551
|
Fathollahi A, Aslani S, Jamshidi A, Mahmoudi M. Epigenetics in osteoarthritis: Novel spotlight. J Cell Physiol 2019; 234:12309-12324. [DOI: 10.1002/jcp.28020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Anwar Fathollahi
- Department of Immunology School of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
552
|
Savvidou O, Milonaki M, Goumenos S, Flevas D, Papagelopoulos P, Moutsatsou P. Glucocorticoid signaling and osteoarthritis. Mol Cell Endocrinol 2019; 480:153-166. [PMID: 30445185 DOI: 10.1016/j.mce.2018.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/03/2018] [Accepted: 11/11/2018] [Indexed: 01/15/2023]
Abstract
Glucocorticoids are steroid hormones synthesized and released by the adrenal cortex. Their main function is to maintain cell homeostasis through a variety of signaling pathways, responding to changes in an organism's environment or developmental status. Mimicking the actions of natural glucocorticoids, synthetic glucocorticoids have been recruited to treat many diseases that implicate glucocorticoid receptor signaling such as osteoarthritis. In osteoarthritis, synthetic glucocorticoids aim to alleviate inflammation and pain. The variation of patients' response and the possibility of complications associated with their long-term use have led to a need for a better understanding of glucocorticoid receptor signaling in osteoarthritis. In this review, we performed a literature search in the molecular pathways that link the osteoarthritic joint to the glucocorticoid receptor signaling. We hope that this information will advance research in the field and propose new molecular targets for the development of more optimized therapies for osteoarthritis.
Collapse
Affiliation(s)
- Olga Savvidou
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Mandy Milonaki
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Stavros Goumenos
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Dimitrios Flevas
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Panayiotis Papagelopoulos
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Paraskevi Moutsatsou
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| |
Collapse
|
553
|
Burke CJ, Alizai H, Beltran LS, Regatte RR. MRI of synovitis and joint fluid. J Magn Reson Imaging 2019; 49:1512-1527. [PMID: 30618151 DOI: 10.1002/jmri.26618] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 12/20/2022] Open
Abstract
Synovitis and joint effusion are common manifestations of rheumatic disease and play an important role in the disease pathophysiology. Earlier detection and accurate assessment of synovial pathology, therefore, can facilitate appropriate clinical management and hence improve prognosis. Magnetic resonance imaging (MRI) allows unparalleled assessment of all joint structures and associated pathology. It has emerged as a powerful tool, which enables not only detection of synovitis and effusion, but also allows quantification, detailed characterization, and noninvasive monitoring of synovial processes. The purpose of this article is to summarize the pathophysiology of synovitis and to review the role of qualitative, semiquantitative, and quantitative MRI in the assessment of synovitis and joint fluid. We also discuss the utility of MRI as an outcome measure to assess treatment response, particularly with respect to osteoarthritis and rheumatoid arthritis. Emerging applications such as hybrid positron emission tomography / MRI and molecular imaging are also briefly discussed. Level of Evidence: 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2019.
Collapse
Affiliation(s)
| | - Hamza Alizai
- Department of Radiology, NYU Langone Health, New York, New York, USA
| | - Luis S Beltran
- Department of Radiology, NYU Langone Health, New York, New York, USA
| | | |
Collapse
|
554
|
Choi WS, Yang JI, Kim W, Kim HE, Kim SK, Won Y, Son YO, Chun CH, Chun JS. Critical role for arginase II in osteoarthritis pathogenesis. Ann Rheum Dis 2019; 78:421-428. [PMID: 30610061 PMCID: PMC6390026 DOI: 10.1136/annrheumdis-2018-214282] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
Objective Osteoarthritis (OA) appears to be associated with various metabolic disorders, but the potential contribution of amino acid metabolism to OA pathogenesis has not been clearly elucidated. Here, we explored whether alterations in the amino acid metabolism of chondrocytes could regulate OA pathogenesis. Methods Expression profiles of amino acid metabolism-regulating genes in primary-culture passage 0 mouse chondrocytes were examined by microarray analysis, and selected genes were further characterised in mouse OA chondrocytes and OA cartilage of human and mouse models. Experimental OA in mice was induced by destabilisation of the medial meniscus (DMM) or intra-articular (IA) injection of adenoviruses expressing catabolic regulators. The functional consequences of arginase II (Arg-II) were examined in Arg2−/− mice and those subjected to IA injection of an adenovirus encoding Arg-II (Ad-Arg-II). Results The gene encoding Arg-II, an arginine-metabolising enzyme, was specifically upregulated in chondrocytes under various pathological conditions and in OA cartilage from human patients with OA and various mouse models. Adenovirus-mediated overexpression of Arg-II in mouse joint tissues caused OA pathogenesis, whereas genetic ablation of Arg2 in mice (Arg2−/−) abolished all manifestations of DMM-induced OA. Mechanistically, Arg-II appears to cause OA cartilage destruction at least partly by upregulating the expression of matrix-degrading enzymes (matrix metalloproteinase 3 [MMP3] and MMP13) in chondrocytes via the nuclear factor (NF)-κB pathway. Conclusions Our results indicate that Arg-II is a crucial regulator of OA pathogenesis in mice. Although chondrocytes of human and mouse do not identically, but similarly, respond to Arg-II, our results suggest that Arg-II could be a therapeutic target of OA pathogenesis.
