501
|
Hu X, Xu J, Li W, Li L, Parungao R, Wang Y, Zheng S, Nie Y, Liu T, Song K. Therapeutic "Tool" in Reconstruction and Regeneration of Tissue Engineering for Osteochondral Repair. Appl Biochem Biotechnol 2019; 191:785-809. [PMID: 31863349 DOI: 10.1007/s12010-019-03214-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Repairing osteochondral defects to restore joint function is a major challenge in regenerative medicine. However, with recent advances in tissue engineering, the development of potential treatments is promising. In recent years, in addition to single-layer scaffolds, double-layer or multilayer scaffolds have been prepared to mimic the structure of articular cartilage and subchondral bone for osteochondral repair. Although there are a range of different cells such as umbilical cord stem cells, bone marrow mesenchyml stem cell, and others that can be used, the availability, ease of preparation, and the osteogenic and chondrogenic capacity of these cells are important factors that will influence its selection for tissue engineering. Furthermore, appropriate cell proliferation and differentiation of these cells is also key for the optimal repair of osteochondral defects. The development of bioreactors has enhanced methods to stimulate the proliferation and differentiation of cells. In this review, we summarize the recent advances in tissue engineering, including the development of layered scaffolds, cells, and bioreactors that have changed the approach towards the development of novel treatments for osteochondral repair.
Collapse
Affiliation(s)
- Xueyan Hu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Jie Xu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Wenfang Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China.,Key Laboratory of Biological Medicines, Universities of Shandong Province Weifang Key Laboratory of Antibody Medicines, School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, China
| | - Liying Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Roxanne Parungao
- Burns Research Group, ANZAC Research Institute, University of Sydney, Concord, NSW, 2139, Australia
| | - Yiwei Wang
- Burns Research Group, ANZAC Research Institute, University of Sydney, Concord, NSW, 2139, Australia
| | - Shuangshuang Zheng
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou, 450000, China
| | - Yi Nie
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou, 450000, China. .,Key Laboratory of Green Process and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Tianqing Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China.
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
502
|
Cell sources of inflammatory mediators present in bone marrow areas inside the meniscus. PLoS One 2019; 14:e0226986. [PMID: 31860662 PMCID: PMC6924665 DOI: 10.1371/journal.pone.0226986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023] Open
Abstract
Purpose To demonstrate the production of inflammatory mediators by cells located in bone marrow spaces inside rodent menisci. Methods Mice subjected to transection of the medial collateral and anterior cruciate ligaments and meniscotomy (osteoarthritis model) or to a sham procedure, as well as non-operated (naive) mice and rats, had knee joints excised. Tissues were stained with hematoxylin-eosin and tartrate-resistant acid phosphatase (TRAP). CD68+ cells, inducible nitric oxide synthase (iNOS), interleukin (IL)-1β, and tumor necrosis factor (TNF) expression were detected using immunohistochemistry. Results Lamellar ossified areas, bone-entrapped osteocytes and bone marrow spaces were found inside menisci of one week up to 6 months-old naïve mice, regardless of gender. Menisci from naive rats also showed the same pattern with bone marrow areas. CD68+ cells were identified in bone marrow areas inside the meniscus of mice. TRAP+ osteoclasts, and hematogenous precursors expressing IL-1β, TNF, and iNOS were identified inside bone marrow areas in meniscal samples from both naïve and sham operated mice. Quantitative immunoexpression of IL-1 β, TNF and iNOS was more intense, P = 0.0194, 0.0293, 0.0124, respectively, in mouse knees from mice sacrificed 49 days after being subjected to an osteoarthritis (OA) model as compared to sham operated animals. Conclusion We provide novel data showing that rodent menisci display bone marrow areas with cells able to produce inflammatory mediators. Immunoexpression of inflammatory mediators in those bone marrow areas is significantly more pronounced in mice subjected to experimental OA
Collapse
|
503
|
Mobasheri A, Saarakkala S, Finnilä M, Karsdal MA, Bay-Jensen AC, van Spil WE. Recent advances in understanding the phenotypes of osteoarthritis. F1000Res 2019; 8. [PMID: 31885861 PMCID: PMC6913225 DOI: 10.12688/f1000research.20575.1] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2019] [Indexed: 01/15/2023] Open
Abstract
Recent research in the field of osteoarthritis (OA) has focused on understanding the underlying molecular and clinical phenotypes of the disease. This narrative review article focuses on recent advances in our understanding of the phenotypes of OA and proposes that the disease represents a diversity of clinical phenotypes that are underpinned by a number of molecular mechanisms, which may be shared by several phenotypes and targeted more specifically for therapeutic purposes. The clinical phenotypes of OA supposedly have different underlying etiologies and pathogenic pathways and they progress at different rates. Large OA population cohorts consist of a majority of patients whose disease progresses slowly and a minority of individuals whose disease may progress faster. The ability to identify the people with relatively rapidly progressing OA can transform clinical trials and enhance their efficiency. The identification, characterization, and classification of molecular phenotypes of rapidly progressing OA, which represent patients who may benefit most from intervention, could potentially serve as the basis for precision medicine for this disabling condition. Imaging and biochemical markers (biomarkers) are important diagnostic and research tools that can assist with this challenge.
Collapse
Affiliation(s)
- Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, 08661, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland.,Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, UK.,ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, DK-2730, Denmark
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland
| | - Mikko Finnilä
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, FI-90014, Finland
| | - Morten A Karsdal
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, DK-2730, Denmark
| | | | - Willem Evert van Spil
- Division of Internal Medicine & Dermatology, Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Rheumatology, Dijklander Hospital, 1620 AR Hoorn, The Netherlands
| |
Collapse
|
504
|
Wang Y, Teichtahl AJ, Pelletier JP, Abram F, Wluka AE, Hussain SM, Martel-Pelletier J, Cicuttini FM. Knee effusion volume assessed by magnetic resonance imaging and progression of knee osteoarthritis: data from the Osteoarthritis Initiative. Rheumatology (Oxford) 2019; 58:246-253. [PMID: 30204907 DOI: 10.1093/rheumatology/key274] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/23/2022] Open
Abstract
Objective To examine whether baseline knee joint effusion volume and the change in effusion volume over 1 year are associated with cartilage volume loss, progression of radiographic OA (ROA) over 4 years and risk of total knee replacement over 6 years. Methods This study included 4115 Osteoarthritis Initiative participants with knee joint effusion volume quantified by MRI at baseline. The change in effusion volume over 1 year was assessed. Cartilage volume loss and progression of ROA over 4 years were assessed using MRI and X-ray and total knee replacement over 6 years was assessed. Multiple linear regression and binary logistic regression were used for data analyses. Results Baseline knee effusion volume (per 5 ml) was positively associated with a loss of medial and lateral cartilage volume [regression coefficient 0.13%/year (95% CI 0.10, 0.17) and 0.13%/year (95% CI 0.10, 0.16), respectively, both P < 0.001], progression of ROA [odds ratio (OR) 1.28 (95% CI 1.20, 1.37), P < 0.001], and risk of knee replacement [OR 1.12 (95% CI 1.05, 1.20), P = 0.001]. A 5 ml increase in knee effusion volume over 1 year was positively associated with medial cartilage volume loss [regression coefficient 0.09%/year (95% CI 0.04, 0.15), P = 0.001], progression of ROA [OR 1.21 (95% CI 1.11, 1.33), P < 0.001] and risk of knee replacement [OR 1.24 (95% CI 1.12, 1.37), P < 0.001]. Conclusions Knee joint effusion volume assessed from MRI provides a continuous and sensitive measure that was associated with cartilage volume loss, progression of ROA and risk of total knee replacement. It may provide a method to identify individuals with an inflammatory OA phenotype who are at higher risk of disease progression.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Andrew J Teichtahl
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - François Abram
- Medical Imaging Research and Development, ArthroLab Inc., Montreal, Quebec, Canada
| | - Anita E Wluka
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sultana Monira Hussain
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Flavia M Cicuttini
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
505
|
Hoshikawa N, Sakai A, Takai S, Suzuki H. Targeting Extracellular miR-21-TLR7 Signaling Provides Long-Lasting Analgesia in Osteoarthritis. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:199-207. [PMID: 31841992 PMCID: PMC6920297 DOI: 10.1016/j.omtn.2019.11.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) is the most prevalent joint disorder associated with severe chronic pain. Although synovial inflammation is well correlated with pain severity, the molecular mechanism responsible for OA pain remains unclear. Here, we show that extracellular miR-21 released from synovial tissue mediates knee OA pain in surgical OA model rats. miR-21 was the most abundant among increased microRNAs (miRNAs) in the synovial tissue. miR-21 was released into extracellular space from the synovial tissue and increased in the synovial fluid. A single intra-articular injection of miR-21 inhibitor exerted long-term analgesia of knee OA pain, whereas miR-21 injection in naive rats caused knee joint pain. miR-21 mutant, which lacks the Toll-like receptor (TLR) binding motif, but not in the seed sequence, did not cause joint pain, suggesting a non-canonical mode of action different from translational repression. Consistent with this, the algesic effect of miR-21 was blocked by antagonizing TLR7. The TLR7 antagonist also exerted a long-lasting analgesic effect on knee OA pain. Therefore, extracellular miR-21 released from synovial tissue mediates knee OA pain through TLR7 activation in surgical OA model rats. Extracellular miRNA in the joint may be a plausible target for pain therapy, providing a novel analgesic strategy for OA.
Collapse
Affiliation(s)
- Naoya Hoshikawa
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Atsushi Sakai
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | - Shinro Takai
- Department of Orthopaedic Surgery, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Hidenori Suzuki
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| |
Collapse
|
506
|
Liu P, Gu L, Ren L, Chen J, Li T, Wang X, Yang J, Chen C, Sun L. Intra-articular injection of etoricoxib-loaded PLGA-PEG-PLGA triblock copolymeric nanoparticles attenuates osteoarthritis progression. Am J Transl Res 2019; 11:6775-6789. [PMID: 31814887 PMCID: PMC6895527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
The current pharmacological therapies for osteoarthritis (OA) are mainly focused on symptomatic relief of pain and inflammation through the use of nonsteroidal anti-inflammatory drugs (NSAIDs). Etoricoxib is a cyclooxygenase-2 (COX-2) selective NSAID with a higher cyclooxygenase-1 (COX-1) to COX-2 selectivity ratio than the other COX-2 selective NSAIDs and a lower risk of gastrointestinal toxicity compared to traditional NSAIDs. In this study, we first evaluated the anti-inflammatory and chondro-protective effects of etoricoxib on interlecukin-1β-stimulated human osteoarthritic chondrocytes. We found that etoricoxib not only inhibited the expression of inflammation mediators COX-2, prostaglandin E2 (PGE2), and nitric oxide, but also had a similar chondro-protective effect to celecoxib through down-regulating matrix degrading enzymes matrix metalloproteinase-13 (MMP-13) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS-5). We then used PLGA-PEG-PLGA triblock copolymeric nanoparticles (NPs) as a drug delivery system to locally deliver etoricoxib into the articular cavity to reduce the risk of cardiovascular toxicity of etoricoxib when administered systemically or orally. The etoricoxib-loaded NPs showed a sustained drug release over 28 days in vitro; in rat OA model, the intra-articular injection of etoricoxib-loaded NPs alleviated the symptoms of subchondral bone, synovium, and cartilage. In conclusion, our study confirmed the chondro-protective role of etoricoxib in OA, and proved the curative effects of etoricoxib-loaded PLGA-PEG-PLGA NPs in vivo.
