1
|
Hu K, Wen H, Song T, Che Z, Song Y, Song M. Deciphering the Role of LncRNAs in Osteoarthritis: Inflammatory Pathways Unveiled. J Inflamm Res 2024; 17:6563-6581. [PMID: 39318993 PMCID: PMC11421445 DOI: 10.2147/jir.s489682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024] Open
Abstract
Long non-coding RNA (LncRNA), with transcripts over 200 nucleotides in length, play critical roles in numerous biological functions and have emerged as significant players in the pathogenesis of osteoarthritis (OA), an inflammatory condition traditionally viewed as a degenerative joint disease. This review comprehensively examines the influence of LncRNA on the inflammatory processes driving OA progression, focusing on their role in regulating gene expression, cellular activities, and inflammatory pathways. Notably, LncRNAs such as MALAT1, H19, and HOTAIR are upregulated in OA and exacerbate the inflammatory milieu by modulating key signaling pathways like NF-κB, TGF-β/SMAD, and Wnt/β-catenin. Conversely, LncRNA like MEG3 and GAS5, which are downregulated in OA, show potential in dampening inflammatory responses and protecting against cartilage degradation by influencing miRNA interactions and cytokine production. By enhancing our understanding of LncRNA' roles in OA inflammation, we can better leverage them as potential biomarkers for the disease and develop innovative therapeutic strategies for OA management. This paper aims to delineate the mechanisms by which LncRNA influence inflammatory responses in OA and propose them as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Kangyi Hu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Haonan Wen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Ting Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Zhixin Che
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Yongjia Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Min Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| |
Collapse
|
2
|
Xie J, Wan X, Yang M, Yu H, Hao J, Xu K, Wang J, Xu P. Circulating cytokines levels and osteoarthritis: A Mendelian randomization study. Cytokine 2024; 179:156625. [PMID: 38677184 DOI: 10.1016/j.cyto.2024.156625] [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: 11/12/2023] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Previous traditional observational studies have suggested the contribution of several cytokines and growth factors to the development of osteoarthritis (OA). This study aimed to determine the association of circulating cytokine and growth factor levels with OA. METHODS We used two-sample Mendelian randomization (MR) to explore the causality between circulating cytokine and growth factor levels and OA [including knee or hip OA (K/HOA), knee OA (KOA), and hip OA (HOA)]. Summary level data for circulating cytokine and growth factor levels were sourced from a genome-wide association study (GWAS) involving 8,293 participants of Finnish ancestry. Single-nucleotide polymorphisms related to K/HOA (39,427 cases and 378,169 controls), KOA (24,955 cases and 378,169 controls), and HOA (15,704 cases and 378,169 controls) were obtained from a previous GWAS. The inverse variance weighted (IVW) method was primarily used for our MR analysis. For exposures to only one relevant SNP as IV, we used the Wald ratio as the major method to assess causal effects. We also conducted a series of sensitivity analyses to improve the robustness of the results. RESULTS Circulating vascular endothelial growth factor levels were suggestively associated with an increased risk of K/HOA (odds ratio (OR) = 1.034; 95 % confidence interval (CI) = 1.013-1.055; P = 0.001), KOA (OR = 1.034; 95 % CI = 1.014-1.065; P = 0.002), and HOA (OR = 1.039; 95 % CI = 1.003-1.067; P = 0.034). Circulating interleukin (IL)-12p70 levels was suggestively associated with K/HOA (OR = 1.047; 95 % CI = 1.018-1.077; P = 0.001), KOA (OR = 1.058; 95 % CI = 1.022-1.095; P = 0.001), and HOA (OR = 1.044; 95 % CI = 1.000-1.091; P = 0.048). Circulating IL-18 levels were suggestively associated with HOA (OR = 1.068; 95 % CI = 1.014-1.125; P = 0.012). However, limited evidence exists to support causal genetic relationships between other circulating cytokines, growth factor levels and K/HOA, KOA, and HOA. CONCLUSIONS Our MR analysis provides suggestive evidence of causal relationships between circulating cytokines and growth factors levels and OA, providing new insights into the etiology of OA.
Collapse
Affiliation(s)
- Jiale Xie
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, China
| | - Xianjie Wan
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, China
| | - Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, China
| | - Hui Yu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, China
| | - Jinrong Hao
- Department of Endocrinology, Xi'an Central Hospital, Xi'an, Shaanxi 710003, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, China
| | - Jiachen Wang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, China.
| |
Collapse
|
3
|
Mukherjee A, Das B. The role of inflammatory mediators and matrix metalloproteinases (MMPs) in the progression of osteoarthritis. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100090. [PMID: 38440290 PMCID: PMC10910010 DOI: 10.1016/j.bbiosy.2024.100090] [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: 09/11/2023] [Revised: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is a chronic musculoskeletal disorder characterized by an imbalance between (synthesis) and catabolism (degradation) in altered homeostasis of articular cartilage mediated primarily by the innate immune system. OA degenerates the joints resulting in synovial hyperplasia, degradation of articular cartilage with damage of the structural and functional integrity of the cartilage extracellular matrix, subchondral sclerosis, osteophyte formation, and is characterized by chronic pain, stiffness, and loss of function. Inflammation triggered by factors like biomechanical stress is involved in the development of osteoarthritis. In OA apart from catabolic effects, anti-inflammatory anabolic processes also occur continually. There is also an underlying chronic inflammation present, not only in cartilage tissue but also within the synovium, which perpetuates tissue destruction of the OA joint. The consideration of inflammation in OA considers synovitis and/or other cellular and molecular events in the synovium during the progression of OA. In this review, we have presented the progression of joint degradation that results in OA. The critical role of inflammation in the pathogenesis of OA is discussed in detail along with the dysregulation within the cytokine networks composed of inflammatory and anti-inflammatory cytokines that drive catabolic pathways, inhibit matrix synthesis, and promote cellular apoptosis. OA pathogenesis, fluctuation of synovitis, and its clinical impact on disease progression are presented here along with the role of synovial macrophages in promoting inflammatory and destructive responses in OA. The role of interplay between different cytokines, structure, and function of their receptors in the inter-cellular signaling pathway is further explored. The effect of cytokines in the increased synthesis and release of matrix-decomposing proteolytic enzymes, such as matrix metalloproteinase (MMPs) and a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS), is elaborated emphasizing the potential impact of MMPs on the chondrocytes, synovial cells, articular and periarticular tissues, and other immune system cells migrating to the site of inflammation. We also shed light on the pathogenesis of OA via oxidative damage particularly due to nitric oxide (NO) via its angiogenic response to inflammation. We concluded by presenting the current knowledge about the tissue inhibitors of metalloproteinases (TIMPs). Synthetic MMP inhibitors include zinc binding group (ZBG), non-ZBG, and mechanism-based inhibitors, all of which have the potential to be therapeutically beneficial in the treatment of osteoarthritis. Improving our understanding of the signaling pathways and molecular mechanisms that regulate the MMP gene expression, may open up new avenues for the creation of therapies that can stop the joint damage associated with OA.
Collapse
Affiliation(s)
- Anwesha Mukherjee
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| |
Collapse
|
4
|
Lee KT, Lin CY, Liu SC, He XY, Tsai CH, Ko CY, Tsai YH, Chao CC, Chen PC, Tang CH. IL-17 promotes IL-18 production via the MEK/ERK/miR-4492 axis in osteoarthritis synovial fibroblasts. Aging (Albany NY) 2024; 16:1829-1844. [PMID: 38261743 PMCID: PMC10866453 DOI: 10.18632/aging.205462] [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: 08/19/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024]
Abstract
The concept of osteoarthritis (OA) as a low-grade inflammatory joint disorder has been widely accepted. Many inflammatory mediators are implicated in the pathogenesis of OA. Interleukin (IL)-18 is a pleiotropic cytokine with versatile cellular functions that are pathogenetically important in immune responses, as well as autoimmune, inflammatory, and infectious diseases. IL-17, a proinflammatory cytokine mainly secreted by Th17 cells, is upregulated in OA patients. However, the role of IL-17 in OA progression is unclear. The synovial tissues collected from healthy donors and OA patients were used to detect the expression level of IL-18 by IHC stain. The OA synovial fibroblasts (OASFs) were incubated with recombinant IL-17 and subjected to Western blot, qPCR, and ELISA to examine IL-18 expression level. The chemical inhibitors and siRNAs which targeted signal pathways were used to investigate signal pathways involved in IL-17-induced IL-18 expression. The microRNAs which participated IL-18 expression were surveyed with online databases miRWalk and miRDB, followed by validation with qPCR. This study revealed significantly higher levels of IL-18 expression in synovial tissue from OA patients compared with healthy controls, as well as increased IL-18 expression in OASFs from rats with severe OA. In vitro findings indicated that IL-17 dose-dependently promoted IL-18 production in OASFs. Molecular investigations revealed that the MEK/ERK/miR-4492 axis stimulated IL-18 production when OASFs were treated with IL-17. This study provides novel insights into the role of IL-17 in the pathogenesis of OA, which may help to inform OA treatment in the future.