Collapse
Affiliation(s)
- Wan-Su Choi
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jeong-In Yang
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Wihak Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Hyo-Eun Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Seul-Ki Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yoonkyung Won
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Young-Ok Son
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Churl-Hong Chun
- Department of Orthopedic Surgery, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Jang-Soo Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
555
|
Chen YJ, Chang WA, Wu LY, Huang CF, Chen CH, Kuo PL. Identification of Novel Genes in Osteoarthritic Fibroblast-Like Synoviocytes Using Next-Generation Sequencing and Bioinformatics Approaches. Int J Med Sci 2019; 16:1057-1071. [PMID: 31523167 PMCID: PMC6743272 DOI: 10.7150/ijms.35611] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/05/2019] [Indexed: 01/15/2023] Open
Abstract
Synovitis in osteoarthritis (OA) the consequence of low grade inflammatory process caused by cartilage breakdown products that stimulated the production of pro-inflammatory mediators by fibroblast-like synoviocytes (FLS). FLS participate in joint homeostasis and low grade inflammation in the joint microenvironment triggers FLS transformation. In the current study, we aimed to identify differentially expressed genes and potential miRNA regulations in human OA FLS through deep sequencing and bioinformatics approaches. The 245 differentially expressed genes in OA FLS were identified, and pathway analysis using various bioinformatics databases indicated their enrichment in functions related to altered extracellular matrix organization, cell adhesion and cellular movement. Moreover, among the 14 dysregulated genes with potential miRNA regulations identified, src kinase associated phosphoprotein 2 (SKAP2), adaptor related protein complex 1 sigma 2 subunit (AP1S2), PHD finger protein 21A (PHF21A), lipoma preferred partner (LPP), and transcription factor AP-2 alpha (TFAP2A) showed similar expression patterns in OA FLS and OA synovial tissue datasets in Gene Expression Omnibus database. Ingenuity Pathway Analysis identified the dysregulated LPP participated in cell migration and cell spreading of OA FLS, which was potentially regulated by miR-141-3p. The current findings suggested new perspectives into understanding the novel molecular signatures of FLS involved in the pathogenesis of OA, which may be potential therapeutic targets.
Collapse
Affiliation(s)
- Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ling-Yu Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ching-Fen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Chia-Hsin Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.,Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Center for Cancer Research, Kaohsiung Medical University
| |
Collapse
|
556
|
Yun Z, Peng HZ, Wang W, Ma Q, Chen J, Wang XL, Ma BA. A20 inhibits the release of inflammatory cytokines by suppressing the activation of the nuclear factor-kappa B pathway in osteoarthritic fibroblast-like synoviocytes. Biochem Biophys Res Commun 2019; 508:877-881. [DOI: 10.1016/j.bbrc.2018.12.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 12/05/2018] [Indexed: 01/31/2023]
|
557
|
Wang B, Lang Y, Zhang L. Histopathological changes in the infrapatellar fat pad in an experimental rabbit model of early patellofemoral osteoarthritis. Knee 2019; 26:2-13. [PMID: 30415972 DOI: 10.1016/j.knee.2018.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/20/2018] [Accepted: 06/08/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND The purpose of this study was to characterise the histopathological changes in the infrapatellar fat pad (IPFP) in the early stage of patellofemoral osteoarthritis (PFOA). METHODS Sixty-four New Zealand white rabbits were randomly divided into experimental (n = 24), sham (n = 16), and control groups (n = 24). In the experimental group, denoted as the patellar ligament uneven shortening group (US group), the patellar ligament (PL) was folded eight millimetres and sutured. After eight weeks, all animals were euthanised, and magnetic resonance imaging (MRI) evaluation, wet IPFP weight measurement, and histopathological and immunohistochemistry analysis were performed to analyse the histopathological changes in the IPFPs. RESULTS The maximum cross-sectional area (CSA) of the IPFPs in the sagittal position of MRI in the control group, sham group, and US group were 45.50 ± 7.19 mm2, 45.88 ± 6.60 mm2 (vs. control group, P = 0.907), and 53.83 ± 8.24 mm2 (vs. control group, P = 0.015; vs. sham group, P = 0.035), respectively. The MRI intensity of the IPFPs in the control group, sham group, and US group were 115.53 ± 28.85, 108.53 ± 26.73 (vs. control group, P = 0.589), and 154.52 ± 18.48 (vs. control group, P = 0.002; vs. sham group, P = 0.002), respectively. The wet weight of the IPFPs in the control group, sham group, and US group were 0.32 ± 0.05 g, 0.32 ± 0.04 g (vs. control group, P = 0.895), and 0.38 ± 0.06 g (vs. control group, P = 0.017; vs. sham group, P = 0.033), respectively. The Osteoarthritis Research Society International (OARSI) scores of the IPFPs in the US group were 6.00 ± 1.91, which was higher than the scores of 2.50 ± 2.02 (P < 0.001) in the control group and of 2.75 ± 1.67 (P = 0.001) in the sham group. CONCLUSIONS The histopathological changes of the IPFPs as determined via MRI and microscopic structure appeared to occur much earlier than cartilage damage in PFOA. Furthermore, detecting and treating the IPFP changes may offer aid in the diagnosis and treatment of PFOA.