Collapse
Affiliation(s)
- Pingju Liu
- Guizhou University of Chinese Traditional MedicineGuiyang 550025, China
- Department of Orthopedics, Guizhou Provincial People’s HospitalGuiyang 550002, China
- Department of Orthopedics, Zunyi Traditional Chinese Medicine HospitalZunyi 563099, China
| | - Liling Gu
- Medical College, Guizhou UniversityGuiyang 550025, China
| | - Lingyan Ren
- Medical College, Guizhou UniversityGuiyang 550025, China
| | - Jiajia Chen
- Biomedical Analysis Center, Army Medical UniversityChongqing 400038, China
| | - Tao Li
- Center for Joint Surgery, Southwest Hospital, Army Medical UniversityChongqing 400038, China
| | - Xin Wang
- Center for Joint Surgery, Southwest Hospital, Army Medical UniversityChongqing 400038, China
| | - Junjun Yang
- Center for Joint Surgery, Southwest Hospital, Army Medical UniversityChongqing 400038, China
| | - Cheng Chen
- Center for Joint Surgery, Southwest Hospital, Army Medical UniversityChongqing 400038, China
| | - Li Sun
- Department of Orthopedics, Guizhou Provincial People’s HospitalGuiyang 550002, China
| |
Collapse
|
507
|
Cho H, Bhatti FUR, Hasty KA, Yi AK. Nanosome-Mediated Delivery Of Protein Kinase D Inhibitor Protects Chondrocytes From Interleukin-1β-Induced Stress And Apoptotic Death. Int J Nanomedicine 2019; 14:8835-8846. [PMID: 31806974 PMCID: PMC6857658 DOI: 10.2147/ijn.s218901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/30/2019] [Indexed: 01/10/2023] Open
Abstract
Background Inflammatory stress caused by protein kinase D (PKD) plays a critical role in damaging chondrocytes and extracellular matrix (ECM) during osteoarthritis (OA). The PKD inhibitor (PKDi) (CRT0066101) has been used to overcome inflammation in different cell types. However, the efficacy of a therapeutic drug can be limited due to off-target distribution, slow cellular internalization, and limited lysosomal escape. In order to overcome this issue, we developed nanosomes carrying CRT0066101 (PKDi-Nano) and tested their efficacy in vitro in chondrocytes. Methods Chondrocytes were subjected to IL-1β-induced inflammatory stress treated with either PKDi or PKDi-Nano. Effects of treatment were measured in terms of cytotoxicity, cellular morphology, viability, apoptosis, phosphorylation of protein kinase B (Akt), and anabolic/catabolic gene expression analyses related to cartilage tissue. Results and Discussion The effects of PKDi-Nano treatment were more pronounced as compared to PKDi treatment. Cytotoxicity and apoptosis were significantly reduced following PKDi-Nano treatment (P < 0.001). Cellular morphology was also restored to normal size and shape. The viability of chondrocytes was significantly enhanced in PKDi-Nano-treated cells (P < 0.001). The data indicated that PKDi-Nano acted independently of the Akt pathway. Gene expression analyses revealed significant increases in the expression levels of anabolic genes with concomitant decreases in the level of catabolic genes. Our results indicate that PKDi-Nano attenuated the effects of IL-1β via the nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) pathway. Conclusion Taken together, these results suggest that PKDi-Nano can be used as a successful strategy to reduce IL1β-induced inflammatory stress in chondrocytes.
Collapse
Affiliation(s)
- Hongsik Cho
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Orthopaedic Surgery, Campbell Clinic, Memphis, TN, USA.,151 Research Service, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Fazal-Ur-Rehman Bhatti
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center, Memphis, TN, USA.,151 Research Service, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Karen A Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Orthopaedic Surgery, Campbell Clinic, Memphis, TN, USA.,151 Research Service, Veterans Affairs Medical Center, Memphis, TN, USA
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
508
|
MiR-33b-3p promotes chondrocyte proliferation and inhibits chondrocyte apoptosis and cartilage ECM degradation by targeting DNMT3A in osteoarthritis. Biochem Biophys Res Commun 2019; 519:430-437. [DOI: 10.1016/j.bbrc.2019.09.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/07/2019] [Indexed: 12/26/2022]
|
509
|
Menarim BC, Gillis KH, Oliver A, Mason C, Ngo Y, Werre SR, Barrett SH, Luo X, Byron CR, Dahlgren LA. Autologous bone marrow mononuclear cells modulate joint homeostasis in an equine in vivo model of synovitis. FASEB J 2019; 33:14337-14353. [PMID: 31665925 DOI: 10.1096/fj.201901684rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is characterized by macrophage-driven synovitis. Macrophages promote synovial health but become inflammatory when their regulatory functions are overwhelmed. Bone marrow mononuclear cells (BMNCs) are a rich source of macrophage progenitors used for treating chronic inflammation and produce essential molecules for cartilage metabolism. This study investigated the response to autologous BMNC injection in normal and inflamed joints. Synovitis was induced in both radiocarpal joints of 6 horses. After 8 h, 1 inflamed radiocarpal and 1 normal tarsocrural joint received BMNC injection. Contralateral joints were injected with saline. Synovial fluid was collected at 24, 96, and 144 h for cytology, cytokine quantification, and flow cytometry. At 144 h, horses were euthanatized, joints were evaluated, and synovium was harvested for histology and immunohistochemistry. Four days after BMNC treatment, inflamed joints had 24% higher macrophage counts with 10% more IL-10+ cells than saline-treated controls. BMNC-treated joints showed gross and analytical improvements in synovial fluid and synovial membrane, with increasing regulatory macrophages and synovial fluid IL-10 concentrations compared with saline-treated controls. BMNC-treated joints were comparable to healthy joints histologically, which remained abnormal in saline-treated controls. Autologous BMNCs are readily available, regulate synovitis through macrophage-associated effects, and can benefit thousands of patients with OA.-Menarim, B. C., Gillis, K. H., Oliver, A., Mason, C., Ngo, Y., Werre, S. R., Barrett, S. H., Luo, X., Byron, C. R., Dahlgren, L. A. Autologous bone marrow mononuclear cells modulate joint homeostasis in an equine in vivo model of synovitis.
Collapse
Affiliation(s)
- Bruno C Menarim
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Kiersten H Gillis
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Andrea Oliver
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Caitlin Mason
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Ying Ngo
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Stephen R Werre
- Laboratory for Study Design and Statistical Analysis, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA; and
| | - Sarah H Barrett
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Xin Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Christopher R Byron
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Linda A Dahlgren
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
510
|
Kuang L, Wu J, Su N, Qi H, Chen H, Zhou S, Xiong Y, Du X, Tan Q, Yang J, Jin M, Luo F, Ouyang J, Zhang B, Wang Z, Jiang W, Chen L, Chen S, Wang Z, Liu P, Yin L, Guo F, Deng C, Chen D, Liu C, Xie Y, Ni Z, Chen L. FGFR3 deficiency enhances CXCL12-dependent chemotaxis of macrophages via upregulating CXCR7 and aggravates joint destruction in mice. Ann Rheum Dis 2019; 79:112-122. [PMID: 31662319 DOI: 10.1136/annrheumdis-2019-215696] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVES This study aims to investigate the role and mechanism of FGFR3 in macrophages and their biological effects on the pathology of arthritis. METHODS Mice with conditional knockout of FGFR3 in myeloid cells (R3cKO) were generated. Gait behaviours of the mice were monitored at different ages. Spontaneous synovial joint destruction was evaluated by digital radiographic imaging and μCT analysis; changes of articular cartilage and synovitis were determined by histological analysis. The recruitment of macrophages in the synovium was examined by immunostaining and monocyte trafficking assay. RNA-seq analysis, Western blotting and chemotaxis experiment were performed on control and FGFR3-deficient macrophages. The peripheral blood from non-osteoarthritis (OA) donors and patients with OA were analysed. Mice were treated with neutralising antibody against CXCR7 to investigate the role of CXCR7 in arthritis. RESULTS R3cKO mice but not control mice developed spontaneous cartilage destruction in multiple synovial joints at the age of 13 months. Moreover, the synovitis and macrophage accumulation were observed in the joints of 9-month-old R3cKO mice when the articular cartilage was not grossly destructed. FGFR3 deficiency in myeloid cells also aggravated joint destruction in DMM mouse model. Mechanically, FGFR3 deficiency promoted macrophage chemotaxis partly through activation of NF-κB/CXCR7 pathway. Inhibition of CXCR7 could significantly reverse FGFR3-deficiency-enhanced macrophage chemotaxis and the arthritic phenotype in R3cKO mice. CONCLUSIONS Our study identifies the role of FGFR3 in synovial macrophage recruitment and synovitis, which provides a new insight into the pathological mechanisms of inflammation-related arthritis.
Collapse
Affiliation(s)
- Liang Kuang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiangyi Wu
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Nan Su
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Huabing Qi
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Siru Zhou
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Xiong
- Department of Orthopedics, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Junjie Ouyang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zuqiang Wang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Wanling Jiang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Liang Chen
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuai Chen
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ziming Wang
- Department of Orthopedics, Daping Hospital, Army Medical University, Chongqing, China
| | - Peng Liu
- Department of Orthopedics, Daping Hospital, Army Medical University, Chongqing, China
| | - Liangjun Yin
- Department of Orthopaedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Fengjin Guo
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Di Chen
- Biochemistry, Rush University Medical Center, Chicago, Illinois, USA
| | - Chuanju Liu
- Department of Orthopaedic Surgery, New York University School of Medicine, New York University Medical Center, New York City, New York, USA
| | - Yangli Xie
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
511
|
Kivitz A, Kwong L, Shlotzhauer T, Lufkin J, Cinar A, Kelley S. A randomized, phase IIa study to assess the systemic exposure of triamcinolone acetonide following injection of extended-release triamcinolone acetonide or traditional triamcinolone acetonide into both knees of patients with bilateral knee osteoarthritis. Ther Adv Musculoskelet Dis 2019; 11:1759720X19881309. [PMID: 31662801 PMCID: PMC6796206 DOI: 10.1177/1759720x19881309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/16/2019] [Indexed: 12/27/2022] Open
Abstract
Background: Intra-articular corticosteroids are commonly used for pain relief in patients
with knee osteoarthritis. Simultaneous intra-articular corticosteroid (CS)
knee injections may be beneficial for the ~80–90% of patients who present
with, or develop, bilateral knee osteoarthritis, but concurrent injections
may increase systemic CS exposure and data on safety/tolerability are
lacking. Triamcinolone acetonide extended release (TA-ER) has shown
decreased systemic triamcinolone acetonide exposure compared with
traditional triamcinolone acetonide crystalline suspension (TAcs) after a
single knee injection in patients with knee osteoarthritis. This phase IIa
study was designed to assess the safety and systemic triamcinolone acetonide
exposure following injections of TA-ER or TAcs into each knee of patients
with bilateral knee osteoarthritis. Methods: Patients (⩾40 years) meeting American College of Rheumatology criteria for
knee osteoarthritis in both knees received concurrent single intra-articular
injections of TA-ER 32 mg or TAcs 40 mg into each knee (total: 64 mg and
80 mg, respectively) and were followed for 6 weeks. Safety was evaluated
based on treatment-emergent adverse events (TEAEs). Blood samples for
pharmacokinetic analysis were collected pre-injection, and at the following
postinjection time points: 1, 2, 3, 4, 5, 6, 8, 10, 12, and 24 h, and days
8, 15, 29, and 43. Results: Baseline characteristics were balanced between patients randomly assigned to
TA-ER (n = 12) or TAcs (n = 12). Both
treatments were well tolerated with comparable TEAE profiles. Peak plasma
triamcinolone acetonide concentrations (Cmax) were lower
following bilateral TA-ER injections [geometric mean, 2277.7 pg/ml (95% CI,
1602.13–3238.04)] compared with bilateral TAcs injections [7394.7 pg/ml
(2201.06–24,843.43)], with median times to Cmax (Tmax)
of 4.5 and 6.5 h, respectively. Conclusions: In patients with bilateral knee osteoarthritis, intra-articular injection of
TA-ER into both knees was well tolerated. Consistent with pharmacokinetic
profiles observed after a single knee injection, plasma triamcinolone
acetonide concentrations were lower after bilateral TA-ER injections
compared with the higher and more variable concentrations observed after
bilateral TAcs injections. ClinicalTrials.gov identifier: NCT03378076
Collapse
Affiliation(s)
- Alan Kivitz
- Altoona Center for Clinical Research, Duncansville, 175 Meadowbrook Lane, PO Box 1018, Duncansville, PA 16635, USA
| | - Louis Kwong
- Department of Orthopaedic Surgery, Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | | | - Amy Cinar
- Flexion Therapeutics, Inc., Burlington, MA, USA
| | | |
Collapse
|
512
|
Song EK, Jeon J, Jang DG, Kim HE, Sim HJ, Kwon KY, Medina-Ruiz S, Jang HJ, Lee AR, Rho JG, Lee HS, Kim SJ, Park CY, Myung K, Kim W, Kwon T, Yang S, Park TJ. ITGBL1 modulates integrin activity to promote cartilage formation and protect against arthritis. Sci Transl Med 2019; 10:10/462/eaam7486. [PMID: 30305454 DOI: 10.1126/scitranslmed.aam7486] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 09/20/2018] [Indexed: 11/02/2022]
Abstract
Developing and mature chondrocytes constantly interact with and remodel the surrounding extracellular matrix (ECM). Recent research indicates that integrin-ECM interaction is differentially regulated during cartilage formation (chondrogenesis). Integrin signaling is also a key source of the catabolic reactions responsible for joint destruction in both rheumatoid arthritis and osteoarthritis. However, we do not understand how chondrocytes dynamically regulate integrin signaling in such an ECM-rich environment. Here, we found that developing chondrocytes express integrin-β-like 1 (Itgbl1) at specific stages, inhibiting integrin signaling and promoting chondrogenesis. Unlike cytosolic integrin inhibitors, ITGBL1 is secreted and physically interacts with integrins to down-regulate activity. We observed that Itgbl1 expression was strongly reduced in the damaged articular cartilage of patients with osteoarthritis (OA). Ectopic expression of Itgbl1 protected joint cartilage against OA development in the destabilization of the medial meniscus-induced OA mouse model. Our results reveal ITGBL1 signaling as an underlying mechanism of protection against destructive cartilage disorders and suggest the potential therapeutic utility of targeting ITGBL1 to modulate integrin signaling in human disease.
