMID: 26556342 PMCID: PMC4661827 DOI: 10.3390/ijms161125968] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/22/2015] [Accepted: 10/29/2015] [Indexed: 11/30/2022] Open
Abstract
Stenosing tenosynovitis of the first dorsal compartment of the wrist (a.k.a. de Quervain’s disease) is common but how estrogen is involved is still unknown. We previously reported that inflammation was involved in the pathogenesis of this ailment. In the present study, we extended our investigation of estrogen receptor (ER)-β expression to determine whether estrogen is involved in the pathogenesis of de Quervain’s. Intraoperative retinaculum samples were collected from 16 patients with the ailment. Specimens were histologically graded by collagen structure and immunohistochemically evaluated by quantifying the expression of ER-β, interleukin (IL)-1β and IL-6 (inflammatory cytokines), cyclooxygenase (COX)-2 (an inflammatory enzyme), and vascular endothelial growth factor (VEGF), and Von Willebrand’s factor (vWF). De Quervain’s occurs primarily in women. The female:male ratio in our study was 7:1. We found that ER-β expression in the retinaculum was positively correlated with disease grade and patient age. Additionally, disease severity was associated with inflammatory factors—IL-1β and IL-6, COX-2, and VEGF and vWF in tenosynovial tissue. The greater the levels of ER-β expression, tissue inflammation, and angiogenesis are, the more severe de Quervain’s disease is. ER-β might be a useful target for novel de Quervain’s disease therapy.
Collapse
|
11
|
Inflammation in joint injury and post-traumatic osteoarthritis. Osteoarthritis Cartilage 2015; 23:1825-34. [PMID: 26521728 PMCID: PMC4630675 DOI: 10.1016/j.joca.2015.08.015] [Citation(s) in RCA: 307] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/24/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023]
Abstract
Inflammation is a variable feature of osteoarthritis (OA), associated with joint symptoms and progression of disease. Signs of inflammation can be observed in joint fluids and tissues from patients with joint injuries at risk for development of post-traumatic osteoarthritis (PTOA). Furthermore, inflammatory mechanisms are hypothesized to contribute to the risk of OA development and progression after injury. Animal models of PTOA have been instrumental in understanding factors and mechanisms involved in chronic progressive cartilage degradation observed after a predisposing injury. Specific aspects of inflammation observed in humans, including cytokine and chemokine production, synovial reaction, cellular infiltration and inflammatory pathway activation, are also observed in models of PTOA. Many of these models are now being utilized to understand the impact of post-injury inflammatory response on PTOA development and progression, including risk of progressive cartilage degeneration and development of chronic symptoms post-injury. As evidenced from these models, a vigorous inflammatory response occurs very early after joint injury but is then sustained at a lower level at the later phases. This early inflammatory response contributes to the development of PTOA features including cartilage erosion and is potentially modifiable, but specific mediators may also play a role in tissue repair. Although the optimal approach and timing of anti-inflammatory interventions after joint injury are yet to be determined, this body of work should provide hope for the future of disease modification tin PTOA.