Collapse
Affiliation(s)
- Binggang Wang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China
| | - Yanfei Lang
- Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, China
| | - Liu Zhang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China; Department of Orthopedic Surgery, The Affiliated Hospital of North China University of Science and Technology, Tangshan, China.
| |
Collapse
|
558
|
Bicho D, Ajami S, Liu C, Reis RL, Oliveira JM. Peptide-biofunctionalization of biomaterials for osteochondral tissue regeneration in early stage osteoarthritis: challenges and opportunities. J Mater Chem B 2019; 7:1027-1044. [DOI: 10.1039/c8tb03173h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Osteoarthritis is a degenerative joint disease characterized by the progressive deterioration of articular cartilage, synovial inflammation and changes in periarticular and subchondral bone, being a leading cause of disability.
Collapse
Affiliation(s)
- D. Bicho
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| | - S. Ajami
- Institute of Orthopaedics and Musculo-Skeletal Sci, University College London, Royal National Orthopaedic Hospital
- Stanmore
- UK
| | - C. Liu
- Institute of Orthopaedics and Musculo-Skeletal Sci, University College London, Royal National Orthopaedic Hospital
- Stanmore
- UK
| | - R. L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| | - J. M. Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| |
Collapse
|
559
|
Oo WM, Yu SPC, Daniel MS, Hunter DJ. Disease-modifying drugs in osteoarthritis: current understanding and future therapeutics. Expert Opin Emerg Drugs 2018; 23:331-347. [PMID: 30415584 DOI: 10.1080/14728214.2018.1547706] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Osteoarthritis (OA) is a leading cause of pain and disability among adults with a current prevalence of around 15% and a predicted prevalence of 35% in 2030 for symptomatic OA. It is increasingly recognized as a heterogeneous multi-faceted joint disease with multi-tissue involvement of varying severity. Current therapeutic regimens for OA are only partially effective and often have significant associated toxicities. There are no disease-modifying drugs approved by the regulatory bodies. Areas covered: We reviewed the opportunities within key OA pathogenetic mechanism: cartilage catabolism/anabolism, pathological remodeling of subchondral bone and synovial inflammation to identify targeted disease-modifying osteoarthritis drugs, based on compounds currently in Phase II and III stages of clinical development in which x-ray and/or MRI was used as the structural outcome with/without symptomatic outcomes according to regulatory requirements. Expert opinion: Given the heterogeneity of the OA disease process and complex overlapping among these phenotypes, a 'one size fits all' approach used in most clinical trials would unlikely be practical and equally effective in all patients, as well as in all anatomical OA sites. On the other hand, it is a challenge to develop a targeted drug with high activity, specificity, potency, and bioavailability in the absence of toxicity for long-term use in this chronic disease of predominantly older adults. Further research and insight into evaluation methods for drug-targeted identification of early OA and specific characterization of phenotypes, improvement of methodological designs, and development/refinement of sensitive imaging and biomarkers will help pave the way to the successful discovery of disease-modifying drugs and the optimal administration strategies in clinical practice.
Collapse
Affiliation(s)
- Win Min Oo
- a Rheumatology Department, Royal North Shore Hospital, and, Institute of Bone and Joint Research, Kolling Institute , University of Sydney , Sydney , Australia
| | - Shirley Pei-Chun Yu
- a Rheumatology Department, Royal North Shore Hospital, and, Institute of Bone and Joint Research, Kolling Institute , University of Sydney , Sydney , Australia
| | - Matthew Sean Daniel
- a Rheumatology Department, Royal North Shore Hospital, and, Institute of Bone and Joint Research, Kolling Institute , University of Sydney , Sydney , Australia
| | - David John Hunter
- a Rheumatology Department, Royal North Shore Hospital, and, Institute of Bone and Joint Research, Kolling Institute , University of Sydney , Sydney , Australia
| |
Collapse
|
560
|
Effect of hypoxia/reoxygenation on the biological effect of IGF system and the inflammatory mediators in cultured synoviocytes. Biochem Biophys Res Commun 2018; 508:17-24. [PMID: 30466784 DOI: 10.1016/j.bbrc.2018.11.099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/07/2018] [Accepted: 11/15/2018] [Indexed: 01/15/2023]
Abstract
Hypoxia/reoxygenation (H/R) plays an important role in the pathogenesis of osteoarthritis. Fibroblast-like synoviocytes (FLS), which are highly sensitive to H/R, are thought to be associated with cartilage degradation during osteoarthritis development. In this study, we investigated the biological effects of insulin-like growth factor (IGF) system and the expression of inflammatory mediators in FLS. We also pretreated FLS with tumor necrosis factor-α (TNF-α) before H/R in order to observe the response of FLS with the background of inflammatory cytokines. H/R increased the levels of TNF-α-induced C-C chemokine ligand 5 (CCL5), interleukin-1β (IL-1β) and interleukin-6 (IL-6) in cell-free culture supernatants; H/R also increased the expression of TNF-α-induced insulin-like growth factor binding protein 3 (IGFBP-3), downregulated the expression of insulin-like growth factor 1 (IGF-1), promoted the loss of mitochondrial membrane potential (MMP), the openness of mitochondrial permeability transition pore (MPTP), the release of intracellular reactive oxygen species (ROS), and mitochondrial matrix swelling, outer membrane rupture and decrease in cristae. Furthermore, H/R induced the expression of catabolic factors and activated the NF-κB signaling pathway in FLS. We therefore concluded that H/R may play a role in inducing inflammation and increase the TNF-α-induced inflammatory effect in FLS, contributing to osteoarthritis pathogenesis.