Collapse
Affiliation(s)
- Kun-Tsan Lee
- Department of Post-Baccalaureate Medicine, National Chung-Hsing University, Taichung, Taiwan
- Department of Orthopedics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Yang Lin
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
| | - Shan-Chi Liu
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Xiu-Yuan He
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Chih-Yuan Ko
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Yuan-Hsin Tsai
- Department of Orthopedics, Show-Chwan Memorial Hospital, Changhua, Taiwan
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Po-Chun Chen
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| |
Collapse
|
5
|
Dias de Oliveira FB, Antonioli E, Dias OFM, de Souza JG, Agarwal S, Chudzinski-Tavassi AM, Ferretti M. Comparative Effects of Intra-Articular versus Intravenous Mesenchymal Stromal Cells Therapy in a Rat Model of Osteoarthritis by Destabilization of Medial Meniscus. Int J Mol Sci 2023; 24:15543. [PMID: 37958526 PMCID: PMC10649289 DOI: 10.3390/ijms242115543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
Transplanted mesenchymal stromal cells (MSCs) exhibit a robust anti-inflammatory and homing capacity in response to high inflammatory signals, as observed in studies focused on rheumatic diseases that target articular cartilage (AC) health. However, AC degradation in osteoarthritis (OA) does not necessarily coincide with a highly inflammatory joint profile. Often, by the time patients seek medical attention, they already have damaged AC. In this study, we examined the therapeutic potential of a single bone marrow MSC transplant (2 × 106 cells/kgbw) through two different routes: intra-articular (MSCs-IAt) and intravenous (MSCs-IVt) in a preclinical model of low-grade inflammatory OA with an established AC degeneration. OA was induced through the destabilization of the medial meniscus (DMM) in female Wistar Kyoto rats. The animals received MSCs 9 weeks after surgery and were euthanized 4 and 12 weeks post-transplant. In vivo and ex vivo tracking of MSCs were analyzed via bioluminescence and imaging flow cytometry, respectively. Cytokine/chemokine modulation in serum and synovial fluid was measured using a multiplex panel. AC degeneration was quantified through histology, and hindlimb muscle balance was assessed with precision weighing. To our knowledge, we are the first group to show the in vivo (8 h) and ex vivo (12 h) homing of cells to the DMM-OA joint following MSCs-IVt. In the case of MSCs-IAt, the detection of cellular bioluminescence at the knee joint persisted for up to 1 week. Intriguingly, intra-articular saline injection (placebo-IAt) resulted in a worse prognosis of OA when compared to a non-invasive control (placebo-IVt) without joint injection. The systemic cytokines/chemokines profile exhibited a time-dependent variation between transplant routes, displaying a transient anti-inflammatory systemic response for both MSCs-IVt and MSCs-IAt. A single injection of MSCs, whether administered via the intra-articular or intravenous route, performed 9 weeks after DMM surgery, did not effectively inhibit AC degeneration when compared to a non-invasive control.
Collapse
Affiliation(s)
| | - Eliane Antonioli
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.B.D.d.O.)
| | | | - Jean Gabriel de Souza
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA;
- CENTD Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo 05503-900, Brazil
| | - Sudha Agarwal
- Division of Rheumatology and Immunology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH 43210, USA
| | - Ana Marisa Chudzinski-Tavassi
- CENTD Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo 05503-900, Brazil
- Laboratório de Desenvolvimento e Inovação, Butantan Institute, São Paulo 05503-900, Brazil
| | - Mario Ferretti
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.B.D.d.O.)
- Departamento de Ortopedia e Traumatologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| |
Collapse
|
6
|
He S, Deng H, Li P, Hu J, Yang Y, Xu Z, Liu S, Guo W, Guo Q. Arthritic Microenvironment-Dictated Fate Decisions for Stem Cells in Cartilage Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207715. [PMID: 37518822 PMCID: PMC10520688 DOI: 10.1002/advs.202207715] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/05/2023] [Indexed: 08/01/2023]
Abstract
The microenvironment and stem cell fate guidance of post-traumatic articular cartilage regeneration is primarily the focus of cartilage tissue engineering. In articular cartilage, stem cells are characterized by overlapping lineages and uneven effectiveness. Within the first 12 weeks after trauma, the articular inflammatory microenvironment (AIME) plays a decisive role in determining the fate of stem cells and cartilage. The development of fibrocartilage and osteophyte hyperplasia is an adverse outcome of chronic inflammation, which results from an imbalance in the AIME during the cartilage tissue repair process. In this review, the sources for the different types of stem cells and their fate are summarized. The main pathophysiological events that occur within the AIME as well as their protagonists are also discussed. Additionally, regulatory strategies that may guide the fate of stem cells within the AIME are proposed. Finally, strategies that provide insight into AIME pathophysiology are discussed and the design of new materials that match the post-traumatic progress of AIME pathophysiology in a spatial and temporal manner is guided. Thus, by regulating an appropriately modified inflammatory microenvironment, efficient stem cell-mediated tissue repair may be achieved.
Collapse
Affiliation(s)
- Songlin He
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Haotian Deng
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Peiqi Li
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Jingjing Hu
- Department of GastroenterologyInstitute of GeriatricsChinese PLA General HospitalBeijing100853China
| | - Yongkang Yang
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Ziheng Xu
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Shuyun Liu
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Weimin Guo
- Department of Orthopaedic SurgeryGuangdong Provincial Key Laboratory of Orthopedics and TraumatologyFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510080China
| | - Quanyi Guo
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| |
Collapse
|
7
|
Su Y, Xing H, Kang J, Bai L, Zhang L. Role of the hedgehog signaling pathway in rheumatic diseases: An overview. Front Immunol 2022; 13:940455. [PMID: 36105801 PMCID: PMC9466598 DOI: 10.3389/fimmu.2022.940455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Hedgehog (Hh) signaling pathway is an evolutionarily conserved signal transduction pathway that plays an important regulatory role during embryonic development, cell proliferation, and differentiation of vertebrates, and it is often inhibited in adult tissues. Recent evidence has shown that Hh signaling also plays a key role in rheumatic diseases, as alterations in their number or function have been identified in rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, systemic sclerosis, and Sjogren's Syndrome. As a result, emerging studies have focused on the blockade of this pathogenic axis as a promising therapeutic target in several autoimmune disorders; nevertheless, a greater understanding of its contribution still requires further investigation. This review aims to elucidate the most recent studies and literature data on the pathogenetic role of Hh signaling in rheumatic diseases.
Collapse
Affiliation(s)
| | | | | | | | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
8
|
Chang X, Kang Y, Yang Y, Chen Y, Shen Y, Jiang C, Shen Y. Pyroptosis: A Novel Intervention Target in the Progression of Osteoarthritis. J Inflamm Res 2022; 15:3859-3871. [PMID: 35845090 PMCID: PMC9285853 DOI: 10.2147/jir.s368501] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
Osteoarthritis (OA) is one of the most common chronic joint diseases and is gradually becoming the main cause of disability and joint pain in the elderly worldwide. Pyroptosis is a regulated programmed cell death triggered by inflammasomes. It leads to cell swelling, lysis, and bioactive molecule secretion. Studies found that the damaged chondrocytes in OA joints had morphological characteristics of pyroptosis, and the cytokines associated with pyroptosis in synovial fluid increased, indicating that pyroptosis may have certain impacts on the pathological progression of OA. This review briefly summarizes the molecular mechanisms of pyroptosis and the epidemiology and pathogenesis of OA. Furthermore, we discussed the role of pyroptosis in articular cartilage and synovium during OA and reviewed the progress of pyroptosis-related molecules in the targeted therapy of OA joints, hoping to provide feasible directions for the diversified treatment of OA.