Collapse
|
12
|
Jackson MT, Moradi B, Zaki S, Smith MM, McCracken S, Smith SM, Jackson CJ, Little CB. Depletion of protease-activated receptor 2 but not protease-activated receptor 1 may confer protection against osteoarthritis in mice through extracartilaginous mechanisms. Arthritis Rheumatol 2015; 66:3337-48. [PMID: 25200274 DOI: 10.1002/art.38876] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 09/04/2014] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To explore the involvement of protease-activated receptor 1 (PAR-1) and PAR-2 in the pathologic processes of osteoarthritis (OA) and to identify the cells/tissues primarily affected by ablation of PAR-1 or PAR-2 in mice. METHODS OA was induced in the joints of wild-type (WT), PAR-1(+/+) , PAR-1(-/-) , and PAR-2(-/-) mice by destabilization of the medial meniscus (DMM), and scores of histologic features (cartilage aggrecan loss and erosion, subchondral bone sclerosis, osteophytes, and synovitis) were compared at 1, 4, and 8 weeks post-DMM. The effects of PAR ablation on cartilage degradation and chondrocyte metalloproteinase expression/activity were studied in cultures of mouse femoral head tissue with or without interleukin-1α (IL-1α). At 1 week post-DMM, synovial expression of cytokines and metalloproteinase genes was measured by reverse transcription-polymerase chain reaction, and populations of inflammatory cells were quantified by flow cytometry. RESULTS Deletion of PAR-2, but not that of PAR-1, in mice significantly delayed the progression of cartilage damage and inhibited subchondral bone sclerosis following DMM. There was no inhibitory effect of PAR-1 or PAR-2 ablation on IL-1α-induced cartilage degradation or chondrocyte metalloproteinase expression/activation. A low but significant level of synovitis persisted in mice subjected to DMM compared to that in control mice subjected to sham surgery, but no differences between the genotypes were seen 4 or 8 weeks post-DMM. One week after DMM, increased synovial expression of proinflammatory cytokines and metalloproteinase genes, along with increased levels of CD4+ T cells, inflammatory monocytes, and activated macrophages, were seen in all genotypes. However, there was a significant reduction in the percentage of activated macrophages in PAR-2(-/-) mice compared to PAR-1(-/-) and WT mice. CONCLUSION Deletion of PAR-2, but not that of PAR-1, results in a significant decrease in DMM-induced cartilage damage. The chondroprotection in PAR-2(-/-) mice appears to occur indirectly through modulation of extracartilaginous events such as subchondral bone remodeling and synovial macrophage activation, rather than through alteration of chondrocyte catabolic responses.
Collapse
Affiliation(s)
- Miriam T Jackson
- Kolling Institute of Medical Research and the University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Albrecht C, Hosiner S, Tichy B, Aldrian S, Hajdu S, Nürnberger S. Comparison of Lentiviral Packaging Mixes and Producer Cell Lines for RNAi Applications. Mol Biotechnol 2015; 57:499-505. [DOI: 10.1007/s12033-015-9843-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Chen HT, Tsou HK, Chen JC, Shih JMK, Chen YJ, Tang CH. Adiponectin enhances intercellular adhesion molecule-1 expression and promotes monocyte adhesion in human synovial fibroblasts. PLoS One 2014; 9:e92741. [PMID: 24667577 PMCID: PMC3965461 DOI: 10.1371/journal.pone.0092741] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 02/24/2014] [Indexed: 12/13/2022] Open
Abstract
Adiponectin is a protein hormone secreted predominantly by differentiated adipocytes and is involved in energy homeostasis. Adiponectin expression is significantly high in the synovial fluid of patients with osteoarthritis (OA). Intercellular adhesion molecule-1 (ICAM-1) is an important adhesion molecule that mediates monocyte adhesion and infiltration during OA pathogenesis. Adiponectin-induced expression of ICAM-1 in human OA synovial fibroblasts (OASFs) was examined by using qPCR, flow cytometry and western blotting. The intracellular signaling pathways were investigated by pretreated with inhibitors or transfection with siRNA. The monocyte THP-1 cell line was used for an adhesion assay with OASFs. Stimulation of OASFs with adiponectin induced ICAM-1 expression. Pretreatment with AMP-activated protein kinase (AMPK) inhibitors (AraA and compound C) or transfection with siRNA against AMPKα1 and two AMPK upstream activator- liver kinase B1 (LKB1) and calmodulin-dependent protein kinase II (CaMKII) diminished the adiponectin-induced ICAM-1 expression. Stimulation of OASFs with adiponectin increased phosphorylation of LKB1, CaMKII, AMPK, and c-Jun, resulting in c-Jun binding to AP-1 element of ICAM-1 promoter. In addition, adiponectin-induced activation of the LKB1/CaMKII, AMPK, and AP-1 pathway increased the adhesion of monocytes to the OASF monolayer. Our results suggest that adiponectin increases ICAM-1 expression in human OASFs via the LKB1/CaMKII, AMPK, c-Jun, and AP-1 signaling pathway. Adiponectin-induced ICAM-1 expression promoted the adhesion of monocytes to human OASFs. These findings may provide a better understanding of the pathogenesis of OA and can utilize this knowledge to design a new therapeutic strategy.
Collapse
Affiliation(s)
- Hsien-Te Chen
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hsi-Kai Tsou
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Early Childhood Care and Education, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Jui-Chieh Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Zhunan, Taiwan
| | | | - Yen-Jen Chen
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|