Collapse
Affiliation(s)
- Eun Kyung Song
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.,Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Jimin Jeon
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Republic of Korea.,Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Republic of Korea.,CIRNO, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong Gil Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ha Eun Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Hyo Jung Sim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Keun Yeong Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Sofia Medina-Ruiz
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Hyun-Jun Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ah Reum Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Jun Gi Rho
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyun-Shik Lee
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Seok Jung Kim
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chan Young Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Taejoon Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Siyoung Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Republic of Korea. .,Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Republic of Korea.,CIRNO, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Tae Joo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea. .,Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| |
Collapse
|
513
|
Chen J, Wu X. MicroRNA-103 contributes to osteoarthritis development by targeting Sox6. Biomed Pharmacother 2019; 118:109186. [DOI: 10.1016/j.biopha.2019.109186] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/26/2019] [Accepted: 06/26/2019] [Indexed: 01/13/2023] Open
|
514
|
Ibrahim IK, Saba EKA, Saad NLM, Mohammed DYA. Relation of interleukin-15 with the severity of primary knee osteoarthritis. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2019. [DOI: 10.4103/err.err_42_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
515
|
Membrane-Free Stem Cell Components Inhibit Interleukin-1α-Stimulated Inflammation and Cartilage Degradation in vitro and in vivo: A Rat Model of Osteoarthritis. Int J Mol Sci 2019; 20:ijms20194869. [PMID: 31575035 PMCID: PMC6801847 DOI: 10.3390/ijms20194869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/20/2022] Open
Abstract
Membrane-free stem cell components (MFSCC) from basal adipose tissue-derived stem cells (ADSCs) are unknown for the treatment strategies in osteoarthritis (OA). OA has been considered to be associated with inflammatory damage and cartilage degradation. In this study, we intended to investigate the molecular mechanism of the anti-inflammation and cartilage protection effect of MFSCC in vitro (rat primary chondrocytes) and in vivo (rat OA model). The MFSCC treatment significantly inhibited interleukin-1α (IL-1α) stimulated inflammation and cartilage degradation. The MFSCC considerably reduced the levels of inflammatory factors such as iNOS, COX-2, NO, and PGE2 and was suppressed NF-κB and MAPKs signaling pathways in IL-1α-stimulated rat chondrocytes. Additionally, biomarkers of OA such as MMP-9, COMP, and CTX-II decreased in the monosodium iodoacetate (MIA)-induced rat OA model by MFSCC treatment. In conclusion, the MFSCC was established to suppress IL-1α induced inflammation and cartilage degradation in vitro and in vivo. These findings provide new insight for understanding OA therapy using membrane-free stem cell approaches.
Collapse
|
516
|
Zhang B, Chen H, Ouyang J, Xie Y, Chen L, Tan Q, Du X, Su N, Ni Z, Chen L. SQSTM1-dependent autophagic degradation of PKM2 inhibits the production of mature IL1B/IL-1β and contributes to LIPUS-mediated anti-inflammatory effect. Autophagy 2019; 16:1262-1278. [PMID: 31500508 DOI: 10.1080/15548627.2019.1664705] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Synovitis is implicated in the pathology of osteoarthritis (OA) and significantly contributes to the development of OA. As a noninvasive physical therapy, low-intensity pulsed ultrasound (LIPUS) has been reported to possess anti-inflammatory effect in recent years. However, the role of LIPUS on synovitis of OA and the underlying mechanisms are little known. The present study showed that LIPUS ameliorated synovial inflammation in destabilization of the medial meniscus (DMM) mouse model and air pouch model, and alleviated pain gait patterns of DMM mouse. LIPUS dramatically inhibited the production of mature IL1B/IL-1β (interleukin 1 beta) in vitro and in vivo. In addition, LIPUS upregulated the macroautophagy/autophagy level as well as accelerated the formation of an SQSTM1 (sequestosome1)-PKM (pyruvate kinase, muscle) complex in the lipopolysaccharide (LPS)-adenosine triphosphate (ATP)-treated macrophages. Besides, LIPUS downregulated the level of PKM2 in LPS-ATP-treated macrophages, which could be reversed by SQSTM1 knockdown. In brief, the present study for the first time demonstrates that LIPUS inhibits the production of mature IL1B partially via SQSTM1-dependent autophagic degradation of PKM2 in LPS-ATP-treated macrophages, which may further ameliorate the synovial inflammation and gait patterns in animal models. Our data provide new clues for the treatments of synovitis and other inflammatory diseases using LIPUS. ABBREVIATIONS 3-MA: 3-methyladenene; ATG7: autophagy-related 7; ATP: adenosine triphosphate; BafA1: bafilomycin A1; BMDMs: bone marrow derived macrophages; CHX: cycloheximide; DMM: destabilization of the medial meniscus; ELISA: enzyme-linked immunosorbent assay; GFP: green fluorescent protein; IL1B/IL-1β: interleukin 1 beta; LIPUS: low-intensity pulsed ultrasound; LIR: LC3-interacting region; LPS: lipopolysaccharide; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MDP: muramyl dipeptide; NFKB/NF-κB: nuclear factor kappa B; NLRP3: NLR family, pyrin domain containing 3; OA: osteoarthritis; PKM/PKM2: pyruvate kinase M1/2; PMA: phorbol-12-myristate-13-acetate; PYCARD/ASC; PYD and CARD domain containing; RFP: red fluorescent protein; siRNAs: small interfering RNAs; SQSTM1: sequestosome 1; TEM: transmission electron microscopy.
Collapse
Affiliation(s)
- Bin Zhang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Hangang Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Junjie Ouyang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Liang Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Qiaoyan Tan
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Nan Su
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Zhenhong Ni
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| |
Collapse
|
517
|
Isorhapontigenin Suppresses Interleukin-1β-Induced Inflammation and Cartilage Matrix Damage in Rat Chondrocytes. Inflammation 2019; 42:2278-2285. [DOI: 10.1007/s10753-019-01092-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
518
|
Wang S, Pillinger MH, Krasnokutsky S, Barbour KE. The association between asymptomatic hyperuricemia and knee osteoarthritis: data from the third National Health and Nutrition Examination Survey. Osteoarthritis Cartilage 2019; 27:1301-1308. [PMID: 31158484 PMCID: PMC6702067 DOI: 10.1016/j.joca.2019.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 05/02/2019] [Accepted: 05/22/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In vitro and clinical studies suggest that urate may contribute to osteoarthritis (OA) risk. We tested the associations between hyperuricemia and knee OA, and examined the role of obesity, using a cross-sectional, nationally representative dataset. METHOD National Health and Nutrition Examination Survey (NHANES) III used a multistage, stratified probability cluster design to select USA civilians from 1988 to 1994. Using NHANES III we studied adults >60 years, with or without hyperuricemia (serum urate > 6.8 mg/dL), excluding individuals with gout (i.e., limiting to asymptomatic hyperuricemia (AH)). Radiographic knee OA (RKOA) was defined as Kellgren-Lawrence grade ≥ 2 in any knee, and symptomatic radiographic knee osteoarthritis (RKOA) (sRKOA) was defined as RKOA plus knee pain (most days for 6 weeks) in the same knee. RESULTS AH prevalence was 17.9% (confidence interval (CI) 15.3-20.5). RKOA prevalence was 37.7% overall (CI 35.0-40.3), and was 44.0% for AH vs 36.3% for normouricemic adults (p = 0.056). symptomatic radiographic knee osteoarthritis (sRKOA) was more prevalent in AH vs normouricemic adults (17.4% vs 10.9%, p = 0.046). In multivariate models adjusting for obesity, model-based associations between AH and knee OA were attenuated (for RKOA, prevalence ratio (PR) = 1.14, 95% CI 0.95, 1.36; for sRKOA, PR = 1.40, 95% CI 0.98, 2.01). In stratified multivariate analyses, AH was associated with sRKOA in adults without obesity (PR = 1.66, 95% CI 1.02, 2.71) but not adults with obesity (PR = 1.21, 95% CI 0.66, 2.23). CONCLUSIONS Among adults aged 60 or older, AH is associated with knee OA risk that is more apparent in adults without obesity.
Collapse
Affiliation(s)
- Shudan Wang
- Division of Rheumatology, Department of Medicine, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY,Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, NY, USA,VA New York Harbor Health Care System, New York Campus, New York, NY, USA
| | - Michael H. Pillinger
- Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, NY, USA,VA New York Harbor Health Care System, New York Campus, New York, NY, USA
| | - Svetlana Krasnokutsky
- Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, NY, USA
| | - Kamil E. Barbour
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
519
|
Seidenstuecker M, Watrinet J, Bernstein A, Suedkamp NP, Latorre SH, Maks A, Mayr HO. Viscoelasticity and histology of the human cartilage in healthy and degenerated conditions of the knee. J Orthop Surg Res 2019; 14:256. [PMID: 31409382 PMCID: PMC6693159 DOI: 10.1186/s13018-019-1308-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/05/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are many studies on osteoarthritis, but only a few studies deal with human arthrosis, comparing the mechanical properties of healthy and diseased samples. In most of these studies, only isolated areas of the tibia are examined. There is currently only one study investigating the complete mapping of cartilage tissue but not the difference between instantaneous modulus (IM) in healthy and diseased samples. The aim of this study is to investigate the relationship between the biomechanical and histological changes of articular cartilage in the pathogenesis of osteoarthritis. METHODS The study compared 25 tibiae with medial gonarthrosis and 13 healthy controls. The IM was determined by automated indentation mapping using a Mach-1 V500css testing machine. A grid was projected over the sample and stored so that all measurements could be taken at the same positions (100 ± 29 positions across the tibiae). This grid was then used to perform the thickness measurement using the needle method. Samples were then taken for histological examinations using a hollow milling machine. Then Giemsa and Safranin O staining were performed. In order to determine the degree of arthrosis according to histological criteria, the assessment was made with regard to Osteoarthritis Research Society International (OARSI) and AHO scores. RESULTS A significant difference (p < 0.05) could be observed in the measured IM between the controls with 3.43 ± 0.36 MPa and the samples with 2.09 ± 0.18 MPa. In addition, there was a significant difference in IM in terms of meniscus-covered and meniscus-uncovered areas. The difference in cartilage thickness between 2.25 ± 0.11 mm controls and 2.0 ± 0.07 mm samples was highly significant with p < 0.001. With regard to the OARSI and AHO scores, the samples differed significantly from the controls. The OARSI and AHO scores showed a significant difference between meniscus-covered and meniscus-uncovered areas. CONCLUSIONS The controls showed significantly better viscoelastic behavior than the arthrotic samples in the measured IM. The measured biomechanical values showed a direct correlation between histological changes and altered biomechanics in gonarthrosis.