Collapse
Affiliation(s)
- Xingyu Chang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Yuchen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Yuxuan Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Yajie Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Yanyu Shen
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Chenjun Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Yi Shen
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| |
Collapse
|
9
|
Li M, Yin H, Yan Z, Li H, Wu J, Wang Y, Wei F, Tian G, Ning C, Li H, Gao C, Fu L, Jiang S, Chen M, Sui X, Liu S, Chen Z, Guo Q. The immune microenvironment in cartilage injury and repair. Acta Biomater 2022; 140:23-42. [PMID: 34896634 DOI: 10.1016/j.actbio.2021.12.006] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023]
Abstract
The ability of articular cartilage to repair itself is limited because it lacks blood vessels, nerves, and lymph tissue. Once damaged, it can lead to joint swelling and pain, accelerating the progression of osteoarthritis. To date, complete regeneration of hyaline cartilage exhibiting mechanical properties remains an elusive goal, despite the many available technologies. The inflammatory milieu created by cartilage damage is critical for chondrocyte death and hypertrophy, extracellular matrix breakdown, ectopic bone formation, and progression of cartilage injury to osteoarthritis. In the inflammatory microenvironment, mesenchymal stem cells (MSCs) undergo aberrant differentiation, and chondrocytes begin to convert or dedifferentiate into cells with a fibroblast phenotype, thereby resulting in fibrocartilage with poor mechanical qualities. All these factors suggest that inflammatory problems may be a major stumbling block to cartilage repair. To produce a milieu conducive to cartilage repair, multi-dimensional management of the joint inflammatory microenvironment in place and time is required. Therefore, this calls for elucidation of the immune microenvironment of cartilage repair after injury. This review provides a brief overview of: (1) the pathogenesis of cartilage injury; (2) immune cells in cartilage injury and repair; (3) effects of inflammatory cytokines on cartilage repair; (4) clinical strategies for treating cartilage defects; and (5) strategies for targeted immunoregulation in cartilage repair. STATEMENT OF SIGNIFICANCE: Immune response is increasingly considered the key factor affecting cartilage repair. It has both negative and positive regulatory effects on the process of regeneration and repair. Proinflammatory factors are secreted in large numbers, and necrotic cartilage is removed. During the repair period, immune cells can secrete anti-inflammatory factors and chondrogenic cytokines, which can inhibit inflammation and promote cartilage repair. However, inflammatory factors persist, which accelerate the degradation of the cartilage matrix. Furthermore, in an inflammatory microenvironment, MSCs undergo abnormal differentiation, and chondrocytes begin to transform or dedifferentiate into fibroblast-like cells, forming fibrocartilage with poor mechanical properties. Consequently, cartilage regeneration requires multi-dimensional regulation of the joint inflammatory microenvironment in space and time to make it conducive to cartilage regeneration.
Collapse
|
10
|
Liu S, Deng Z, Chen K, Jian S, Zhou F, Yang Y, Fu Z, Xie H, Xiong J, Zhu W. Cartilage tissue engineering: From proinflammatory and anti‑inflammatory cytokines to osteoarthritis treatments (Review). Mol Med Rep 2022; 25:99. [PMID: 35088882 PMCID: PMC8809050 DOI: 10.3892/mmr.2022.12615] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA), one of the most common joint diseases, is characterized by fibrosis, rhagadia, ulcers and attrition of articular cartilage due to a number of factors. The etiology of OA remains unclear, but its occurrence has been associated with age, obesity, inflammation, trauma and genetic factors. Inflammatory cytokines are crucial for the occurrence and progression of OA. The intra-articular proinflammatory and anti-inflammatory cytokines jointly maintain a dynamic balance, in accordance with the physiological metabolism of articular cartilage. However, dynamic imbalance between proinflammatory and anti-inflammatory cytokines can cause abnormal metabolism in knee articular cartilage, which leads to deformation, loss and abnormal regeneration, and ultimately destroys the normal structure of the knee joint. The ability of articular cartilage to self-repair once damaged is limited, due to its inability to obtain nutrients from blood vessels, nerves and lymphatic vessels, as well as limitations in the extracellular matrix. There are several disadvantages inherent to conventional repair methods, while cartilage tissue engineering (CTE), which combines proinflammatory and anti-inflammatory cytokines, offers a new therapeutic approach for OA. The aim of the present review was to examine the proinflammatory factors implicated in OA, including IL-1β, TNF-α, IL-6, IL-15, IL-17 and IL-18, as well as the key anti-inflammatory factors reducing OA-related articular damage, including IL-4, insulin-like growth factor and TGF-β. The predominance of proinflammatory over anti-inflammatory cytokine effects ultimately leads to the development of OA. CTE, which employs mesenchymal stem cells and scaffolding technology, may prevent OA by maintaining the homeostasis of pro- and anti-inflammatory factors.
Collapse
Affiliation(s)
- Shuyu Liu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Kang Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Shengsheng Jian
- Department of Orthopedics, Luo Hu Hospital, Shenzhen, Guangdong 518001, P.R. China
| | - Feifei Zhou
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Yuan Yang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zicai Fu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Huanyu Xie
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Jianyi Xiong
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Weimin Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
11
|
Waszczykowski M, Fabiś-Strobin A, Bednarski I, Narbutt J, Fabiś J. Serum and synovial fluid concentrations of interleukin-18 and interleukin-20 in patients with osteoarthritis of the knee and their correlation with other markers of inflammation and turnover of joint cartilage. Arch Med Sci 2022; 18:448-458. [PMID: 35316898 PMCID: PMC8924851 DOI: 10.5114/aoms.2020.96717] [Citation(s) in RCA: 4] [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: 03/30/2020] [Accepted: 05/22/2020] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is the most common degenerative joint disease, and its aetiology is not entirely known. The aim of the study was to evaluate the involvement of interleukin-18 (IL-18) and interleukin-20 (IL-20) in the pathogenesis of knee OA and their correlations with other markers of inflammation and destruction of joint cartilage, as well as clinical and radiological changes. MATERIAL AND METHODS The study included 25 patients with knee OA and a control group. The concentration of IL-18, IL-20, IL-6, MMP-1, MMP-3, COMP, PG-AG, and YKL-40 in serum and synovial fluid (SF) were determined. We also evaluated radiological lesions of the knee joint according to the Kellgren-Lawrence (K-L) scale, and clinical severity of the disease according to Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) and Lequesne Index. RESULTS The concentrations of IL-18 and IL-20 were statistically significantly higher in serum of patients with OA than in the control group (106.00 ±189.76 pg/ml vs. 16.73 ±16.99 pg/ml, p < 0.001, 17.69 ±13.45 pg/ml vs. 9.76 ±9.00 pg/ml, p < 0.014). Serum concentration of IL-18 positively correlated with MMP-3 (R = 0.58; p = 0.006) and YKL-40 (R = 0.48; p = 0.002). The degree of radiological advancement of OA (K-L scale) correlated positively with clinical evaluation (WOMAC, R = 0.74, p ≤ 0.001; Lequesne Index, R = 0.57, p = 0.003). CONCLUSIONS The analysis of ROC curves showed that IL-20 as well as COMP, MMP-3, and YKL-40 may be diagnostic markers of knee OA. The observations indicate that IL-18 potentially mediates mainly in intra-articular processes and IL-20 could be primarily responsible for the systemic inflammatory reaction.
Collapse
Affiliation(s)
- Michał Waszczykowski
- Department of Arthroscopy, Minimally Invasive Surgery, and Sports Traumatology, Medical University of Lodz, Lodz, Poland
| | - Anna Fabiś-Strobin
- Clinic of Orthopaedic and Traumatology, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Igor Bednarski
- Dermatology, Paediatric Dermatology, and Oncology Clinic, Medical University of Lodz, Lodz, Poland
| | - Joanna Narbutt
- Dermatology, Paediatric Dermatology, and Oncology Clinic, Medical University of Lodz, Lodz, Poland
| | - Jarosław Fabiś
- Department of Arthroscopy, Minimally Invasive Surgery, and Sports Traumatology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
12
|
Molnar V, Matišić V, Kodvanj I, Bjelica R, Jeleč Ž, Hudetz D, Rod E, Čukelj F, Vrdoljak T, Vidović D, Starešinić M, Sabalić S, Dobričić B, Petrović T, Antičević D, Borić I, Košir R, Zmrzljak UP, Primorac D. Cytokines and Chemokines Involved in Osteoarthritis Pathogenesis. Int J Mol Sci 2021; 22:9208. [PMID: 34502117 PMCID: PMC8431625 DOI: 10.3390/ijms22179208] [Citation(s) in RCA: 221] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/15/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis is a common cause of disability worldwide. Although commonly referred to as a disease of the joint cartilage, osteoarthritis affects all joint tissues equally. The pathogenesis of this degenerative process is not completely understood; however, a low-grade inflammation leading to an imbalance between anabolic and katabolic processes is a well-established factor. The complex network of cytokines regulating these processes and cell communication has a central role in the development and progression of osteoarthritis. Concentrations of both proinflammatory and anti-inflammatory cytokines were found to be altered depending on the osteoarthritis stage and activity. In this review, we analyzed individual cytokines involved in the immune processes with an emphasis on their function in osteoarthritis.
Collapse
Affiliation(s)
- Vilim Molnar
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vid Matišić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Ivan Kodvanj
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Roko Bjelica
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Željko Jeleč
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Nursing, University North, 48000 Varaždin, Croatia
| | - Damir Hudetz
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Orthopaedic Surgery, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Eduard Rod
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
| | - Fabijan Čukelj
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
- Department of Health Studies, University of Split, 21000 Split, Croatia
- Department of Traumatology, Medical University Merkur Hospital, 10000 Zagreb, Croatia
| | - Trpimir Vrdoljak
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Orthopaedic Surgery, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Dinko Vidović
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
| | | | - Srećko Sabalić
- University Hospital “Sisters of Mercy”, Clinic for Traumatology, Draškovićeva 19, 10000 Zagreb, Croatia;
| | - Borut Dobričić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Orthopaedics and Traumatology, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Tadija Petrović
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Darko Antičević
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Igor Borić
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Department of Traumatology, Medical University Merkur Hospital, 10000 Zagreb, Croatia
- Medical School, University of Split, 21000 Split, Croatia;
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
| | - Rok Košir
- Molecular Biology Laboratory, BIA Separations CRO, Labena Ltd., 1000 Ljubljana, Slovenia; (R.K.); (U.P.Z.)
| | - Uršula Prosenc Zmrzljak
- Molecular Biology Laboratory, BIA Separations CRO, Labena Ltd., 1000 Ljubljana, Slovenia; (R.K.); (U.P.Z.)
| | - Dragan Primorac
- St. Catherine Specialty Hospital, 49210 Zabok, Croatia; (V.M.); (V.M.); (R.B.); (Ž.J.); (D.H.); (E.R.); (F.Č.); (T.V.); (D.V.); (B.D.); (T.P.); (D.A.); (I.B.)