Collapse
|
561
|
Rakow A, Perka C, Trampuz A, Renz N. Origin and characteristics of haematogenous periprosthetic joint infection. Clin Microbiol Infect 2018; 25:845-850. [PMID: 30678837 DOI: 10.1016/j.cmi.2018.10.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Recognition of infectious origin of haematogenous periprosthetic joint infections (PJI) is crucial. We investigated the primary focus and characteristics of haematogenous PJI. METHODS Consecutive patients who presented with haematogenous PJI between 01/2010 and 01/2018 were retrospectively analysed. Haematogenous PJI was defined by diagnosis of infection ≥1 month after surgery, acute manifestation after a pain-free period and positive blood or prosthetic-site culture and/or evidence of distant infectious focus consistent with the pathogen. Fisher's exact, Student's t and Mann-Whitney U tests were used, as appropriate. RESULTS A total of 106 episodes of PJI were included, involving 59 knee, 45 hip, one shoulder and one elbow prostheses. The median time from last surgery until haematogenous PJI was 47 months (range, 1-417 months). The pathogen was identified in 105 episodes (99%), including Staphylococcus aureus (n = 43), streptococci (n = 32), enterococci (n = 13), Gram-negative bacteria (n = 9) and coagulase-negative staphylococci (n = 8). Gram-negative bacteria were significantly more often found in hip joints than in knee joints. Blood cultures grew the pathogen in 43 of 70 episodes (61%). The primary infectious focus was identified in 72 episodes (68%) and included infections of intravascular devices or heart valves (22 episodes), skin and soft tissue (16 episodes), the oral cavity (12 episodes), urogenital (12 episodes) or gastrointestinal tract (seven episodes) and other sites (three episodes). CONCLUSIONS In acute PJI manifesting after a pain-free period, the haematogenous infection route should be considered and the primary infectious focus should be actively searched for. The cardiovascular system, skin and soft tissue, oral cavity, urogenital and gastrointestinal tracts were common origins of haematogenous PJI.
Collapse
Affiliation(s)
- A Rakow
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Musculoskeletal Surgery (CMSC), Berlin, Germany
| | - C Perka
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Musculoskeletal Surgery (CMSC), Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - A Trampuz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Musculoskeletal Surgery (CMSC), Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - N Renz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Musculoskeletal Surgery (CMSC), Berlin, Germany.
| |
Collapse
|
562
|
Van Ginckel A, Hall M, Dobson F, Calders P. Effects of long-term exercise therapy on knee joint structure in people with knee osteoarthritis: A systematic review and meta-analysis. Semin Arthritis Rheum 2018; 48:941-949. [PMID: 30392703 DOI: 10.1016/j.semarthrit.2018.10.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/19/2018] [Accepted: 10/11/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate effects of long-term exercise therapy for people with knee osteoarthritis (OA) on radiographic disease severity and cartilage integrity (primary outcomes) as well as severity of bone marrow lesions (BMLs), synovitis and/or effusion (secondary outcomes). METHODS We sourced randomized controlled trials in people with clinical and/or radiographic OA investigating the effect of land-based exercise therapy of > 6 months on radiographic disease severity and magnetic resonance imaging outcomes of cartilage integrity (morphology or composition) as well as BML, synovitis and/or effusion severity, when compared to no exercise. Two independent reviewers extracted data and assessed risk of bias. Random-effects meta-analysis was used to pool standardised mean differences (SMD) (95% confidence intervals (CI)) or odds ratios (OR) (95% CI) and estimate heterogeneity (I2, %). Quality of the pooled body of evidence was rated implementing the GRADE approach. Studies unsuitable for meta-analysis were summarized in a best-evidence synthesis. RESULTS Meta-analysis showed moderate quality evidence of no treatment effect on tibiofemoral radiographic disease severity ((SMD) 95% (CI): 0.06 (-0.07, 0.20), I2 = 0%) and low-quality evidence of no effect on tibiofemoral cartilage morphology (SMD (95%): 0.06 (-0.20, 0.36), I2 = 0%). Low quality evidence revealed no treatment effect on the odds of change in synovitis ((OR) (95% CI): 0.90 (0.51,1.60), I2 = 0%) and effusion ((OR (95% CI): 0.88 (0.64, 1.20), I2 = 0%), but greater odds of tibiofemoral BMLs worsening (OR (95% CI): 1.90 (1.11, 3.26), I2 = 0%). In best-evidence synthesis, limited evidence was found for changes in patellar cartilage composition following exercise in women with mild knee OA compared to no exercise, but not for tibiofemoral cartilage. CONCLUSION Long-term exercise therapy did not change tibiofemoral radiographic disease severity, cartilage morphology or synovitis/effusion, but may slightly increase the likelihood for increased BML severity. Overall, meta-analysis findings were limited in directness and precision and restricted to relatively imprecise effect estimates in people who were obese on average. Limited evidence suggested some protective effects on patellar cartilage composition.