Collapse
Affiliation(s)
- Michael Seidenstuecker
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany.
| | - Julius Watrinet
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Anke Bernstein
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Norbert P Suedkamp
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Sergio H Latorre
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Anastasija Maks
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Hermann O Mayr
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany.,Schoen Clinic Munich Harlaching, Teaching Hospital of Paracelsus Medical University Salzburg, Salzburg, Austria
| |
Collapse
|
520
|
Choi MC, Jo J, Park J, Kang HK, Park Y. NF-κB Signaling Pathways in Osteoarthritic Cartilage Destruction. Cells 2019; 8:cells8070734. [PMID: 31319599 PMCID: PMC6678954 DOI: 10.3390/cells8070734] [Citation(s) in RCA: 313] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a type of joint disease associated with wear and tear, inflammation, and aging. Mechanical stress along with synovial inflammation promotes the degradation of the extracellular matrix in the cartilage, leading to the breakdown of joint cartilage. The nuclear factor-kappaB (NF-κB) transcription factor has long been recognized as a disease-contributing factor and, thus, has become a therapeutic target for OA. Because NF-κB is a versatile and multi-functional transcription factor involved in various biological processes, a comprehensive understanding of the functions or regulation of NF-κB in the OA pathology will aid in the development of targeted therapeutic strategies to protect the cartilage from OA damage and reduce the risk of potential side-effects. In this review, we discuss the roles of NF-κB in OA chondrocytes and related signaling pathways, including recent findings, to better understand pathological cartilage remodeling and provide potential therapeutic targets that can interfere with NF-κB signaling for OA treatment.
Collapse
Affiliation(s)
- Moon-Chang Choi
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea.
| | - Jiwon Jo
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea
| | - Jonggwan Park
- Department of Bioinformatics, Kongju National University, Kongju 38065, Korea
| | - Hee Kyoung Kang
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea
| | - Yoonkyung Park
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea.
| |
Collapse
|
521
|
Intra-articular targeting of nanomaterials for the treatment of osteoarthritis. Acta Biomater 2019; 93:239-257. [PMID: 30862551 DOI: 10.1016/j.actbio.2019.03.010] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/28/2019] [Accepted: 03/06/2019] [Indexed: 12/31/2022]
Abstract
Osteoarthritis is a prevalent and debilitating disease that involves pathological contributions from numerous joint tissues and cells. The joint is a challenging arena for drug delivery, since the joint has poor bioavailability for systemically administered drugs and experiences rapid clearance of therapeutics after intra-articular injection. Moreover, each tissue within the joint presents unique barriers to drug localization. In this review, the various applications of nanotechnology to overcome these drug delivery limitations are investigated. Nanomaterials have reliably shown improvements to retention profiles of drugs within the joint space relative to injected free drugs. Additionally, nanomaterials have been modified through active and passive targeting strategies to facilitate interactions with and localization within specific joint tissues such as cartilage and synovium. Last, the limitations of drawing cross-study comparisons, the implications of synovial fluid, and the potential importance of multi-modal therapeutic strategies are discussed. As emerging, cell-specific disease modifying osteoarthritis drugs continue to be developed, the need for targeted nanomaterial delivery will likely become critical for effective clinical translation of therapeutics for osteoarthritis. STATEMENT OF SIGNIFICANCE: Improving drug delivery to the joint is a pressing clinical need. Over 27 million Americans live with osteoarthritis, and this figure is continuously expanding. Numerous drugs have been investigated but have failed in clinical trials, likely related to poor bioavailability to target cells. This article comprehensively reviews the advances in nano-scale delivery vehicles designed to overcome the delivery barriers in the joint. This is the first review to analyze active and passive targeting strategies systematically for different target sites while also delineating between tissue homing and whole joint retention. By bringing together the lessons learned across numerous nano-scale platforms, researchers may be able to hone future nanomaterial designs, allowing emerging therapeutics to perform with clinically relevant efficacy and disease modifying potential.
Collapse
|
522
|
Ni Z, Kuang L, Chen H, Xie Y, Zhang B, Ouyang J, Wu J, Zhou S, Chen L, Su N, Tan Q, Luo X, Chen B, Chen S, Yin L, Huang H, Du X, Chen L. The exosome-like vesicles from osteoarthritic chondrocyte enhanced mature IL-1β production of macrophages and aggravated synovitis in osteoarthritis. Cell Death Dis 2019; 10:522. [PMID: 31285423 PMCID: PMC6614358 DOI: 10.1038/s41419-019-1739-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023]
Abstract
Synovitis, a common clinical symptom for osteoarthritis (OA) patients, is highly related to OA pathological progression and pain manifestation. The activated synovial macrophages have been demonstrated to play an important role in synovitis, but the mechanisms about macrophage activation are still not clear. In this study, we found that the exosome-like vesicles from osteoarthritic chondrocytes could be a new biological factor to stimulate inflammasome activation and increase mature IL-1β production in macrophages. The degraded cartilage explants produced more exosome-like vesicles than the nondegraded ones, while the exosome-like vesicles from chondrocytes could enter into joint synovium tissue and macrophages. Moreover, the exosome-like vesicles from osteoarthritic chondrocytes enhanced the production of mature IL-1β in macrophages. These vesicles could inhibit ATG4B expression via miR-449a-5p, leading to inhibition of autophagy in LPS-primed macrophages. The decreased autophagy promoted the production of mitoROS, which further enhanced the inflammasome activation and subsequent IL-1β processing. Ultimately, the increase of mature IL-1β may aggravate synovial inflammation and promote the progression of OA disease. Our study provides a new perspective to understand the activation of synovial macrophages and synovitis in OA patients, which may be beneficial for therapeutic intervention in synovitis-related OA patients.
Collapse
Affiliation(s)
- Zhenhong Ni
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Liang Kuang
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Hangang Chen
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Yangli Xie
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Bin Zhang
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Junjie Ouyang
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Jiangyi Wu
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Siru Zhou
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Liang Chen
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Nan Su
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - QiaoYan Tan
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Xiaoqing Luo
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Bo Chen
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Shuai Chen
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Liangjun Yin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, 400010, Chongqing, China
| | - Haiyang Huang
- Department of Orthopedic Surgery, Qianjiang Nationality Hospital, 409000, Chongqing, China
| | - Xiaolan Du
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Lin Chen
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China.
| |
Collapse
|
523
|
Charlier E, Deroyer C, Ciregia F, Malaise O, Neuville S, Plener Z, Malaise M, de Seny D. Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol 2019; 165:49-65. [DOI: 10.1016/j.bcp.2019.02.036] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
|
524
|
陈 永, 邱 富, 朱 兴, 莫 海, 吴 自, 肖 长. [Pannus does not occur only in rheumatoid arthritis: a pathological observation of pannus of knee osteoarthritis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:747-750. [PMID: 31270057 PMCID: PMC6743915 DOI: 10.12122/j.issn.1673-4254.2019.06.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To compare the histopathological features of the synovium between rheumatoid arthritis (RA) and osteoarthritis (OA). METHODS We retrospectively analyzed the synovial specimens obtained after synovial surgery in 72 cases of RA and 24 cases of OA. Two independent pathologists reviewed the sections of the synovial tissues with HE staining, quantitatively scored the degree of fibroblast-like synoviocyte (FLS) hyperplasia, vascular hyperplasia, fibroplasia, and lymphocyte infiltration, and examined the presence plasma cell infiltration. The pathological morphology of the synovial tissues was evaluated in relation with the clinical data of the patients. RESULTS Pannus formation was also detected in the synovium of OA patients, which showed a lesser degree of OA-FLS hyperplasia, fibrosis and lymphocyte infiltration and a significantly lower rate of plasma cell infiltration compared with the pannus in RA patients. Vascular proliferation was also milder in the pannus of OA patients than in RA pannus, but the difference was not statistically significant. In OA patients, the pannus could be observed under a microscope and was difficult to distinguish from that in RA patients. CONCLUSIONS Pannus formation occurs also in the synovium of OA patients but with milder FLS hyperplasia, fibrosis and lymphocyte infiltration and a lower rate of plasma cell infiltration compared with the pannus in RA patients. These differences in the pannus between OA and RA can be of potential value in the diagnosis and treatment of the patients.
Collapse
Affiliation(s)
- 永 陈
- 南方医科大学中西医结合医院 风湿免疫科,广东 广州 510330Department of Rheumatology, Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510330, China
| | - 富娟 邱
- 南方医科大学中西医结合医院 风湿免疫科,广东 广州 510330Department of Rheumatology, Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510330, China
| | - 兴旺 朱
- 南方医科大学中西医结合医院 风湿免疫科,广东 广州 510330Department of Rheumatology, Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510330, China
| | - 海月 莫
- 南方医科大学中西医结合医院 病理科,广东 广州 510330Department of Pathology, Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510330, China
| | - 自勍 吴
- 南方医科大学中西医结合医院 病理科,广东 广州 510330Department of Pathology, Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510330, China
| | - 长虹 肖
- 南方医科大学中西医结合医院 风湿免疫科,广东 广州 510330Department of Rheumatology, Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510330, China
| |
Collapse
|
525
|
Lee GJ, Cho IA, Oh JS, Seo YS, You JS, Kim SG, Kim JS. Anticatabolic Effects of Morin through the Counteraction of Interleukin-1β-Induced Inflammation in Rat Primary Chondrocytes. Cells Tissues Organs 2019; 207:21-33. [PMID: 31256148 DOI: 10.1159/000500323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/12/2019] [Indexed: 11/19/2022] Open
Abstract
Morin, a flavonoid isolated from various medicinal herbal plants, has an anti-inflammatory effect. This study aimed to elucidate the anticatabolic effects and cellular mechanism of morin against interleukin-1β (IL-1β) in rat primary chondrocytes. Morin at 10-100 μM did not affect the viability of rat primary chondrocytes. Treatment with morin for 21 days ameliorated the IL-1β-induced decrease in extracellular matrix. Furthermore, treatment with morin attenuated IL-1β-induced proteoglycan loss in the articular cartilage through suppression of catabolic factors, such as matrix metalloproteinases, inflammatory mediators, and pro-inflammatory cytokines. These data indicated that morin exerted anticatabolic effects that can prevent and reduce progressive degeneration of the articular cartilage, and thus may be a potential candidate treatment for osteoarthritis.
Collapse
Affiliation(s)
- Gyeong-Je Lee
- Department of Prosthodontics, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - In-A Cho
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Ji-Su Oh
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Yo-Seob Seo
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Jae-Seek You
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Su-Gwan Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Jae-Sung Kim
- Pre-Dentistry, School of Dentistry, Chosun University, Gwangju, Republic of Korea,
| |
Collapse
|
526
|
Abstract
PURPOSE OF THE REVIEW Osteoarthritis is widely regarded as a spectrum of conditions that affect all joint tissues, typified by a common entity: cartilage loss. Here, we review recent progress and challenges in chondroprotection and discuss new strategies to prevent cartilage loss in osteoarthritis. RECENT FINDINGS Advances in clinical, molecular, and cellular characterization are enabling improved stratification of osteoarthritis subtypes. Integration of next-generation sequencing and "omics" approaches with clinically relevant readouts shows promise in delineating both subtypes of disease and meaningful trial end points. Novel delivery strategies are enabling joint-specific delivery. Chondroprotection requires a whole joint approach, stratification of patient groups, and use of patient-relevant end points. Drug development should continue to explore new targets, while using modern technologies and recent knowledge to re-visit unsuccessful therapeutics from the past. The overarching goal for chondroprotection is to provide the right treatment(s) for the right patient at the right time.