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Split, 21000 Split, Croatia;
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96450 Coburg, Germany
- Eberly College of Science, State College, The Pennsylvania State University, University Park, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
| |
Collapse
|
13
|
Ansari MY, Ahmad N, Haqqi TM. Oxidative stress and inflammation in osteoarthritis pathogenesis: Role of polyphenols. Biomed Pharmacother 2020; 129:110452. [PMID: 32768946 PMCID: PMC8404686 DOI: 10.1016/j.biopha.2020.110452] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/13/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint degenerative disease leading to irreversible structural and functional changes in the joint and is a major cause of disability and reduced life expectancy in ageing population. Despite the high prevalence of OA, there is no disease modifying drug available for the management of OA. Oxidative stress, a result of an imbalance between the production of reactive oxygen species (ROS) and their clearance by antioxidant defense system, is high in OA cartilage and is a major cause of chronic inflammation. Inflammatory mediators, such as interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) are highly upregulated in OA joints and induce ROS production and expression of matrix degrading proteases leading to cartilage extracellular matrix degradation and joint dysfunction. ROS and inflammation are interdependent, each being the target of other and represent ideal target/s for the treatment of OA. Plant polyphenols possess potent antioxidant and anti-inflammatory properties and can inhibit ROS production and inflammation in chondrocytes, cartilage explants and in animal models of OA. The aim of this review is to discuss the chondroprotective effects of polyphenols and modulation of different molecular pathways associated with OA pathogenesis and limitations and future prospects of polyphenols in OA treatment.
Collapse
Affiliation(s)
- Mohammad Yunus Ansari
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, 4209, ST RT 44, Rootstown, Ohio, 44272, USA.
| | - Nashrah Ahmad
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, 4209, ST RT 44, Rootstown, Ohio, 44272, USA; School of Biomedical Sciences, Kent State University, Kent, Ohio, USA.
| | - Tariq M Haqqi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, 4209, ST RT 44, Rootstown, Ohio, 44272, USA.
| |
Collapse
|
14
|
Cherlin S, Lewis MJ, Plant D, Nair N, Goldmann K, Tzanis E, Barnes MR, McKeigue P, Barrett JH, Pitzalis C, Barton A, Cordell HJ. Investigation of genetically regulated gene expression and response to treatment in rheumatoid arthritis highlights an association between IL18RAP expression and treatment response. Ann Rheum Dis 2020; 79:1446-1452. [PMID: 32732242 PMCID: PMC7569378 DOI: 10.1136/annrheumdis-2020-217204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 01/08/2023]
Abstract
Objectives In this study, we sought to investigate whether there was any association between genetically regulated gene expression (as predicted using various reference panels) and anti-tumour necrosis factor (anti-TNF) treatment response (change in erythrocyte sedimentation rate (ESR)) using 3158 European ancestry patients with rheumatoid arthritis. Methods The genetically regulated portion of gene expression was estimated in the full cohort of 3158 subjects (as well as within a subcohort consisting of 1575 UK patients) using the PrediXcan software package with three different reference panels. Estimated expression was tested for association with anti-TNF treatment response. As a replication/validation experiment, we also investigated the correlation between change in ESR with measured gene expression at the Interleukin 18 Receptor Accessory Protein (IL18RAP) gene in whole blood and synovial tissue, using an independent replication data set of patients receiving conventional synthetic disease modifying anti-rheumatic drugs, with directly measured (via RNA sequencing) gene expression. Results We found that predicted expression of IL18RAP showed a consistent signal of association with treatment response across the reference panels. In our independent replication data set, IL18RAP expression in whole blood showed correlation with the change in ESR between baseline and follow-up (r=−0.35, p=0.0091). Change in ESR was also correlated with the expression of IL18RAP in synovial tissue (r=−0.28, p=0.02). Conclusion Our results suggest that IL18RAP expression is worthy of further investigation as a potential predictor of treatment response in rheumatoid arthritis that is not specific to a particular drug type.
Collapse
Affiliation(s)
- Svetlana Cherlin
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Darren Plant
- Centre of Genetics & Genomics Versus Arthritis, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Nisha Nair
- Centre of Genetics & Genomics Versus Arthritis, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Katriona Goldmann
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Evan Tzanis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael R Barnes
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Paul McKeigue
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Jennifer H Barrett
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.,School of Medicine, University of Leeds, Leeds, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anne Barton
- Centre of Genetics & Genomics Versus Arthritis, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Heather J Cordell
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| |
Collapse
|
15
|
Kulkarni P, Martson A, Vidya R, Chitnavis S, Harsulkar A. Pathophysiological landscape of osteoarthritis. Adv Clin Chem 2020; 100:37-90. [PMID: 33453867 DOI: 10.1016/bs.acc.2020.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A sharp rise in osteoarthritis (OA) incidence is expected as over 25% of world population ages in the coming decade. Although OA is considered a degenerative disease, mounting evidence suggests a strong connection with chronic metabolic conditions and low-grade inflammation. OA pathology is increasingly understood as a complex interplay of multiple pathological events including oxidative stress, synovitis and immune responses revealing its intricate nature. Cellular, biochemical and molecular aspects of these pathological events along with major outcomes of the relevant research studies in this area are discussed in the present review. With reference to their published and unpublished work, the authors strongly propose synovitis as a central OA pathology and the key OA pathological events are described in connection with it. Recent research outcomes also have succeeded to establish a linkage between metabolic syndrome and OA, which has been precisely included in the present review. Impact of aging process cannot be neglected in OA. Cell senescence is an important mechanism of aging through which it facilitates development of OA like other degenerative disorders, also discussed within a frame of OA. Conclusively, the reviewers urge low-grade inflammation linked to aging and derailed immune function as a pathological platform for OA development and progression. Thus, interventions targeted to prevent inflammaging hold a promising potential in effective OA management and efforts should be invested in this direction.
Collapse
Affiliation(s)
- Priya Kulkarni
- Department of Pathophysiology, Biomedicine and Translational medicine, University of Tartu, Tartu, Estonia; Department of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia
| | - Aare Martson
- Department of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia; Clinic of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia
| | - Ragini Vidya
- Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India
| | - Shreya Chitnavis
- Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India
| | - Abhay Harsulkar
- Department of Pathophysiology, Biomedicine and Translational medicine, University of Tartu, Tartu, Estonia; Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India.
| |
Collapse
|
16
|
Interleukin-18, interleukin-20, and matrix metalloproteinases (MMP-1, MMP-3) as markers of psoriatic arthritis disease severity and their correlations with biomarkers of inflammation and turnover of joint cartilage. Postepy Dermatol Alergol 2020; 37:1001-1008. [PMID: 33603622 PMCID: PMC7874883 DOI: 10.5114/ada.2020.94903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 01/09/2023] Open
Abstract
Introduction Psoriatic arthritis (PsA) is a chronic, seronegative spondyloarthropathy characterised by joint inflammation and psoriatic skin changes. Recent data indicate that interleukin-18 (IL-18) and interleukin-20 (IL-20) may be involved in the aetiopathogenesis of PsA. Aim To evaluate the potential role of IL-18, IL-20, and matrix metalloproteinases (MMP-1, MMP-3) in the pathogenesis of PsA and their correlations with other markers of inflammation and destruction of joint cartilage, as well as clinical changes. Material and methods The study included 24 patients with PsA and 26 healthy volunteers as a control group. The concentration of IL-18 and IL-20, c-reactive protein (CRP), metalloproteinase-1 and -3 (MMP-1, MMP-3), cartilage oligomeric matrix protein (COMP), aggrecan (PG-AG), and human cartilage glycoprotein (YKL-40) in serum was determined. Clinical severity of the disease according to the BSA, PASI, and DLQI as well as tender and swollen joint count (TJC, SJC) were also evaluated. Results The concentration of IL-18 was statistically significantly higher in the serum of patients with PsA than in the control group (62.87 pg/ml vs. 16.73 pg/ml, p < 0.0049). Serum IL-20 levels in PsA patients were also higher than in the control group, but without statistical significance (p = 0.2939). The ROC curves showed: AUC = 0.81 for IL-18, AUC = 0.75 for IL-20, AUC = 0.96 for COMP, and AUC = 0.89 for MMP-3. Conclusions IL-18 and IL-20 as well as MMP-3 and COMP may be sensitive markers in the diagnosis of PsA.