Collapse
Affiliation(s)
- Ans Van Ginckel
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Corneel Heymanslaan 10, Building B3, 9000 Ghent, Belgium.
| | - Michelle Hall
- Centre for Health, Exercise and Sports Medicine, Department of Physiotherapy, The University of Melbourne, Alan Gilbert Building, 161 Barry Street, Carlton, VIC 3053, Australia.
| | - Fiona Dobson
- Centre for Health, Exercise and Sports Medicine, Department of Physiotherapy, The University of Melbourne, Alan Gilbert Building, 161 Barry Street, Carlton, VIC 3053, Australia.
| | - Patrick Calders
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Corneel Heymanslaan 10, Building B3, 9000 Ghent, Belgium.
| |
Collapse
|
563
|
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a major cause of pain and disability worldwide. There is, however, a relatively poor correlation between the severity of OA based on plain radiograph changes and symptoms. In this review, we consider the mechanisms of pain in OA. RECENT FINDINGS It is now widely recognised that OA is a disease of the whole joint. Data from large observational studies which have used magnetic resonance imaging (MRI) suggest that pain in OA is associated with a number of structural factors including the presence of bone marrow lesions (BMLs) and also synovitis. There is evidence also of alterations in nerve processing and that both peripheral and central nerve sensitisation may contribute to pain in OA. Identification of the causes of pain in an individual patient may be of benefit in helping to better target with appropriate therapy to help reduce their symptoms and improve function.
Collapse
Affiliation(s)
- Terence W O'Neill
- Arthritis Research UK Centre for Epidemiology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, The Stopford Building, Oxford Road, Manchester, M13 9PT, UK. terence.o'
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK. terence.o'
- Salford Royal NHS Foundation Trust, Salford, UK. terence.o'
| | - David T Felson
- Arthritis Research UK Centre for Epidemiology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, The Stopford Building, Oxford Road, Manchester, M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
- Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
564
|
Mao G, Zhang Z, Hu S, Zhang Z, Chang Z, Huang Z, Liao W, Kang Y. Exosomes derived from miR-92a-3p-overexpressing human mesenchymal stem cells enhance chondrogenesis and suppress cartilage degradation via targeting WNT5A. Stem Cell Res Ther 2018; 9:247. [PMID: 30257711 PMCID: PMC6158854 DOI: 10.1186/s13287-018-1004-0] [Citation(s) in RCA: 309] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/30/2018] [Accepted: 09/03/2018] [Indexed: 12/13/2022] Open
Abstract
Background WNT5A is known to be involved in the pathogenesis of osteoarthritis. This study investigated the molecular mechanism of exosomal miR-92a-3p and WNT5A in chondrogenesis and cartilage degeneration. Methods Exosomal miR-92a-3p expression was assessed in vitro in a human mesenchymal stem cell (MSC) model of chondrogenesis and in normal and OA primary human chondrocytes (PHCs). MSCs and PHCs were treated with exosomes derived from MSC-miR-92a-3p (MSC-miR-92a-3p-Exos) or its antisense inhibitor (MSC-anti-miR-92a-3p-Exos), respectively. Small interfering RNAs (siRNAs) and luciferase reporter assay were used to reveal the molecular role of exosomal miR-92a-3p and WNT5A in chondrogenesis. The protective effect of exosomes in vivo was measured using Safranin-O and Fast Green staining and immunohistochemical staining. Results Exosomal miR-92a-3p expression was elevated in the MSC chondrogenic exosome, while it was significantly reduced in the OA chondrocyte-secreted exosome compared with normal cartilage. Treatment with MSC-miR-92a-3p-Exos promoted cartilage proliferation and matrix genes expression in MSCs and PHCs, respectively. In contrast, treatment with MSC-anti-miR-92a-3p-Exos repressed chondrogenic differentiation and reduced cartilage matrix synthesis by enhancing the expression of WNT5A. Luciferase reporter assay demonstrated that miR-92a-3p suppressed the activity of a reporter construct containing the 3’-UTR and inhibited WNT5A expression in both MSCs and PHCs. MSC-miR-92a-3p-Exos inhibit cartilage degradation in the OA mice model. Conclusions Our results suggest that exosomal miR-92a-3p regulates cartilage development and homeostasis by directly targeting WNT5A. This indicates that exosomal miR-92a-3p may act as a Wnt inhibitor and exhibits potential as a disease-modifying osteoarthritis drug. Electronic supplementary material The online version of this article (10.1186/s13287-018-1004-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guping Mao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ziji Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shu Hu
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zhiqi Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zongkun Chang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zhiyu Huang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Weiming Liao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Yan Kang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
565
|