Collapse
Affiliation(s)
- Jolet Y Mimpen
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, OX3 7LD, UK
| | - Sarah J B Snelling
- The Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
527
|
Li YN, Fan ML, Liu HQ, Ma B, Dai WL, Yu BY, Liu JH. Dihydroartemisinin derivative DC32 inhibits inflammatory response in osteoarthritic synovium through regulating Nrf2/NF-κB pathway. Int Immunopharmacol 2019; 74:105701. [PMID: 31228817 DOI: 10.1016/j.intimp.2019.105701] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/05/2019] [Accepted: 06/13/2019] [Indexed: 12/30/2022]
Abstract
Synovitis is an aseptic inflammation that leads to joint effusion, pain and swelling. As one of the main drivers of pathogenesis in osteoarthritis (OA), the presence of synovitis contributes to pain, incidence and progression of OA. In our previous study, DC32 [(9α,12α-dihydroartemisinyl) bis(2'-chlorocinnmate)], a dihydroartemisinin derivative, was found to have an antirheumatic ability via immunosuppression, but the effect of DC32 on synovitis has not been fully illuminated. In this study, we chose to evaluate the effect and mechanism of DC32 on attenuating synovial inflammation. Fibroblast-like synoviocytes (FLSs) of papain-induced OA rats were isolated and cultured. And DC32 significantly inhibited the invasion and migration of cultured OA-FLSs, as well as the transcription of IL-6, IL-1β, CXCL12 and CX3CL1 in cultured OA-FLSs measured by qPCR. DC32 remarkably inhibited the activation of ERK and NF-κB pathway, increased the expression of Nrf2 and HO-1 in cultured OA-FLSs detected by western blot. DC32 inhibited the degradation and phosphorylation of IκBα which further prevented the phosphorylation of NF-κB p65 and the effect of DC32 could be relieved by siRNA for Nrf2. In papain-induced OA mice, DC32 significantly alleviated papain-induced mechanical allodynia, knee joint swelling and infiltration of inflammatory cell in synovium. DC32 upregulated the mRNA expression of Type II collagen and aggrecan, and downregulated the mRNA expression of MMP2, MMP3, MMP13 and ADAMTS-5 in the knee joints of papain-induced OA mice measured by qPCR. The level of TNF-α in the serum and secretion of TNF-α in the knee joints were also reduced by DC32 in papain-induced OA mice. In conclusion, DC32 inhibited the inflammatory response in osteoarthritic synovium through regulating Nrf2/NF-κB pathway and attenuated OA. In this way, DC32 may be a potential agent in the treatment of OA.
Collapse
Affiliation(s)
- Ya-Nan Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meng-Lin Fan
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Han-Qing Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Bin Ma
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wen-Ling Dai
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Bo-Yang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Ji-Hua Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
528
|
Shorter E, Sannicandro AJ, Poulet B, Goljanek-Whysall K. Skeletal Muscle Wasting and Its Relationship With Osteoarthritis: a Mini-Review of Mechanisms and Current Interventions. Curr Rheumatol Rep 2019; 21:40. [PMID: 31203463 PMCID: PMC6571089 DOI: 10.1007/s11926-019-0839-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a subset of joint disorders resulting in degeneration of synovial joints. This leads to pain, disability and loss of independence. Knee and hip OA are extremely prevalent, and their occurrence increases with ageing. Similarly, loss of muscle mass and function, sarcopenia, occurs during ageing. RECENT FINDINGS Little is known about the impact of muscle wasting on OA progression; nevertheless, it has been suggested that muscle wasting directly affects the stability of the joints and loss of mobility leads to gradual degeneration of articular cartilage. The molecular mechanisms underlying muscle wasting in OA are not well understood; however, these are probably related to changes in gene expression, as well as epigenetic modifications. It is becoming clear that skeletal muscle wasting plays an important role in OA development and/or progression. Here, we discuss mechanisms, current interventions, such as exercise, and potentially novel approaches, such as modulation of microRNAs, aiming at ameliorating OA symptoms through maintaining muscle mass and function.
Collapse
Affiliation(s)
- Emily Shorter
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, West Derby Road, Liverpool, L7 8TX UK
| | - Anthony J Sannicandro
- Department of Physiology, School of Medicine, REMEDI, NUI Galway, Human Biology Building, University Road, Galway, Ireland
| | - Blandine Poulet
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, West Derby Road, Liverpool, L7 8TX UK
| | - Katarzyna Goljanek-Whysall
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, West Derby Road, Liverpool, L7 8TX UK
- Department of Physiology, School of Medicine, REMEDI, NUI Galway, Human Biology Building, University Road, Galway, Ireland
| |
Collapse
|
529
|
Haraden CA, Huebner JL, Hsueh MF, Li YJ, Kraus VB. Synovial fluid biomarkers associated with osteoarthritis severity reflect macrophage and neutrophil related inflammation. Arthritis Res Ther 2019; 21:146. [PMID: 31196179 PMCID: PMC6567574 DOI: 10.1186/s13075-019-1923-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/22/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND To identify a synovial fluid (SF) biomarker profile characteristic of individuals with an inflammatory osteoarthritis (OA) endotype. METHODS A total of 48 knees (of 25 participants) were characterized for an extensive array of SF biomarkers quantified by Rules Based Medicine using the high-sensitivity multiplex immunoassay, Myriad Human InflammationMAP® 1.0, which included 47 different cytokines, chemokines, and growth factors related to inflammation. Multivariable regression with generalized estimating equations (GEE) and false discovery rate (FDR) correction was used to assess associations of SF RBM biomarkers with etarfolatide imaging scores reflecting synovial inflammation; radiographic knee OA severity (based on Kellgren-Lawrence (KL) grade, joint space narrowing, and osteophyte scores); knee joint symptoms; and SF biomarkers associated with activated macrophages and knee OA progression including CD14 and CD163 (shed by activated macrophages) and elastase (shed by activated neutrophils). RESULTS Significant associations of SF biomarkers meeting FDR < 0.05 included soluble (s)VCAM-1 and MMP-3 with synovial inflammation (FDR-adjusted p = 0.025 and 1.06 × 10-7); sVCAM-1, sICAM-1, TIMP-1, and VEGF with radiographic OA severity (p = 1.85 × 10-5 to 3.97 × 10-4); and VEGF, MMP-3, TIMP-1, sICAM-1, sVCAM-1, and MCP-1 with OA symptoms (p = 2.72 × 10-5 to 0.050). All these SF biomarkers were highly correlated with macrophage markers CD163 and CD14 in SF (r = 0.43 to 0.90, FDR < 0.05); all but MCP-1 were also highly correlated with neutrophil elastase in SF (r = 0.62 to 0.89, FDR < 0.05). CONCLUSIONS A subset of six SF biomarkers was related to synovial inflammation in OA, as well as radiographic and symptom severity. These six OA-related SF biomarkers were specifically linked to indicators of activated macrophages and neutrophils. These results attest to an inflammatory OA endotype that may serve as the basis for therapeutic targeting of a subset of individuals at high risk for knee OA progression. TRIAL REGISTRATION Written informed consent was received from participants prior to inclusion in the study; the study was registered at ClinicalTrials.gov ( NCT01237405 ) on November 9, 2010, prior to enrollment of the first participant.
Collapse
Affiliation(s)
- Collin A Haraden
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA
| | - Janet L Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA
| | - Ming-Feng Hsueh
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA
| | - Yi-Ju Li
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA.,Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA. .,Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
530
|
Li X, Yu M, Han L, Chen L, Zhang D, Zhou G, Zhao Q, Sun T. LINC00305 represses miR-124 expression to trigger inflammatory insults in the presence of lipopolysaccharide. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2352-2360. [PMID: 31190561 DOI: 10.1080/21691401.2019.1624559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Xiaoyan Li
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Manqiu Yu
- Department of Trauma Reconstruction, Shandong Wendeng Orthopaedic & Traumatic Hospital Weihai, China
| | - Liang Han
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lei Chen
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Daxue Zhang
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guannan Zhou
- Emergency Department, Jining No. 1 People’s Hospital, Jining, China
| | - Qiang Zhao
- Department of Orthopaedics, Jining No.1 People’s Hospital, Jining, China
| | - Taitao Sun
- Department of Orthopaedics, Jining No.1 People’s Hospital, Jining, China
- Department of Orthopaedics, Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining, China
| |
Collapse
|
531
|
Fleischmann RM, Bliddal H, Blanco FJ, Schnitzer TJ, Peterfy C, Chen S, Wang L, Feng S, Conaghan PG, Berenbaum F, Pelletier J, Martel‐Pelletier J, Vaeterlein O, Kaeley GS, Liu W, Kosloski MP, Levy G, Zhang L, Medema JK, Levesque MC. A Phase
II
Trial of Lutikizumab, an Anti–Interleukin‐1α/β Dual Variable Domain Immunoglobulin, in Knee Osteoarthritis Patients With Synovitis. Arthritis Rheumatol 2019; 71:1056-1069. [DOI: 10.1002/art.40840] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 01/10/2019] [Indexed: 12/30/2022]
Affiliation(s)
| | - Henning Bliddal
- Bispebjerg‐Frederiksberg Hospital and University of Copenhagen Copenhagen Denmark
| | | | | | | | - Su Chen
- AbbVie Inc. North Chicago Illinois
| | - Li Wang
- AbbVie Inc. North Chicago Illinois
| | | | | | - Francis Berenbaum
- Sorbonne UniversitéINSERM, and AP‐HP Hospital Saint‐Antoine Paris France
| | | | | | | | | | - Wei Liu
- AbbVie Inc. North Chicago Illinois
| | | | | | | | | | | |
Collapse
|
532
|
Bernardo-Bueno MM, Gonzalez-Suarez CB, Malvar AK, Cua R, Feliciano D, Tan-Sales BG, Aycardo SMO, Tan-Ong M, Chan R, De Los Reyes F. Stratifying Minimal Versus Severe Pain in Knee Osteoarthritis Using a Musculoskeletal Ultrasound Protocol. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2019; 38:1411-1423. [PMID: 30239017 DOI: 10.1002/jum.14819] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE The aim of this cross-sectional correlational study was to determine the association of pain with morphologic and inflammatory sonographic findings in patients with knee osteoarthritis. METHODS A total of 113 participants with knee osteoarthritis were assessed using visual analog scale pain score and sonography. Ultrasound evaluation included morphologic changes (ie, articular cartilage degeneration, medial and lateral meniscal protrusion, and presence of osteophytes on the joint margins) and inflammatory changes (ie suprapatellar effusion and/or synovitis, Baker cyst, superficial and deep infrapatellar effusion, pes anserine tendinopathy, and Hoffa panniculitis). RESULTS Cluster analysis via Ward's method grouped patients with minimal pain (visual analog scale score, 0-4) and with substantial pain (visual analog scale score, 5-10). Stepwise logistic regression yielded 5 variables that significantly explained the variation in the probability of perceived substantial pain at 10% level of significance: lateral cartilage clarity (LCC; P = .025), medial cartilage clarity (MCC; P = .20), medial cartilage thickness (MCT; P = .041), medial meniscus protrusion (MMP) (P = .029), and osteophytes at medial femoral margin (P = .082), with 63% overall prediction accuracy. When age and sex were added, 4 variables remained significant at a 10% level of significance: LCC, MCC, MCT, and MMP, with 65% overall prediction accuracy. The receiver operating characteristic curve of this model was 0.667. CONCLUSION The study was able to demonstrate that morphologic abnormalities in the ultrasound parameters for LCC, MCC, MCT, and MMP were able to predict significant joint pain in knee osteoarthritis. There were no inflammatory changes that contributed to significant joint pain in this study.