Collapse
|
17
|
Identifying effector molecules, cells, and cytokines of innate immunity in OA. Osteoarthritis Cartilage 2020; 28:532-543. [PMID: 32044352 DOI: 10.1016/j.joca.2020.01.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 02/06/2023]
Abstract
Inflammatory changes are observed in affected joints of osteoarthritis (OA) patients and are thought to be involved in the pathology that develops along OA progression. This narrative review provides an overview of the various cell types that are present in the joint during OA and which alarmins, cytokines, chemokines, growth factors, and other mediators they produce. Moreover, the involvement of more systemic processes like inflammaging and its associated cellular senescence in the context of OA are discussed.
Collapse
|
18
|
Bao J, Chen Z, Xu L, Wu L, Xiong Y. Rapamycin protects chondrocytes against IL-18-induced apoptosis and ameliorates rat osteoarthritis. Aging (Albany NY) 2020; 12:5152-5167. [PMID: 32182210 PMCID: PMC7138594 DOI: 10.18632/aging.102937] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/27/2020] [Indexed: 12/22/2022]
Abstract
Interleukin 18 (IL-18) promotes inflammation and apoptosis in chondrocytes, thereby contributing to the development and progression of osteoarthritis (OA). Here, we investigated the effects of IL-18 treatment and inhibition in rat chondrocytes in vitro and in vivo. We used RT-PCR and Western blotting to measure the mRNA and protein levels of the chondrocyte-specific genes Collagen II and Aggrecan as well as the protein levels of apoptosis-related (Bax, Bcl2, Caspase3/9), autophagy-related (Atg5, Atg7, Beclin1, LC3), and mTOR pathway-related genes (PI3K, Akt, mTOR). We observed a decrease in Collagen II and Aggrecan mRNA and protein levels, upregulation of chondrocyte apoptosis, downregulation of chondrocyte autophagy, and activation of the PI3K/Akt/mTOR pathway upon IL-18 treatment. PI3K/Akt/mTOR pathway activation and inhibition tests using rat 740Y-P (PI3K activator), SC79 (AKT activator), 3BDO (mTOR activator), or LY294002 (PI3K inhibitor) revealed that activation of the PI3K/Akt/mTOR pathway enhances chondrocyte-specific gene degradation induced by IL-18, while its inhibition has protective effects on chondrocytes. We also found that treatment with rapamycin (a selective mTOR inhibitor) also exerts chondro-protective effects that ameliorate OA by promoting autophagy. These results suggest that inhibition of the mTOR pathway could be exploited for therapeutic benefits in the treatment of OA.
Collapse
Affiliation(s)
- Jiapeng Bao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Zhonggai Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Langhai Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Lidong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Yan Xiong
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| |
Collapse
|
19
|
Koh SM, Chan CK, Teo SH, Singh S, Merican A, Ng WM, Abbas A, Kamarul T. Elevated plasma and synovial fluid interleukin-8 and interleukin-18 may be associated with the pathogenesis of knee osteoarthritis. Knee 2020; 27:26-35. [PMID: 31917106 DOI: 10.1016/j.knee.2019.10.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 02/02/2023]
Abstract
PURPOSE Osteoarthritis (OA) of the knee is a multifactorial degenerative disease typically defined as the 'wear and tear' of articular joint cartilage. However, recent studies suggest that OA is a disease arising from chronic low-grade inflammation. We conducted a study to investigate the relationship between chronic inflammatory mediators present in both the systemic peripheral blood system and localised inflammation in synovial fluid (SF) of OA and non-OA knees; and subsequently made direct comparative analyses to understand the mechanisms that may underpin the processes involved in OA. METHODS 20-Plex proteins were quantified using Human Magnetic Luminex® assay (R&D Systems, USA) from plasma and SF of OA (n = 14) and non-OA (n = 14) patients. Ingenuity Pathway Analysis (IPA) software was used to predict the relationship and possible interaction of molecules pertaining to OA. RESULTS There were significant differences in plasma level for matrix metalloproteinase (MMP)-3, interleukin (IL)-27, IL-8, IL-4, tumour necrosis factor-alpha, MMP-1, IL-15, IL-21, IL-10, and IL-1 beta between the groups, as well as significant differences in SF level for IL-15, IL-8, vascular endothelial growth factor (VEGF), MMP-1, and IL-18. Our predictive OA model demonstrated that toll-like receptor (TLR) 2, macrophage migration inhibitory factor (MIF), TLR4 and IL-1 were the main regulators of IL-1B, IL-4, IL-8, IL-10, IL-15, IL-21, IL-27, MMP-1 and MMP-3 in the plasma system; whilst IL-1B, TLR4, IL-1, and basigin (BSG) were the regulators of IL-4, IL-8, IL-10, IL-15, IL-18, IL-21, IL-27, MMP-1, and MMP-3 in the SF system. CONCLUSION The elevated plasma IL-8 and SF IL-18 may be associated with the pathogenesis of OA via the activation of MMP-3.
Collapse
Affiliation(s)
- S M Koh
- Tissue Engineering Group (TEG), Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - C K Chan
- Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - S H Teo
- Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - S Singh
- Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - A Merican
- Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - W M Ng
- Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - A Abbas
- Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - T Kamarul
- Tissue Engineering Group (TEG), Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
20
|
Liu Y, Mo CF, Luo XY, Li H, Guo HJ, Sun H, Hu S, Li LM, Wang YT, Yang SX, Chang S, Zou Q. Activation of Toll-Like Receptor 3 Induces Interleukin-1 Receptor Antagonist Expression by Activating the Interferon Regulatory Factor 3. J Innate Immun 2019; 12:304-320. [PMID: 31865314 DOI: 10.1159/000504321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptor 3 (TLR3) is a sensor of endogenous cell necrosis during the process of acute inflammation. Interleukin (IL)-1 receptor antagonist (IL-1Ra) is an anti-inflammatory cytokine and can negatively regulate the pathogenesis of inflammation. However, whether and how activation of TLR3 can regulate IL-1Ra expression has not been clarified. Here, we show that poly(I:C) induces IL-1Ra expression in primarily cultured human fibroblast-like synoviocytes and other types of cells. Induction of IL-1Ra by poly(I:C) was dependent on TLR3, but was independent of melanoma differentiation--associated protein 5 or retinoic acid-inducible gene I. Interferon regulatory factor 3 (IRF3) directly binds to the IL-1Ra promoter and promotes IL-1Ra expression in response to poly(I:C) stimulation. Induction of IL-1Ra by poly(I:C) was abolished by the inhibition of the NF-κB signaling, attenuated by the inhibition of the PI3K-Akt signaling, enhanced by inhibition of the ERK1/2 or MSK1/2 activation, but was independent of the p38 MAPK signaling. Treatment with poly(I:C) or Sendai virus elevated the levels of serum IL-1Ra in wild-type, but not in TLR3-/- or IRF3-/- mice. Our findings may provide new insights into the intrinsic anti-inflammatory function of TLR3 and double-stranded RNA-induced IL-Ra expression by TLR3 and its regulation.
Collapse
Affiliation(s)
- Yang Liu
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Chun-Fen Mo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Xing-Yan Luo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Hua Li
- Cancer Center, Chengdu Military General Hospital, Chengdu, China
| | - Hui-Jie Guo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Hai Sun
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Song Hu
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Li-Mei Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Yan-Tang Wang
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Shu-Xia Yang
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China.,Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Shan Chang
- Department of Orthopedics, First Teaching Hospital, Chengdu Medical College, Chengdu, China
| | - Qiang Zou
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China, .,Center of Science and Research, Chengdu Medical College, Chengdu, China,
| |
Collapse
|
21
|
Wang X, Tang K, Wang Y, Chen Y, Yang M, Gu C, Wang J, Wang Y, Yuan Y. Elevated microRNA‑145‑5p increases matrix metalloproteinase‑9 by activating the nuclear factor‑κB pathway in rheumatoid arthritis. Mol Med Rep 2019; 20:2703-2711. [PMID: 31322192 PMCID: PMC6691224 DOI: 10.3892/mmr.2019.10499] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
The present study explored whether miR‑145‑5p can aggravate the development and progression of rheumatoid arthritis (RA) by regulating the expression of matrix metalloproteinases (MMPs). ELISAs, reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR), and western blotting were used to examine the expression levels of MMP‑1, MMP‑3, MMP‑9, and MMP‑13 in fibroblast‑like synoviocytes (FLS) from patients with RA. Levels of MMP‑1, MMP‑3, MMP‑9, and MMP‑13 were assessed in the right hind ankles of a murine collagen‑induced arthritis (CIA) model by RT‑qPCR and immunohistochemical (IHC) analysis. The effects of activation or inhibition of the nuclear factor‑κB (NF‑κB) pathway on MMPs were evaluated by RT‑qPCR and western blotting. Subcellular localization of NF‑κB p65 was visualized by confocal microscopy. Overexpression of miR‑145‑5p increased the expression of MMP‑3, MMP‑9, and MMP‑13 in RA‑FLS. Moreover, injection of a miR‑145‑5p agomir into mice increased MMP‑3, MMP‑9, and MMP‑13, as demonstrated by RT‑qPCR and IHC analysis. A chemical inhibitor that selectively targets NF‑κB (BAY11‑7082) significantly attenuated MMP‑9 expression, while it did not influence the levels of MMP‑3 and MMP‑13. Immunofluorescence analysis revealed that nuclear localization of p65 was significantly enhanced, indicating that miR‑145‑5p enhances activation of the NF‑κB pathway by promoting p65 nuclear translocation. miR‑145‑5p overexpression also significantly increased phosphorylated p65 levels; however, the levels of IkB‑a were reduced in response to this miRNA. Moreover, our results indicated that miR‑145‑5p aggravated RA progression by activating the NF‑κB pathway, which enhanced secretion of MMP‑9. In conclusion, modulation of miR‑145‑5p expression is potentially useful for the treatment of RA inflammation, by regulating the expression of MMPs, and MMP‑9 in particular, through inhibition of the NF‑κB pathway.