Zhou J, Zhao Y, Wu G, Lin B, Li Z, Liu X. Differential miRNAomics of the synovial membrane in knee osteoarthritis induced by bilateral anterior cruciate ligament transection in rats. Mol Med Rep 2018; 18:4051-4057. [PMID: 30106113 DOI: 10.3892/mmr.2018.9385] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 07/13/2018] [Indexed: 11/05/2022] Open
Abstract
The differential microRNA (miRNA) omics of the synovial membrane were investigated using a rat model of knee osteoarthritis (KOA) induced by bilateral anterior cruciate ligament transection, which produced pathological biomarkers in KOA. Sprague‑Dawley rats were randomly divided into two groups; Sham‑operated and KOA‑operated group. The KOA rats were subjected to bilateral anterior cruciate ligament transection. After 6 weeks, total RNA was extracted from the knee joint synovial membrane of the rats and a microRNA (miR) microarray was performed to identify differentially expressed miRs. Subsequently, the obtained differentially expressed miRs were validated by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) analysis. A total of 24 miRs were identified with alterations ≥1.5‑fold in the synovial membrane in the KOA‑operated group compared with the sham‑operated group, of which 4 miRs (miR‑532‑5p, ‑200b‑5p, ‑377‑3p and ‑759‑5p) were decreased and 20 miRs (miR‑382‑3p, ‑223‑3p, ‑100‑5p, ‑30d‑5p, ‑183‑5p, ‑130, ‑92b‑3p, ‑125b‑3p, ‑151‑3p, ‑155‑3p, 27a‑3p, ‑146b‑3p, ‑885‑5p, ‑352, ‑184, ‑345‑5p, ‑30a‑5p and ‑9a‑5p) were increased. Subsequently, RT‑qPCR was used to validate the expressions of miR‑223, ‑100, ‑345, ‑130, ‑382, ‑377, ‑352, ‑200b, ‑9a and ‑183, which were upregulated by a fold change of ≥1.5 in synovial membranes of KOA rats compared with shams. Furthermore, in vitro miR‑223 mimic could suppress the luciferase activity of NACHT, LRR and PYD domains‑containing protein 3 (NLRP3) 3' untranslated region by detecting of dual luciferase reporter vector. Additionally, the expression of NLRP3, interleukin (IL)‑1β and IL‑18 significantly increased in the synovial membrane of KOA rats. A total of 24 different miRs were determined by comparing the miRNAomics in the synovial membrane of the KOA model rats. Furthermore, the miR‑233‑regulated NLRP3 inflammasome was implicated in synovial membrane injury, which may be an important mechanism of KOA pathogenesis.
Collapse
Affiliation(s)
- Jiangtao Zhou
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yina Zhao
- Department of Orthopedics and Traumatology, Affiliated Wenzhou Hospital of Traditional Chinese Medicine, Zhejiang University of Traditional Chinese Medicine, Wenzhou, Zhejiang 325000, P.R. China
| | - Guangwen Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Bingbing Lin
- Department of Pharmacy, Affiliated Wenzhou Hospital of Traditional Chinese Medicine, Zhejiang University of Traditional Chinese Medicine, Wenzhou, Zhejiang 325000, P.R. China
| | - Zuanfang Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xianxiang Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
566
|
Steen-Louws C, Popov-Celeketic J, Mastbergen SC, Coeleveld K, Hack CE, Eijkelkamp N, Tryfonidou M, Spruijt S, van Roon JAG, Lafeber FPJG. IL4-10 fusion protein has chondroprotective, anti-inflammatory and potentially analgesic effects in the treatment of osteoarthritis. Osteoarthritis Cartilage 2018; 26:1127-1135. [PMID: 29775732 DOI: 10.1016/j.joca.2018.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Effective disease-modifying drugs for osteoarthritis (DMOAD) should preferably have chondroprotective, anti-inflammatory, and analgesic activity combined in a single molecule. We developed a fusion protein of IL4 and IL10 (IL4-10 FP), in which the biological activity of both cytokines is preserved. The present study evaluates the chondroprotective, anti-inflammatory, and analgesic activity of IL4-10 FP in in vitro and in vivo models of osteoarthritis. METHODS Human osteoarthritic cartilage tissue and synovial tissue were cultured with IL4-10 FP. Cartilage proteoglycan turnover and release of pro-inflammatory, catabolic, and pain mediators by cartilage and synovial tissue were measured. The analgesic effect of intra-articularly injected IL4-10 FP was evaluated in a canine model of osteoarthritis by force-plate analysis. RESULTS IL4-10 FP increased synthesis (P = 0.018) and decreased release (P = 0.018) of proteoglycans by osteoarthritic cartilage. Release of pro-inflammatory IL6 and IL8 by cartilage and synovial tissue was reduced in the presence of IL4-10 FP (all P < 0.05). The release of MMP3 by osteoarthritic cartilage and synovial tissue was decreased (P = 0.018 and 0.028) whereas TIMP1 production was not significantly changed. Furthermore, IL4-10 FP protected cartilage against destructive properties of synovial tissue mediators shown by the increased cartilage proteoglycan synthesis (P = 0.0235) and reduced proteoglycan release (P = 0.0163). Finally, intra-articular injection of IL4-10 FP improved the deficient joint loading in dogs with experimentally induced osteoarthritis. CONCLUSION The results of current preliminary study suggest that IL4-10 FP has DMOAD potentials since it shows chondroprotective and anti-inflammatory effects in vitro, as well as potentially analgesic effect in a canine in vivo model of osteoarthritis.