Collapse
Affiliation(s)
- Mary Monica Bernardo-Bueno
- Department of Physical Medicine and Rehabilitation, University of Santo Tomas Hospital
- Department of Physical Medicine and Rehabilitation, Our Lady of Lourdes Hospital, Manila, Philippines
- East Avenue Medical Center Quezon City, Philippines College of Rehabilitation Sciences, University of Santo Tomas, Manila, Philippines
| | - Consuelo B Gonzalez-Suarez
- Department of Physical Medicine and Rehabilitation, University of Santo Tomas Hospital
- Department of Physical Medicine and Rehabilitation, Our Lady of Lourdes Hospital, Manila, Philippines
| | | | - Ronald Cua
- Department of Physical Medicine and Rehabilitation, University of Santo Tomas Hospital
| | - Daniel Feliciano
- Department of Physical Medicine and Rehabilitation, University of Santo Tomas Hospital
| | - Bee Giok Tan-Sales
- Department of Physical Medicine and Rehabilitation, University of Santo Tomas Hospital
| | | | - Millicent Tan-Ong
- Faculty of Medicine and Surgery, Department of Medicine, Section of Rheumatology, Clinical Immunology and Osteoporosis, University of Santo Tomas, Manila Philippines
| | - Robert Chan
- Department of Orthopedic Surgery, University of Santo Tomas Hospital
| | | |
Collapse
|
533
|
Szentesi M, Nagy Z, Géher P, Papp I, Kampen WU. A prospective observational study on the long-term results of 90Yttrium citrate radiosynoviorthesis of synovitis in osteoarthritis of the knee joint. Eur J Nucl Med Mol Imaging 2019; 46:1633-1641. [DOI: 10.1007/s00259-019-04350-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/16/2019] [Indexed: 01/21/2023]
|
534
|
Diaz-Rodriguez P, Erndt-Marino J, Chen H, Diaz-Quiroz JF, Samavedi S, Hahn MS. A Bioengineered In Vitro Osteoarthritis Model with Tunable Inflammatory Environments Indicates Context-Dependent Therapeutic Potential of Human Mesenchymal Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00109-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
535
|
Karateev AE, Lila AM, Pogozheva EY, Amirdzhanova VN, Filatova ES. [What factors affect the effectiveness of long - term analgesic therapy for osteoarthritis? Data analysis of the multi - center 3-month PARACELSUS study]. TERAPEVT ARKH 2019; 91:68-75. [PMID: 32598679 DOI: 10.26442/00403660.2019.05.000233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 12/16/2022]
Abstract
There are factors that can affect the effectiveness of treatment of osteoarthritis (OA). Aim to identify factors affecting the effectiveness of long - term analgesic therapy in OA. Materials and methods. The study included 6448 patients (70.9% female and 29.1% male), middle age 57.8±10.2 years, with severe pain [≥40 mm on the visual analog scale]. All patients received the preparation of avocado - soybean unsaponifiables (ASU) 300 mg/day. For pain relief at the beginning and during the study, the drug Ketoprofen lysine salt (KLS) 320 mg/day was used. The efficiency criterion was pain reduction ≥50% and satisfaction with treatment ≥4 on a 5-point scale. The influence of a number of factors on the result of treatment was evaluated. Results. For 3 months of treatment, the pain decreased from 63.7±12.0 to 14.2±11.7 mm VAS. The result was evaluated as "good" or "excellent" 81.7% of patients. Adverse reactions were rare. In total, a good response to therapy was noted in 87.4% of patients. Gender, body mass index ≥30 kg/m2, type 2 diabetes mellitus, poor effect of non - steroidal anti - inflammatory drugs (NSAIDs) and Symptomatic Slow-Acting Drugs in Osteoarthritis (SYSADOA) in history did not affect the result. The effect was lower in persons >65 years [odds ratio (OR) 0.418; 95% confidence interval (CI) 0.342-0.509, p2 by Kellgren-Lawrence (OR 0.556; 95% CI 0.298-0.738, p.
Collapse
Affiliation(s)
- A E Karateev
- V.A. Nasonova Scientific and Research Institute of Rheumatology
| | - A M Lila
- V.A. Nasonova Scientific and Research Institute of Rheumatology
| | - E Y Pogozheva
- V.A. Nasonova Scientific and Research Institute of Rheumatology
| | | | - E S Filatova
- V.A. Nasonova Scientific and Research Institute of Rheumatology
| |
Collapse
|
536
|
Udomsinprasert W, Jittikoon J, Honsawek S. Interleukin-34 as a promising clinical biomarker and therapeutic target for inflammatory arthritis. Cytokine Growth Factor Rev 2019; 47:43-53. [PMID: 31126875 DOI: 10.1016/j.cytogfr.2019.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/09/2019] [Indexed: 01/15/2023]
Abstract
Interleukin-34 (IL-34), recently identified as a novel inflammatory cytokine and the second ligand for colony-stimulating factor-1 receptor, is known to play regulatory roles in the development, maintenance, and function of mononuclear phagocyte lineage cells - especially osteoclasts. Regarding its primary effect on osteoclasts, IL-34 has been shown to stimulate formation and activation of osteoclasts, which in turn magnifies osteoclasts-resorbing activity. In addition to its role in osteoclastogenesis, IL-34 has been implicated in inflammation of synovium via augmenting production of inflammatory mediators, in which altered IL-34 expression is regulated by pro-inflammatory cytokines responsible for cartilage degradation. Indeed, IL-34 has been documented to be highly expressed in inflamed synovium of rheumatoid arthritis (RA) and knee osteoarthritis (OA) patients, which are recognized as inflammatory arthritis. Furthermore, a number of clinical studies demonstrated that IL-34 levels were significantly increased in the circulation and synovial fluid of patients with RA and knee OA. Its levels were also found to be positively associated with disease severity - especially radiographic severity of both RA and knee OA patients. Interestingly, emerging evidence has accumulated that functional blockage of IL-34 with specific antibody can alleviate the severity of inflammatory arthritis. It is therefore reasonable to speculate that IL-34 may be developed as a potential biomarker and a new therapeutic candidate for inflammatory arthritis. To date, there are numerous studies showing IL-34 involvement and association with many aspects of inflammatory arthritis. Herein, this review aimed to summarize the recent findings regarding regulatory role of IL-34 in synovial inflammation-mediated cartilage destruction and update the current comprehensive knowledge on usefulness of IL-34-based treatment in inflammatory arthritis - particularly RA and knee OA.
Collapse
Affiliation(s)
| | - Jiraphun Jittikoon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Sittisak Honsawek
- Department of Biochemistry, Osteoarthritis and Musculoskeleton Research Unit, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
537
|
Rasheed Z, Rasheed N, Abdulmonem WA, Khan MI. MicroRNA-125b-5p regulates IL-1β induced inflammatory genes via targeting TRAF6-mediated MAPKs and NF-κB signaling in human osteoarthritic chondrocytes. Sci Rep 2019; 9:6882. [PMID: 31053727 PMCID: PMC6499837 DOI: 10.1038/s41598-019-42601-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 04/04/2019] [Indexed: 12/23/2022] Open
Abstract
Abnormal post-transcriptional modulations in inflammatory genes by microRNAs (miRNAs) play a crucial role in human disorders including arthritis. In this study, we determined the effect of hsa-miR-125b-5p on interleukin (IL)-1β induced inflammatory genes in human osteoarthritic (OA) chondrocytes. Bioinformatics algorithms showed 3′untranslated region (3′UTR) of TRAF6 mRNA (NM_004620.3) has perfectly matched ‘seed-sequence’ for hsa-miR-125b-5p. Treatment of cells with IL-1β up-regulates TRAF6 mRNA and down-regulates hsa-miR-125b-5p expression. This negative correlation between TRAF6 and hsa-miR-125b-5p was verified by transfection with miR-125b mimic (pre-miR-125b). Moreover, transfection with miR-125b mimic caused marked inhibition of IL-1β-induced phosphorylation of p38-MAPK, JNK-MAPKs and ERK-MAPKs and also suppressed the nuclear levels of NF-κBp50, NF-κBp65 and inhibited the activation of IκBα. Furthermore, transfected chondrocytes with miR-125b mimic in the presence of IL-1β also showed marked inhibition in the secretion of several proinflammatory cytokines, chemokines and growth factors including IL-6, IL-8, INF-γ, TGF-β1, IGFBP-1 and PGDF-BB. Importantly, this transfection also significantly inhibited IL-1β- induced MMP-13 expression/production. In short, this study concludes that hsa-miR-125b-5p acts as a negative co-regulator of inflammatory genes including MMP-13 via targeting TRAF6/MAPKs/NF-κB pathway in human OA chondrocytes.
Collapse
Affiliation(s)
- Zafar Rasheed
- Department of Medical Biochemistry, College of Medicine, Buraidah, Qassim University, Buraidah, Saudi Arabia.
| | - Naila Rasheed
- Department of Medical Biochemistry, College of Medicine, Buraidah, Qassim University, Buraidah, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Buraidah, Qassim University, Buraidah, Saudi Arabia
| | - Muhammad Ismail Khan
- Faculty of Medicine, School of Public Health, University of Queensland, Brisbane, Australia
| |
Collapse
|
538
|
Enteshari-Moghaddam A, Isazadehfar K, Habibzadeh A, Hemmati M. Efficacy of Methotrexate on Pain Severity Reduction and Improvement of Quality of Life in Patients with Moderate to Severe Knee Osteoarthritis. Anesth Pain Med 2019; 9:e89990. [PMID: 31497519 PMCID: PMC6712359 DOI: 10.5812/aapm.89990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 12/22/2022] Open
Abstract
Background Knee osteoarthritis (OA) leads to low quality of life due to pain and limitation in daily activities. Recent studies indicated that Methotrexate (MTX) could reduce pain due to its anti-inflammatory effects. Objectives In this study, the researchers aimed at evaluating the efficacy of MTX in pain control and improvement of quality of life in patients with moderate to severe knee OA. Methods In this randomized clinical trial, 100 patients with moderate to severe knee OA were allocated to receive MTX (n = 50) 7.5 mg weekly to be increased to 15 mg weekly after first months or placebo (n = 50) for six months. Pain severity was measured using the numerical rating scale (NRS), so was functional status by Western Ontario and McMaster Universities Arthritis Index (WOMAC) and quality of life by SF-12 questionnaire before the treatment, and three months and six months after the intervention. The results were compared between the groups subsequently. Nine patients from the MTX group were excluded due to the use of corticosteroids during the treatment period. Results The MTX group compared to the placebo group had significant improvement in pain severity and quality of life during six months and WOMAC parameters at three and six months after the intervention. The need for NSAIDS was slightly higher in the placebo group with no significant difference (22% versus 36%, P = 0.14). The MTX adverse effects were not observed. Conclusions Treatment of moderate to severe knee OA with MTX could reduce pain severity and improve functional status and quality of life in OA patients.