Collapse
Affiliation(s)
- Xiaoxue Wang
- Clinical Laboratory Diagnostics, Tianjin Medical University General Hospital Airport Site, Tianjin 300308, P.R. China
| | - Ke Tang
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yuanyuan Wang
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yaqing Chen
- School of Laboratory Medicine, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Mengchen Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Chungang Gu
- Clinical Laboratory Diagnostics, Tianjin Third Center Hospital, Tianjin 300170, P.R. China
| | - Jing Wang
- Clinical Laboratory Diagnostics, Tianjin Medical University General Hospital Airport Site, Tianjin 300308, P.R. China
| | - Yi Wang
- Clinical Laboratory Diagnostics, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Yuhua Yuan
- Clinical Laboratory Diagnostics, Tianjin Medical University General Hospital Airport Site, Tianjin 300308, P.R. China
| |
Collapse
|
22
|
Ruan G, Xu J, Wang K, Wu J, Zhu Q, Ren J, Bian F, Chang B, Bai X, Han W, Ding C. Associations between knee structural measures, circulating inflammatory factors and MMP13 in patients with knee osteoarthritis. Osteoarthritis Cartilage 2018; 26:1063-1069. [PMID: 29753949 DOI: 10.1016/j.joca.2018.05.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 02/25/2018] [Accepted: 05/03/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate cross-sectional associations between serum level of Matrix metalloproteinase (MMP)13 and knee structural measures and circulating inflammatory factors in patients with symptomatic knee osteoarthritis (OA). DESIGN A total of 149 subjects with symptomatic knee OA were included. Magnetic resonance imaging was used to measure infrapatellar fat pad (IPFP) volume, IPFP signal intensity alternation, cartilage volume and cartilage defects. Knee radiography was used to assess radiographic OA using the Kellgren-Lawrence (K-L) grading system. Enzyme-linked immunosorbent assay was used to measure the serum levels of inflammatory factors and MMP13. RESULTS In multivariable analyses, serum MMP13 was negatively associated with cartilage volume at patellar site (β: -32.94 mm3 per 10 ng/ml, P < 0.05), and positively associated with cartilage defect at medial femoral site (OR: 1.13 per 10 ng/ml, P < 0.05). Also, MMP13 was positively associated with K-L grading and IPFP signal intensity alteration (OR: 1.14 and 1.15 per 10 ng/ml, respectively, both P < 0.05), and negatively associated with IPFP volume (β: -0.34 cm3 per 10 ng/ml, P < 0.05). Furthermore, serum level of adiponectin was negatively associated serum MMP13 quartiles (OR: 0.66 per 10 μg/ml, P < 0.05), and serum levels of tumor necrosis factor (TNF)-α, interleukin (IL)-8 and IL-18 were positively associated with serum MMP13 quartiles (ORs: 1.01-1.18 per 10 pg/ml, all P < 0.05). CONCLUSIONS Serum level of MMP13 was associated with knee structural abnormalities as well as serum inflammatory factors. These suggest that systemic MMP13 may play a role in knee OA, and could be regulated by inflammatory factors.
Collapse
Affiliation(s)
- G Ruan
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - J Xu
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - K Wang
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - J Wu
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Q Zhu
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - J Ren
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - F Bian
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - B Chang
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - X Bai
- Translational Research Centre, Academy of Orthopaedics, Guangdong Province, China; School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - W Han
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - C Ding
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Translational Research Centre, Academy of Orthopaedics, Guangdong Province, China; School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
23
|
Bao J, Ma C, Ran J, Xiong Y, Yan S, Wu L. Wnt/β-catenin and Hedgehog pathways are involved in the inflammatory effect of Interleukin 18 on rat chondrocytes. Oncotarget 2017; 8:109962-109972. [PMID: 29299122 PMCID: PMC5746357 DOI: 10.18632/oncotarget.20584] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 08/08/2017] [Indexed: 01/30/2023] Open
Abstract
To investigate the inflammatory effect of Interleukin 18 (IL-18) on rat chondrocytes and the involvement of Wnt/β-catenin and Hedgehog pathways, the mRNA and protein level of matix-degrading enzymes (MMP-2, 3, 9,13 and aggrecanses) and chondrocyte-specific proteins (Collagen II and aggrecan) were evaluated by qRT-PCR and Western blot, and key protein level of Wnt/β-catenin and Hedgehog pathways including β-catenin, GSK-3β, DKK-1, IHH, SHH, and Gli-2 were evaluated by Western blot. Dickkopf-1 (DKK-1) and Cyclopamine were used as antagonist of Wnt/β-catenin and Hedgehog pathways to perform pathway inhibition tests. In addition, location and expression of β-catenin, GSK-3β, Gli-2 and Smo were assessed by Immunofluorescence microscopy. The results showed up-regulation of matix-degrading enzymes (MMP-2, 3, 9,13 and aggrecanses) and down-regulation of chondrocyte-specific proteins (Collagen II and aggrecan) at both mRNA and protein level and activation of Wnt/β-catenin and Hedgehog pathways in the inflammatory reaction on rat chondrocytes caused by IL-18 treatment was observed. As conclusion, Wnt/β-catenin and Hedgehog pathways are involved in the inflammatory effect of IL-18 in vitro.
Collapse
Affiliation(s)
- Jiapeng Bao
- Department of Orthopedics Surgery, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Chiyuan Ma
- Department of Orthopedics Surgery, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Jisheng Ran
- Department of Orthopedics Surgery, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Yan Xiong
- Department of Orthopedics Surgery, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Shigui Yan
- Department of Orthopedics Surgery, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Lidong Wu
- Department of Orthopedics Surgery, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| |
Collapse
|
24
|
Chen JH, Tsai CH, Lin HY, Huang CF, Leung YM, Lai SW, Tsai CF, Chang PC, Lu DY, Lin C. Interlukin-18 Is a Pivot Regulatory Factor on Matrix Metalloproteinase-13 Expression and Brain Astrocytic Migration. Mol Neurobiol 2015; 53:6218-6227. [PMID: 26558633 DOI: 10.1007/s12035-015-9529-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/05/2015] [Indexed: 01/28/2023]
Abstract
The expression of matrix metalloproteinase-13 (MMP-13) has been shown to be elevated in some pathophysiological conditions and is involved in the degradation of extracellular matrix in astrocytes. In current study, the function of MMP-13 was further investigated. The conditioned medium (CM) collected from activated microglia increased interleukin (IL)-18 production and enhanced MMP-13 expression in astrocytes. Furthermore, treatment with recombinant IL-18 increased MMP-13 protein and mRNA levels in astrocytes. Recombinant IL-18 stimulation also increased the enzymatic activity of MMP-13 and the migratory activity of astrocytes, while administration of MMP-13 or pan-MMP inhibitors antagonized IL-18-induced migratory activity of astrocytes. In addition, administration of recombinant IL-18 to astrocytes led to the phosphorylation of JNK, Akt, or PKCδ, and treatment of astrocytes with JNK, PI3 kinase/Akt, or PKCδ inhibitors significantly decreased the IL-18-induced migratory activity. Taken together, the results suggest that IL-18-induced MMP-13 expression in astrocytes is regulated by JNK, PI3 kinase/Akt, and PKCδ signaling pathways. These findings also indicate that IL-18 is an important regulator leading to MMP-13 expression and cell migration in astrocytes.