Collapse
Affiliation(s)
- C Steen-Louws
- Laboratory of Translational Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| | - J Popov-Celeketic
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| | - S C Mastbergen
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| | - K Coeleveld
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| | - C E Hack
- Laboratory of Translational Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| | - N Eijkelkamp
- Laboratory of Translational Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands; Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| | - M Tryfonidou
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, University Utrecht, The Netherlands.
| | - S Spruijt
- St. Maartens Hospital, The Netherlands.
| | - J A G van Roon
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| | - F P J G Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, University Utrecht, The Netherlands.
| |
Collapse
|
567
|
Bay-Jensen A, Kjelgaard-Petersen C, Petersen K, Arendt-Nielsen L, Quasnichka H, Mobasheri A, Karsdal M, Leeming D. Aggrecanase degradation of type III collagen is associated with clinical knee pain. Clin Biochem 2018; 58:37-43. [DOI: 10.1016/j.clinbiochem.2018.04.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 04/17/2018] [Accepted: 04/23/2018] [Indexed: 11/16/2022]
|
568
|
Valdes AM, Ravipati S, Pousinis P, Menni C, Mangino M, Abhishek A, Chapman V, Barrett DA, Doherty M. Omega-6 oxylipins generated by soluble epoxide hydrolase are associated with knee osteoarthritis. J Lipid Res 2018; 59:1763-1770. [PMID: 29986999 PMCID: PMC6121933 DOI: 10.1194/jlr.p085118] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 07/02/2018] [Indexed: 11/30/2022] Open
Abstract
Omega-6 FAs are inflammatory mediators that are increased in joints with osteoarthritis (OA), but their association with OA progression is not yet well defined. To investigate the relationship between omega-6 FAs and knee OA, we measured with LC-MS the levels of 22 omega-6 lipids (arachidonic acid, linoleic acid, and 20 oxylipins) in synovial fluid (SF) from 112 knees of 102 individuals (58 with knee OA; 44 controls). We hypothesized that oxylipin metabolites would increase in OA knee SF and with radiographically progressive disease. We validated results by comparing samples from affected and unaffected knees in 10 individuals with unilateral OA. In adjusted analysis, SF levels of three omega-6 oxylipins [prostaglandin D2, 11,12-dihydroxyeicosatrienoic acid (DHET), and 14,15-DHET] were associated with OA. Of these, 11,12-DHET and 14,15-DHET were higher in affected versus unaffected knees of people with unilateral disease (P < 0.014 and P < 0.003, respectively). Levels of these and 8,9-DHET were also associated with radiographic progression over 3.3 years in 87 individuals. Circulating levels of all three were associated with gene variants at the soluble epoxide hydrolase enzyme. Lipidomic profiling in SF identified an additional inflammatory pathway associated with knee OA and radiographic progression.
Collapse
Affiliation(s)
- Ana M Valdes
- Academic Rheumatology, Nottingham City Hospital, Nottingham, United Kingdom; National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom; Department of Twin Research, King's College London, St Thomas' Hospital, London, United Kingdom; Arthritis Research UK Centre of Excellence for Pain, University of Nottingham, Nottingham, United Kingdom.