Collapse
Affiliation(s)
| | - Khatereh Isazadehfar
- Social Determinants of Health Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Afshin Habibzadeh
- Department of Internal Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
- Corresponding Author: Department of Internal Medicine, Imam Khomeini Hospital, Ardabil University of Medical Sciences, Postal Code: 5618985991, Ardabil, Iran. Tel/Fax: +98-4533522391,
| | - Mehdi Hemmati
- Internal Medicine Resident at MedStar Health, MedStar Georgetown University Hospital, Washington, United States of America
| |
Collapse
|
539
|
Wojcik-Pastuszka D, Krzak J, Macikowski B, Berkowski R, Osiński B, Musiał W. Evaluation of the Release Kinetics of a Pharmacologically Active Substance from Model Intra-Articular Implants Replacing the Cruciate Ligaments of the Knee. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E1202. [PMID: 31013801 PMCID: PMC6515312 DOI: 10.3390/ma12081202] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/23/2022]
Abstract
Implants are readily applied as a convenient method of therapy. There is great interest in the prolonged release of active substances from implants. The objective of this work was to evaluate the dissolution kinetics of steroidal anti-inflammatory preparation (SAP) released from novel implants, and to test the influence of the technology on SAP release kinetics. The proposed long-acting preparations may overcome difficulties resulting from repeated injections and often visits to ambulatory clinic, as the stabilizing function of the artificial ligament would be enriched with pharmacological activity. The potential advantages provided by the new coatings of knee implants include the continuous, sustained, and prolonged release of an active substance. The study was carried out using a modified United States Pharmacopoeia (USP) apparatus 4. The amount of SAP was measured spectroscopically. It was revealed that the transport of the drug was mainly a diffusion process. The drug release kinetics was analyzed using zero-, first-, and second-order kinetics as well as Korsmeyer-Peppas, Higuchi, and Hixon-Crowell models. The highest values of the release rate constants were k0 = (7.49 ± 0.05) × 10-5 mg × min-1, k1 = (6.93 ± 0.05) × 10-6 min-1, and k2 = (7.70 ± 0.05) × 10-7 mg-1 × min-1 as calculated according to zero-, first-, and second-order kinetics equations, respectively. The values of the rate constants obtained for the slowest process were k0 = (3.63 ± 0.06) × 10-5 mg × min-1, k1 = (2.50 ± 0.03) × 10-6 min-1, and k2 = (2.80 ± 0.03) × 10-7 mg-1 × min-1. They may suggest the possibility of sustained release of betamethasone from the system. Due to the statistical analysis, differences were observed between most of the studied implants. Incubation, temperature, time of stabilization of layers, and the method of SAP deposition on the matrix affected the drug release.
Collapse
Affiliation(s)
- Dorota Wojcik-Pastuszka
- Department of Physical Chemistry, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211A, 55-556 Wroclaw, Poland.
| | - Justyna Krzak
- Department of Mechanics, Materials Science and Engineering, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, ul. Smoluchowskiego 25, 50-370 Wrocław, Poland.
| | - Bartosz Macikowski
- Department of Physical Chemistry, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211A, 55-556 Wroclaw, Poland.
| | - Ryszard Berkowski
- Department of Physical Chemistry, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211A, 55-556 Wroclaw, Poland.
| | - Bogdan Osiński
- Department and Clinic of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 51, 50-366 Wroclaw, Poland.
| | - Witold Musiał
- Department of Physical Chemistry, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211A, 55-556 Wroclaw, Poland.
| |
Collapse
|
540
|
Stefani RM, Halder SS, Estell EG, Lee AJ, Silverstein AM, Sobczak E, Chahine NO, Ateshian GA, Shah RP, Hung CT. A Functional Tissue-Engineered Synovium Model to Study Osteoarthritis Progression and Treatment. Tissue Eng Part A 2019; 25:538-553. [PMID: 30203722 PMCID: PMC6482911 DOI: 10.1089/ten.tea.2018.0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/31/2018] [Indexed: 01/15/2023] Open
Abstract
IMPACT STATEMENT The synovium envelops the diarthrodial joint and plays a key regulatory role in defining the composition of the synovial fluid through filtration and biosynthesis of critical boundary lubricants. Synovium changes often precede cartilage damage in osteoarthritis. We describe a novel in vitro tissue engineered model, validated against native synovium explants, to investigate the structure-function of synovium through quantitative solute transport measures. Synovium was evaluated in the presence of a proinflammatory cytokine, interleukin-1, or the clinically relevant corticosteroid, dexamethasone. We anticipate that a better understanding of synovium transport would support efforts to develop more effective strategies aimed at restoring joint health.
Collapse
Affiliation(s)
- Robert M. Stefani
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Saiti S. Halder
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Eben G. Estell
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Andy J. Lee
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Amy M. Silverstein
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Evie Sobczak
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Nadeen O. Chahine
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Mechanical Engineering, Columbia University, New York, New York
| | - Roshan P. Shah
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
541
|
Restoring knee joint kinematics after anterior cruciate ligament injury might inhibit synovial membrane inflammation. SPORT SCIENCES FOR HEALTH 2019. [DOI: 10.1007/s11332-018-0481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
542
|
Contribution of Infrapatellar Fat Pad and Synovial Membrane to Knee Osteoarthritis Pain. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6390182. [PMID: 31049352 PMCID: PMC6462341 DOI: 10.1155/2019/6390182] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is the most common form of joint disease and a major cause of pain and disability in the adult population. Interestingly, there are patients with symptomatic OA displaying pain, while patients with asymptomatic OA that do not experience pain but show radiographic signs of joint damage. Pain is a complex experience integrating sensory, affective, and cognitive processes related to several peripheral and central nociceptive factors besides inflammation. During the last years, the role of infrapatellar fat pad (IFP), other than the synovial membrane, has been investigated as a potential source of pain in OA. Interestingly, new findings suggest that IFP and synovial membrane might act as a functional unit in OA pathogenesis and pain. The present review discuss the role of IFP and synovial membrane in the development of OA, with a particular focus on pain onset and the possible involved mediators that may play a role in OA pathology and pain mechanisms. Inflammation of IFP and synovial membrane may drive peripheral and central sensitization in KOA. Since sensitization is associated with pain severity in knee OA and may potentially contribute to the transition from acute to chronic, persistent pain in knee OA, preventing sensitization would be a potentially effective and novel means of preventing worsening of pain in knee OA.
Collapse
|
543
|
Regulation of fibroblast-like synoviocyte transformation by transcription factors in arthritic diseases. Biochem Pharmacol 2019; 165:145-151. [PMID: 30878552 DOI: 10.1016/j.bcp.2019.03.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
Inflammation in the synovium is known to mediate joint destruction in several forms of arthritis. Fibroblast-like synoviocytes (FLS) are cells that reside in the synovial lining of joints and are known to be key contributors to inflammation associated with arthritis. FLS are a major source of inflammatory cytokines and catabolic enzymes that promote joint degeneration. We now know that there exists a direct correlation between the signaling pathways that are activated by the pro-inflammatory molecules produced by the FLS, and the severity of joint degeneration in arthritis. Research focused on understanding the signaling pathways that are activated by these pro-inflammatory molecules has led to major advancements in the understanding of the joint pathology in arthritis. Transcription factors (TFs) that act as downstream mediators of the pro-inflammatory signaling cascades in various cell types have been reported to play an important role in inducing the deleterious transformation of the FLS. Interestingly, recent studies have started uncovering that several TFs that were previously reported to play role in embryonic development and cancer, but not known to have pronounced roles in tissue inflammation, can actually play crucial roles in the regulation of the pathological properties of the FLS. In this review, we will discuss reports that have been able to impart novel arthritogenic roles to TFs that are specialized in embryonic development. We also discuss the therapeutic potential of targeting these newly identified regulators of FLS transformation in the treatment of arthritis.
Collapse
|
544
|
NLRX1 alleviates lipopolysaccharide-induced apoptosis and inflammation in chondrocytes by suppressing the activation of NF-κB signaling. Int Immunopharmacol 2019; 71:7-13. [PMID: 30861394 DOI: 10.1016/j.intimp.2019.03.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/20/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a chronic debilitating disease characterized by joint degeneration. Excessive chondrocyte apoptosis and inflammation contributes to articular cartilage destruction in OA pathology. Nucleotide-binding oligomerization domain (NOD)-like receptor X1 (NLRX1) has emerged as a critical regulator of inflammation that participates in the pathology of diverse diseases. To date, little is known about the role of NLRX1 in OA. In the present study, we aimed to explore the function of NLRX1 in lipopolysaccharide (LPS)-induced injury in chondrocytes, an in vitro model of OA. NLRX1 mRNA was detected by quantitative polymerase chain reaction (qPCR) analysis. Protein expression of NLRX1, phosphorylated IκB kinase β (IKKβ), and phosphorylated nuclear factor-κB (NF-κB) p65 were examined by western blot. Cell viability was assessed by the MTT assay. Cell apoptosis was evaluated by measuring caspase-3 activity. Cytokine release was assessed by enzyme-linked immunosorbent assay (ELISA). NF-κB signaling activation was analyzed with a luciferase reporter assay. Herein, our results revealed that NLRX1 expression was markedly decreased in LPS-treated chondrocytes. Functional experiments demonstrated that NLRX1 overexpression significantly improved cell viability and attenuated LPS-treated chondrocyte apoptosis and inflammation, while NLRX1 silencing caused the opposite effects. Moreover, our results showed that NLRX1 regulated LPS-induced NF-κB signaling activation. Notably, NF-κB signaling inhibition significantly reversed the NLRX1-knockdown-mediated enhanced effects on LPS-induced apoptosis and inflammation. Overall, these results demonstrate that NLRX1 alleviates LPS-induced apoptosis and inflammation in chondrocytes by negatively regulating NF-κB signaling, results that indicate an anti-inflammatory role for NLRX1 in OA. Our findings suggest that NLRX1 may serve as a potential therapeutic target for OA.
Collapse
|
545
|
Caron JP, Gandy JC, Brown JL, Sordillo LM. Omega-3 fatty acids and docosahexaenoic acid oxymetabolites modulate the inflammatory response of equine recombinant interleukin1β-stimulated equine synoviocytes. Prostaglandins Other Lipid Mediat 2019; 142:1-8. [PMID: 30836143 DOI: 10.1016/j.prostaglandins.2019.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/21/2018] [Accepted: 02/28/2019] [Indexed: 01/15/2023]
Abstract
Omega-3 fatty acid (n-3 PUFA) supplementation may have beneficial effects in certain chronic diseases, potentially including osteoarthritis. Favorable effects are attributed, in part, to downstream pro-resolving oxylipid metabolites. We investigated the role of n-3 PUFA and docosahexaenoic acid (DHA)-derived oxylipids (docosanoids) on equine synoviocyte metabolism. We hypothesized that n-3 PUFA and selected docosanoids would modulate inflammatory mediator gene expression by recombinant equine (re)IL-1β-stimulated synovial fibroblasts. Synoviocyte monolayer cultures were prepared from grossly normal equine carpal synovium. Cellular incorporation of eicosapentaenoic acid (EPA) and DHA was determined using LC-MS and docosanoid biosynthesis by LC-MS-MS. The influence of n-3 PUFA and docosanoids on osteoarthritis marker gene expression was determined by quantitative real time polymerase chain reaction (qPCR). Synoviocytes incorporated EPA and DHA in significant amounts and DHA treatment augmented the synthesis of several docosanoids. Synoviocyte cultures pre-treated with EPA or DHA followed by reIL-1β stimulation had significant reductions in expression of ADAMTS4, MMP-1, MMP-13, IL-1β, IL-6 and COX-2. The docosanoids resolvin D1 and D2, maresin 1 and protectin DX, alone and in combination, abrogated ADAMTS4, MMP-1, MMP-13, and IL-6 gene expression in reIL-1β-stimulated synoviocytes. Similarly, both resolvins and maresin 1 stifled COX-2 expression. Our results demonstrate that synoviocytes readily incorporate n-3 PUFA. DHA incorporation was sufficient for biosynthesis of significant concentrations of several docosanoids which modulated the synovial inflammatory response in vitro. These data indicate n-3 PUFA supplementation may prove useful in the prevention or treatment of osteoarthritis.