Collapse
Affiliation(s)
- Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chon-Haw Tsai
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Chien-Fang Huang
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Yuk-Man Leung
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Sheng-Wei Lai
- Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan. .,Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan.
| |
Collapse
|
25
|
Wang Y, Xu D, Long L, Deng X, Tao R, Huang G. Correlation between plasma, synovial fluid and articular cartilage Interleukin-18 with radiographic severity in 33 patients with osteoarthritis of the knee. Clin Exp Med 2015; 14:297-304. [PMID: 23958877 DOI: 10.1007/s10238-013-0251-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 07/15/2013] [Indexed: 11/30/2022]
Abstract
Osteoarthritis (OA) is a complex disease characterized by cartilage degeneration, secondary synovial membrane inflammation and subchondral bone changes. In recent years, many studies have confirmed that interleukin-18 (IL-18) is involved in the inflammatory process of inflammatory joint diseases. In the present study, we investigated IL-18 levels in plasma, synovial fluid and articular cartilage of patients with primary knee OA (n = 33) to analyze their relationship with radiographic severity. Compared to healthy controls (n = 15), OA patients had higher plasma and synovial fluid IL-18 concentrations(45.8 ± 22.1 vs. 23.7 ± 13.6 pg/ml, P<0.001 and 75.2 ± 40.1 vs. 28.3 ± 11.6 pg/ml, P<0.001) as measured by enzyme-linked immunosorbent assay. Also,the percentage of immunofluorescent IL-18 positive cells in articular cartilage was significantly increased in OA compared to controls (46.5 ± 10.3 vs. 2.9 ± 1.7, P<0.001). Moreover, plasma, synovial fluid and articular cartilage IL-18 significantly positively correlated with radiographic severity, respectively (r = 0.663, P<0.001, r = 0.56, P = 0.001 and r = 0.884, P<0.001). Subsequent analysis revealed that plasma, synovial fluid and articular cartilage IL-18 levels positively correlated with each other (r = 0.632, P<0.001, r = 0.489, P = 0.004 and r = 0.620, P<0.001). These data suggested that plasma, synovial fluid and articular cartilage IL-18 levels were significantly increased in OA patients, and these elevated levels were positively correlated with radiographic severity. Accordingly, our study supports the role of IL-18 in the pathophysiology of OA.
Collapse
|
26
|
The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm 2014; 2014:561459. [PMID: 24876674 PMCID: PMC4021678 DOI: 10.1155/2014/561459] [Citation(s) in RCA: 1046] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 04/12/2014] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic disease of human joints. The basis of pathologic changes involves all the tissues forming the joint; already, at an early stage, it has the nature of inflammation with varying degrees of severity. An analysis of the complex relationships indicates that the processes taking place inside the joint are not merely a set that (seemingly) only includes catabolic effects. Apart from them, anti-inflammatory anabolic processes also occur continually. These phenomena are driven by various mediators, of which the key role is attributed to the interactions within the cytokine network. The most important group controlling the disease seems to be inflammatory cytokines, including IL-1β, TNFα, IL-6, IL-15, IL-17, and IL-18. The second group with antagonistic effect is formed by cytokines known as anti-inflammatory cytokines such as IL-4, IL-10, and IL-13. The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of OA with respect to inter- and intracellular signaling pathways is still under investigation. This paper summarizes the current state of knowledge. The cytokine network in OA is put in the context of cells involved in this degenerative joint disease. The possibilities for further implementation of new therapeutic strategies in OA are also pointed.
Collapse
|
27
|
Beaulieu LM, Lin E, Mick E, Koupenova M, Weinberg EO, Kramer CD, Genco CA, Tanriverdi K, Larson MG, Benjamin EJ, Freedman JE. Interleukin 1 receptor 1 and interleukin 1β regulate megakaryocyte maturation, platelet activation, and transcript profile during inflammation in mice and humans. Arterioscler Thromb Vasc Biol 2014; 34:552-64. [PMID: 24458711 DOI: 10.1161/atvbaha.113.302700] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Interleukin 1 Receptor 1 (IL1R1) and its ligand, IL1β, are upregulated in cardiovascular disease, obesity, and infection. Previously, we reported a higher level of IL1R1 transcripts in platelets from obese individuals of the Framingham Heart Study (FHS), but its functional effect in platelets has never been described. Additionally, IL1β levels are increased in atherosclerotic plaques and in bacterial infections. The aim of this work is to determine whether IL1β, through IL1R1, can activate platelets and megakaryocytes to promote atherothrombosis. APPROACH AND RESULTS We found that IL1β-related genes from platelets, as measured in 1819 FHS participants, were associated with increased body mass index, and a direct relationship was shown in wild-type mice fed a high-fat diet. Mechanistically, IL1β activated nuclear factor-κB and mitogen-activated protein kinase signaling pathways in megakaryocytes. IL1β, through IL1R1, increased ploidy of megakaryocytes to 64+ N by 2-fold over control. IL1β increased agonist-induced platelet aggregation by 1.2-fold with thrombin and 4.2-fold with collagen. IL1β increased adhesion to both collagen and fibrinogen, and heterotypic aggregation by 1.9-fold over resting. High fat diet-enhanced platelet adhesion was absent in IL1R1(-/-) mice. Wild-type mice infected with Porphyromonas gingivalis had circulating heterotypic aggregates (1.5-fold more than control at 24 hours and 6.2-fold more at 6 weeks) that were absent in infected IL1R1(-/-) and IL1β(-/-) mice. CONCLUSIONS In summary, IL1R1- and IL1β-related transcripts are elevated in the setting of obesity. IL1R1/IL1β augment both megakaryocyte and platelet functions, thereby promoting a prothrombotic environment during infection and obesity; potentially contributing to the development of atherothrombotic disease.
Collapse
Affiliation(s)
- Lea M Beaulieu
- From the Department of Medicine (L.M.B., M.K., K.T., J.E.F.) and Quantitative Health Sciences (E.M.), University of Massachusetts Medical School, Worcester, MA; Department of Medicine (E.L., M.K., E.O.W., C.D.K., C.A.G., E.J.B.), Section of Infectious Disease (C.A.G.), and Department of Microbiology (C.A.G.), Boston University School of Medicine, MA; NHLBI and Boston University's Framingham Heart Institute, Framingham, MA (M.G.L., E.J.B.); and Department of Mathematics and Statistics, Boston University, MA (M.G.L.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Shen W, Chen J, Zhu T, Yin Z, Chen X, Chen L, Fang Z, Heng BC, Ji J, Chen W, Ouyang HW. Osteoarthritis prevention through meniscal regeneration induced by intra-articular injection of meniscus stem cells. Stem Cells Dev 2013; 22:2071-82. [PMID: 23461527 DOI: 10.1089/scd.2012.0563] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Meniscus injury is frequently encountered in clinical practice. Current surgical therapy involving partial or complete meniscectomy relieves pain in the short-term but often leads to osteoarthritis (OA) in the long-term. Here, this study aimed to identify and characterize a novel population of meniscus-derived stem cells (MeSCs) and develop a new strategy of articular cartilage protection by intra-articular injection of these cells. The "stemness" and immune properties of MeSCs were investigated in vitro, while the efficacy of intra-articular injection of MeSCs for meniscus regeneration and OA prevention were investigated in vivo at 4, 8, and 12 weeks postsurgery. MeSCs displayed typical stem cell characteristics such as low immunogenicity and even possessed immunosuppressive function. In a rabbit meniscus injury model, transplantation of allogenous MeSCs did not elicit immunological rejection, but promoted neo-tissue formation with better-defined shape and more matured extracellular matrix. In a rabbit experimental OA model, transplantation of MeSCs further protected joint surface cartilage and maintained joint space at 12 weeks postsurgery, whereas extensive joint surface irregularities and joint space stenosis were observed in the control group. This study thus evoked a new strategy for articular cartilage protection and meniscus regeneration by intra-articular injection of MeSCs for patients undergoing meniscectomy.
Collapse
Affiliation(s)
- Weiliang Shen
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Williams EL, Edwards CJ, Cooper C, Oreffo ROC. Impact of inflammation on the osteoarthritic niche: implications for regenerative medicine. Regen Med 2012; 7:551-70. [DOI: 10.2217/rme.12.34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis worldwide and is the sixth leading cause of disability. It costs the UK economy approximately 1% of gross national product per annum. With an aging population, the cost of chronic conditions such as OA continues to rise. Historically, treatments for OA have been limited to painkillers, physiotherapy and joint injections. When these fail, patients are referred for joint replacement surgery. With the advent of tissue engineering strategies aimed at generating new bone and cartilage for repair of osteochondral defects, there has been considerable interest in exploiting these techniques to devise new treatments for OA. To date, little consideration has been given to the OA niche and attendant inflammatory milieu for any regenerative skeletal strategy. This review highlights the importance of understanding the osteoarthritic niche in order to modify existing tissue engineering and regenerative medicine strategies for the future treatment of OA.
Collapse
Affiliation(s)
- Emma L Williams
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development & Health, University of Southampton Medical School, Southampton, UK
| | - Christopher J Edwards
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development & Health, University of Southampton Medical School, Southampton, UK
- Rheumatology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Cyrus Cooper
- Rheumatology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- MRC Lifecourse Epidemiology Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Richard OC Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development & Health, University of Southampton Medical School, Southampton, UK
| |
Collapse
|
30
|
Liu YP, Yu GR. Comment on “The matrix metalloproteinases as pharmacological target in osteoarthritis: Statins may be of therapeutic benefit”. Med Hypotheses 2010; 74:394-5. [DOI: 10.1016/j.mehy.2009.09.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2009] [Revised: 09/21/2009] [Accepted: 09/23/2009] [Indexed: 11/29/2022]
|
31
|
|
32
|
Abstract
As the cellular component of articular cartilage, chondrocytes are responsible for maintaining in a low-turnover state the unique composition and organization of the matrix that was determined during embryonic and postnatal development. In joint diseases, cartilage homeostasis is disrupted by mechanisms that are driven by combinations of biological mediators that vary according to the disease process, including contributions from other joint tissues. In osteoarthritis (OA), biomechanical stimuli predominate with up-regulation of both catabolic and anabolic cytokines and recapitulation of developmental phenotypes, whereas in rheumatoid arthritis (RA), inflammation and catabolism drive cartilage loss. In vitro studies in chondrocytes have elucidated signaling pathways and transcription factors that orchestrate specific functions that promote cartilage damage in both OA and RA. Thus, understanding how the adult articular chondrocyte functions within its unique environment will aid in the development of rational strategies to protect cartilage from damage resulting from joint disease. This review will cover current knowledge about the specific cellular and biochemical mechanisms that regulate cartilage homeostasis and pathology.
Collapse
Affiliation(s)
- Mary B Goldring
- Research Division, Hospital for Special Surgery, Affiliated with Weill College of Medicine of Cornell University, New York, NY 10021, USA.
| | | |
Collapse
|
33
|
Sakao K, Takahashi KA, Arai Y, Saito M, Honjo K, Hiraoka N, Asada H, Shin-Ya M, Imanishi J, Mazda O, Kubo T. Osteoblasts derived from osteophytes produce interleukin-6, interleukin-8, and matrix metalloproteinase-13 in osteoarthritis. J Bone Miner Metab 2009; 27:412-23. [PMID: 19333684 DOI: 10.1007/s00774-009-0058-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Accepted: 10/10/2008] [Indexed: 10/20/2022]
Abstract
To clarify the significance of the osteophytes that appear during the progression of osteoarthritis (OA), we investigated the expression of inflammatory cytokines and proteases in osteoblasts from osteophytes. We also examined the influence of mechanical stress loading on osteoblasts on the expression of inflammatory cytokines and proteases. Osteoblasts were isolated from osteophytes in 19 patients diagnosed with knee OA and from subchondral bone in 4 patients diagnosed with femoral neck fracture. Messenger RNA expression and protein production of inflammatory cytokines and proteases were analyzed using real-time RT-PCR and ELISA, respectively. To examine the effects of mechanical loading, continuous hydrostatic pressure was applied to the osteoblasts. We determined the mRNA expression and protein production of IL-6, IL-8, and MMP-13, which are involved in the progression of OA, were increased in the osteophytes. Additionally, when OA pathological conditions were simulated by applying a nonphysiological mechanical stress load, the gene expression of IL-6 and IL-8 increased. Our results suggested that nonphysiological mechanical stress may induce the expression of biological factors in the osteophytes and is involved in OA progression. By controlling the expression of these genes in the osteophytes, the progression of cartilage degeneration in OA may be reduced, suggesting a new treatment strategy for OA.
Collapse
Affiliation(s)
- Kei Sakao
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Inoue H, Hiraoka K, Hoshino T, Okamoto M, Iwanaga T, Zenmyo M, Shoda T, Aizawa H, Nagata K. High levels of serum IL-18 promote cartilage loss through suppression of aggrecan synthesis. Bone 2008; 42:1102-10. [PMID: 18374640 DOI: 10.1016/j.bone.2008.01.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 01/04/2008] [Accepted: 01/27/2008] [Indexed: 11/29/2022]
Abstract
Osteoarthritis (OA) is closely related to the function of several inflammatory cytokines. It has been reported that older age is associated with higher serum levels of the inflammatory cytokine IL-18. In the present study, we investigated the long-term role of serum IL-18 in cartilage loss in vivo using a new strain of IL-18 transgenic mouse (Tg) in comparison with wild-type (WT) mice. The IL-18 Tg mouse strain we developed constitutively overproduces soluble mature IL-18 in the lungs but not in other tissues, including joints. These Tg mice showed high levels of serum IL-18, but not IL-1beta. No inflammatory cells, fibrillation or synovitis were observed in the knee joints of either IL-18 Tg or WT mice. However, the cartilage cellularity of the femoral and tibial condyles of IL-18 Tg mice was significantly reduced in comparison with control WT mice. Aggrecan was detected in only a few cells in the deep zone of the articular cartilage of Tg mice. The expression of aggrecan mRNA was also significantly decreased in articular chondrocytes from Tg mice when compared with WT mice. In contrast, endogenous IL-18 mRNA was significantly increased in the chondrocytes of Tg mice in comparison with WT mice. Expression of IFN-gamma was also significantly increased in the Tg mice. Moreover, IL-18 transgene-positive caspase-1-deficient mice showed articular cartilage loss that was independent of endogenous IL-1beta. In cultured chondrocytes isolated from WT mice, the expression of aggrecan mRNA was dosage-dependently suppressed by treatment with recombinant IL-18. In contrast, IL-18 stimulated the expression of mRNA for endogenous IL-18 and IFN-gamma. These results suggest that high levels of serum IL-18 promote the overexpression of endogenous IL-18 in articular chondrocytes, resulting in cartilage loss through suppression of aggrecan synthesis. Thus IL-18 may play an important role in the pathogenesis of articular cartilage loss in osteoarthritis.
Collapse
Affiliation(s)
- Hidetake Inoue
- Department of Orthopedics, Kurume University School of Medicine, 67 Ashahi-machi, Kurume 830-0011, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Rheumatoid arthritis (RA) is one of the inflammatory joint diseases in a heterogeneous group of disorders that share features of destruction of the extracellular matrices of articular cartilage and bone. The underlying disturbance in immune regulation that is responsible for the localized joint pathology results in the release of inflammatory mediators in the synovial fluid and synovium that directly and indirectly influence cartilage homeostasis. Analysis of the breakdown products of the matrix components of joint cartilage in body fluids and quantitative imaging techniques have been used to assess the effects of the inflammatory joint disease on the local remodeling of joint structures. The role of the chondrocyte itself in cartilage destruction in the human rheumatoid joint has been difficult to address but has been inferred from studies in vitro and in animal models. This review covers current knowledge about the specific cellular and biochemical mechanisms that account for the disruption of the integrity of the cartilage matrix in RA.
Collapse
Affiliation(s)
- Miguel Otero
- Research Division of the Hospital for Special Surgery, Weill College of Medicine of Cornell University, Caspary Research Building, 535 E. 70th Street, New York, NY 10021, USA
| | | |
Collapse
|
36
|
Abstract
Recent data are presented which indicate a critical role for interleukin (IL)-18 in rheumatoid arthritis (RA). The T cells and macrophages invading the synovium or in the synovial fluid are the chief cellular targets of IL-18 in RA. Neutrophils, dendritic cells and endothelial cells may also be cellular mediators of IL-18. The direct effect of IL-18 on fibroblast-like synoviocytes or chondrocytes may not be essential or important. In RA, IL-18, which is mainly produced by macrophages, activates T cells and macrophages to produce proinflammatory cytokines, chemokines, adhesion molecules and RANKL which, in turn, perpetuate chronic inflammation and induce bone and cartilage destruction.
Collapse
Affiliation(s)
- Sheng-Ming Dai
- Department of Rheumatology and Immunology, Changhai Hospital, Second Military Medical University, 174 Changhai Road, Shanghai 200433, P R China.
| | | | | | | |
Collapse
|
37
|
Kang MJ, Homer RJ, Gallo A, Lee CG, Crothers KA, Cho SJ, Rochester C, Cain H, Chupp G, Yoon HJ, Elias JA. IL-18 is induced and IL-18 receptor alpha plays a critical role in the pathogenesis of cigarette smoke-induced pulmonary emphysema and inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:1948-59. [PMID: 17237446 DOI: 10.4049/jimmunol.178.3.1948] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Th1 inflammation and remodeling characterized by local tissue destruction coexist in pulmonary emphysema and other diseases. To test the hypothesis that IL-18 plays an important role in these responses, we characterized the regulation of IL-18 in lungs from cigarette smoke (CS) and room air-exposed mice and characterized the effects of CS in wild-type mice and mice with null mutations of IL-18Ralpha (IL-18Ralpha(-/-)). CS was a potent stimulator and activator of IL-18 and caspases 1 and 11. In addition, although CS caused inflammation and emphysema in wild-type mice, both of these responses were significantly decreased in IL-18Ralpha(-/-) animals. CS also induced epithelial apoptosis, activated effector caspases and stimulated proteases and chemokines via IL-18Ralpha-dependent pathways. Importantly, the levels of IL-18 and its targets, cathepsins S and B, were increased in pulmonary macrophages from smokers and patients with chronic obstructive lung disease. Elevated levels of circulating IL-18 were also seen in patients with chronic obstructive lung disease. These studies demonstrate that IL-18 and the IL-18 pathway are activated in CS-exposed mice and man. They also demonstrate, in a murine modeling system, that IL-18R signaling plays a critical role in the pathogenesis of CS-induced inflammation and emphysema.
Collapse
Affiliation(s)
- Min-Jong Kang
- Section of Pulmonary and Critical Care Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|