Collapse
Affiliation(s)
- John P Caron
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48823-1314, United States.
| | - Jeffrey C Gandy
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48823-1314, United States
| | - Jennifer L Brown
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48823-1314, United States
| | - Lorraine M Sordillo
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48823-1314, United States
| |
Collapse
|
546
|
Belluzzi E, Olivotto E, Toso G, Cigolotti A, Pozzuoli A, Biz C, Trisolino G, Ruggieri P, Grigolo B, Ramonda R, Favero M. Conditioned media from human osteoarthritic synovium induces inflammation in a synoviocyte cell line. Connect Tissue Res 2019; 60:136-145. [PMID: 29695173 DOI: 10.1080/03008207.2018.1470167] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM Osteoarthritis (OA) is a whole joint pathology involving cartilage, synovial membrane, meniscus, subchondral bone, and infrapatellar fat pad (IFP). Synovitis has been widely documented in OA suggesting its important role in pathogenesis. The aim of this study was to investigate the role of different joint tissues in promoting synovitis. MATERIALS AND METHODS Conditioned media (CM) from cartilage, synovial membrane, meniscus, and IFP were generated from tissues of five patients undergoing total knee replacement and used to stimulate a human fibroblast-like synoviocytes cell line (K4IM). Cytokines, chemokines, and metalloproteases release was analyzed in all CM by Bio-Plex Assay and sulfated glycosaminoglycan (GAG) content by dimethylmethylene blue assay. Gene expression of several markers was evaluated by real-time PCR in K4IM cells stimulated with the CM obtained from joint tissues. RESULTS CM from all tissues produced high levels of IL-6, IL-8, and CCL2. CCL21, MMP-3, and -13 levels were detected in all CM except IFP. MMP-10 was present only in CM of cartilage and synovial tissues. IL-1β, IL-15, TNF-α, CCL5, and CCL19 were undetectable. However, only K4IM cells stimulated by the CM from OA synovium showed an increase of IL-6, CXCL-8, CCL21, MMP10, and IL-1β expression. CONCLUSION Our study showed that K4IM might be a suitable in vitro model for evaluating different cellular pathways in OA studies. Importantly, we demonstrated that in OA, all joint tissues might be involved in the progression of synovitis with a predominant role of synovial membrane itself compared to the other joint tissues.
Collapse
Affiliation(s)
- Elisa Belluzzi
- a Rheumatology Unit, Department of Medicine-DIMED , University Hospital of Padova , Padova , Italy.,b Musculoskeletal Pathology and Oncology Laboratory, Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Eleonora Olivotto
- c RAMSES Laboratory-RIT Department , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Giovanna Toso
- b Musculoskeletal Pathology and Oncology Laboratory, Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Augusto Cigolotti
- d Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Assunta Pozzuoli
- b Musculoskeletal Pathology and Oncology Laboratory, Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Carlo Biz
- d Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Giovanni Trisolino
- e Reconstructive Hip and Knee Joint Surgery , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy.,f Pediatric orthopedic and Traumatology , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Pietro Ruggieri
- d Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Brunella Grigolo
- c RAMSES Laboratory-RIT Department , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy.,g Laboratory of Immunorheumatology and Tissue Regeneration , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Roberta Ramonda
- a Rheumatology Unit, Department of Medicine-DIMED , University Hospital of Padova , Padova , Italy
| | - Marta Favero
- a Rheumatology Unit, Department of Medicine-DIMED , University Hospital of Padova , Padova , Italy.,c RAMSES Laboratory-RIT Department , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| |
Collapse
|
547
|
Pontes-Quero GM, García-Fernández L, Aguilar MR, San Román J, Pérez Cano J, Vázquez-Lasa B. Active viscosupplements for osteoarthritis treatment. Semin Arthritis Rheum 2019; 49:171-183. [PMID: 30878154 DOI: 10.1016/j.semarthrit.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/05/2019] [Accepted: 02/12/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Osteoarthritis is a chronic, painful and disabling disease which prevalence is increasing in developing countries. Patients with osteoarthritis present a reduced synovial fluid viscoelasticity due to a reduction in concentration and molecular weight of hyaluronic acid. Currently, the main treatment used to restore the compromised rheological properties of synovial fluid is the viscosupplementation by hyaluronic acid injections that can be combined with oral anti-inflammatory drugs for pain relief. Combination of viscosupplements with chemical agents or drugs is emerging as a new strategy to provide a double action of synovial fluid viscoelasticity recovery and the therapeutic effect of the bioactive principle. METHODS In this review, we present the latest research on the combination of viscosupplements with active molecules. We conducted a literature review of articles published in different web search engines and categorized according to the active molecule introduced into the viscosupplement. RESULTS Generally, the introduction of anti-inflammatory molecules have shown to improve pain relief although some cytotoxicity has been demonstrated especially for non-steroidal anti-inflammatory drugs. Other molecules such as antioxidant or disease modifying osteoarthritis drugs have been reported to improve viscosupplementation action. Drug delivery systems combined with hyaluronic acid could enhance the activity of the encapsulated molecules and provide better control over the drug release. Finally, biological approaches such as the use of stem cells or platelet-rich plasma seem to be the most promising strategies for cartilage recovery. CONCLUSIONS Combination therapy of viscosupplements with therapeutic agents, drug delivery systems or regenerative therapies can improve viscosupplementation outcome in terms of pain relief and joint functionality. However, further research is needed in order to reach more conclusive results.
Collapse
Affiliation(s)
- Gloria María Pontes-Quero
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; Alodia Farmacéutica SL, Madrid, Spain
| | - Luis García-Fernández
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - María Rosa Aguilar
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| | - Julio San Román
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | | | - Blanca Vázquez-Lasa
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
548
|
Huang H, Zhang ZF, Qin FW, Tang W, Liu DH, Wu PY, Jiao F. The mechanism of TDP-43 gene expression on inflammatory factors and the JNK and p38 MAPK signalling pathways in ischaemic hypoxic stress dependence. Int Wound J 2019; 16:724-729. [PMID: 30784197 DOI: 10.1111/iwj.13087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
In this study, the mechanism of TDP-43 gene expression on inflammatory factors and Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) signalling pathways in ischaemic hypoxic stress dependence was investigated. Sixty SD rats were selected and divided into the control group, the osteoarthritis (OA) model group, and the TDP-43-mMSCs+OA group. In the OA model group and the TDP-43-mMSCs+OA group, OA was established by collagenase injection. Western blotting assays were used to detect the expression of TDP-43 in cartilage tissues of each rat. The secretion of tumour necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the serum of rats was determined by enzyme-linked immunosorbent assay (ELISA). The formation of cytoplasmic stress granules (SGs) and the expression of receptor for activated c-kinase 1 (RACK1) were detected by Western blotting assays in each group of rats. The expression of MTK1 and MAPKKK phosphorylation and changes in the JNK and p38 MAPK signalling pathways were detected by Western blotting assays. Compared with the control group, the expression of TDP-43 in the cartilage tissue of rats in the OA model group was significantly decreased. The expression of TDP-43 in the cartilage tissue of rats in the TDP-43-mMSCs+OA group was significantly higher than that of the control group and the OA model group, which indicates that TDP-43-mMSC transplantation was successful. Enzyme-linked immunosorbent assay results showed that the plasma TNF-α and IL-1β levels in the OA model group were significantly increased (P < 0.01) when compared with the control group. However, the secretion of TNF-α and IL-1β in the serum of the TDP-43-mMSCs+OA group was significantly lower than that of the model group (P < 0.01) but still higher than the control group. This indicates that overexpression of TDP-43 reduces the inflammatory response induced by OA. Western blotting assays showed that the amount of cytoplasmic SGs in the cartilage tissue of rats in the OA model group was significantly decreased when compared with the control group. The amount of SGs in the cartilage of rats in the TDP-43-mMSCs+OA group was significantly higher than that of the model group. The expression of RACK1 in the cartilage tissue of rats in the OA model group was significantly higher than that of the control group. Overexpression of the TDP-43 gene can interfere with the secretion of inflammatory factors and inhibit the activation of the JNK and p38 MAPK signalling pathways by ischaemic hypoxia stress. Thus, the molecular mechanism of chondrocytopathic lesions was reversed, which provided a new theoretical basis for the treatment of OA.
Collapse
Affiliation(s)
- He Huang
- Department of Orthopedic Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Zhao-Fei Zhang
- Department of Orthopedic Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Feng-Wei Qin
- Department of Orthopedic Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Wang Tang
- Department of Orthopedic Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Dong-Hua Liu
- Department of Orthopedic Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Pei-Yu Wu
- Department of Orthopedic Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Feng Jiao
- Department of Orthopedic Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| |
Collapse
|
549
|
Harth M, Nielson WR. Pain and affective distress in arthritis: relationship to immunity and inflammation. Expert Rev Clin Immunol 2019; 15:541-552. [PMID: 30669892 DOI: 10.1080/1744666x.2019.1573675] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Most arthritides are associated with pain and psychological distress (clinically significant depression and anxiety). Pain and depression are mutually exacerbating; both may continue even when joint involvement appears well controlled. Area covered: There is strong evidence that arthritis-related stress impacts the central nervous system and, together with peripheral inflammatory changes, can cause central sensitization that can lead to chronic pain and worsening of affective distress. Cytokines and chemokines participate both in joint inflammation and in central sensitization. We review evidence of these relationships in five arthritides, namely rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, gout, and in osteoarthritis of the hips and knees. Central sensitization in these conditions results in long-lasting pain and psychological distress. Expert commentary: Chronic pain and depression are important but often neglected in the clinical assessment and treatment of arthritis. The potential role of biologic cytokines and Janus kinase inhibitors in dealing with these symptoms needs further study.
Collapse
Affiliation(s)
- Manfred Harth
- a Medicine , Western University , London , Ontario , Canada
| | - Warren R Nielson
- b Lawson Health Research Institute , London , Ontario , Canada.,c Department of Psychology , Western University , London , Ontario , Canada
| |
Collapse
|
550
|
Castrogiovanni P, Di Rosa M, Ravalli S, Castorina A, Guglielmino C, Imbesi R, Vecchio M, Drago F, Szychlinska MA, Musumeci G. Moderate Physical Activity as a Prevention Method for Knee Osteoarthritis and the Role of Synoviocytes as Biological Key. Int J Mol Sci 2019; 20:ijms20030511. [PMID: 30691048 PMCID: PMC6387266 DOI: 10.3390/ijms20030511] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to investigate the influence of moderate physical activity (MPA) on the expression of osteoarthritis (OA)-related (IL-1β, IL-6, TNF-α, MMP-13) and anti-inflammatory and chondroprotective (IL-4, IL-10, lubricin) biomarkers in the synovium of an OA-induced rat model. A total of 32 rats were divided into four groups: Control rats (Group 1); rats performing MPA (Group 2); anterior cruciate ligament transection (ACLT)-rats with OA (Group 3); and, ACLT-rats performing MPA (Group 4). Analyses were performed using Hematoxylin & Eosin (H&E) staining, histomorphometry and immunohistochemistry. In Group 3, OA biomarkers were significantly increased, whereas, IL-4, IL-10, and lubricin were significantly lower than in the other experimental groups. We hypothesize that MPA might partake in rescuing type B synoviocyte dysfunction at the early stages of OA, delaying the progression of the disease.
Collapse
Affiliation(s)
- Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia n°87, 95124 Catania, Italy.
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia n°87, 95124 Catania, Italy.
| | - Silvia Ravalli
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia n°87, 95124 Catania, Italy.
| | - Alessandro Castorina
- School of Life Sciences, Faculty of Science, University of Technology Sydney, P.O. Box 123, Broadway, Sydney, NSW 2007, Australia.
- Discipline of Anatomy and Histology, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Claudia Guglielmino
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia n°87, 95124 Catania, Italy.
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia n°87, 95124 Catania, Italy.
| | - Michele Vecchio
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, via S. Sofia 67, 95123 Catania, Italy.
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, via S. Sofia 67, 95123 Catania, Italy.
| | - Marta Anna Szychlinska
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia n°87, 95124 Catania, Italy.
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia n°87, 95124 Catania, Italy.
- School of the Sport of the Italian National Olympic Committee "CONI" Sicily, Via Emanuele Notarbartolo, 90141 Palermo, Italy.
| |
Collapse
|