| | - Srinivasarao Ravipati
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Petros Pousinis
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Cristina Menni
- Department of Twin Research, King's College London, St Thomas' Hospital, London, United Kingdom
| | - Massimo Mangino
- Department of Twin Research, King's College London, St Thomas' Hospital, London, United Kingdom
| | - Abhishek Abhishek
- Academic Rheumatology, Nottingham City Hospital, Nottingham, United Kingdom
| | - Victoria Chapman
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom; Arthritis Research UK Centre of Excellence for Pain, University of Nottingham, Nottingham, United Kingdom
| | - David A Barrett
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Michael Doherty
- Academic Rheumatology, Nottingham City Hospital, Nottingham, United Kingdom; National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom; Arthritis Research UK Centre of Excellence for Pain, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
569
|
Knee Joint Tissues Effectively Separate Mixed Sized Molecules Delivered in a Single Bolus to the Heart. Sci Rep 2018; 8:10254. [PMID: 29980695 PMCID: PMC6035244 DOI: 10.1038/s41598-018-28228-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 06/19/2018] [Indexed: 12/17/2022] Open
Abstract
The role of molecular size selectivity in the onset and progression of osteoarthritis (OA), a degenerative disease of the musculoskeletal system and the most common cause of disability in aging adults, is unknown. Here we delivered a mixture of Texas-red (70 kDa), and Rhodamine-green (10 kDa) tagged, dextrans of neutral charge in a single bolus via heart injection to middle aged (8-10 months) and aged (17-19 months) Dunkin-Hartley Guinea pigs, a natural model for OA. We quantified tracer transport in serial-sectioned, cryofixed block specimens after five minutes' circulation. A remarkable separation of the molecules was observed in serial fluorescent images of whole joint sections. The larger, 70 kDa red tracer was abundant in the marrow cavity albeit less prevalent or absent in the bone, cartilage, meniscus and other tissues of the joint. Tissues of the meniscus, ligament, and tendon exhibited abundant 10 kDa tracer; volumes of tissue containing this molecular tracer were significantly lower in older than in younger animals. Surprisingly, muscle fiber bundles exhibited little fluorescence, while their bounding fasciae fluoresced either red or green. Small caliber channels through the articular cartilage appeared to show a degree of green fluorescence not observed in the surrounding cartilage matrix. This study opens up new avenues for study of musculoskeletal physiology in health and disease as well as new strategies for drug delivery.
Collapse
|
570
|
Konieczynski P, Szreder G, Tamowska E, Wesolowski M. Essential elements in synovial fluid samples obtained from patients living in Northern Poland. J Trace Elem Med Biol 2018; 48:20-24. [PMID: 29773182 DOI: 10.1016/j.jtemb.2018.02.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/16/2018] [Accepted: 02/26/2018] [Indexed: 11/21/2022]
Abstract
Many people after 50 year of life suffer pains caused by osteoarthritis (OA), which make their life difficult and painful. There is a probable association of OA with the concentration of certain elements in body fluids. Therefore the aim of the study was to relate the level of essential elements in synovial fluids of patients with OA with the treatment using antiarthritic drugs. Thus, flame-atomic absorption spectrometry was used to determine the concentrations of Fe, Zn, Cu, Mn, Na, K, Ca and Mg in 74 samples of synovial fluids taken from 36 patients. Prior to analysis, the samples of synovial fluids were centrifuged and obtained supernatants were digested by microwave technique. It was found that the level of investigated essential elements was differentiated in patients suffering with OA. High correlation between Zn and K, Na and K, as well as Cu and Mn were revealed. Based on cluster analysis, synovial fluids of patients treated with Diprophos, Euflexxa and Synocrom, can be found almost in all clusters. ANOVA has shown that there is a significant difference between Na level in synovial fluids of patients treated with antirheumatic drugs, and those who were not treated. For the patients treated with Euflexxa, similar relations for Ca and K exist. Principal component analysis has shown that essential elements responsible for variability within studied samples, are Ca, Mg, Cu, Mn and Fe. However, there were no statistically significant relations between the studied macro- and microelements, and treatment using specific preparations.
Collapse
Affiliation(s)
- Pawel Konieczynski
- Medical University of Gdansk, Department of Analytical Chemistry, Gen. J. Hallera 107, 80-416, Gdansk, Poland.
| | | | - Ewelina Tamowska
- Medical University of Gdansk, Department of Analytical Chemistry, Gen. J. Hallera 107, 80-416, Gdansk, Poland
| | - Marek Wesolowski
- Medical University of Gdansk, Department of Analytical Chemistry, Gen. J. Hallera 107, 80-416, Gdansk, Poland
| |
Collapse
|
571
|
Qadri MM, Jay GD, Ostrom RS, Zhang LX, Elsaid KA. cAMP attenuates TGF-β's profibrotic responses in osteoarthritic synoviocytes: involvement of hyaluronan and PRG4. Am J Physiol Cell Physiol 2018; 315:C432-C443. [PMID: 29898378 DOI: 10.1152/ajpcell.00041.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is characterized by synovitis and synovial fibrosis. Synoviocytes are fibroblast-like resident cells of the synovium that are activated by transforming growth factor (TGF)-β to proliferate, migrate, and produce extracellular matrix. Synoviocytes secrete hyaluronan (HA) and proteoglycan-4 (PRG4). HA reduces synovial fibrosis in vivo, and the Prg4-/- mouse exhibits synovial hyperplasia. We investigated the antifibrotic effects of increased intracellular cAMP in TGF-β-stimulated human OA synoviocytes. TGF-β1 stimulated collagen I (COL1A1), α-smooth muscle actin (α-SMA), tissue inhibitor of metalloproteinase (TIMP)-1, and procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) expression, and procollagen I, α-SMA, HA, and PRG4 production, migration, and proliferation of OA synoviocytes were measured. Treatment of OA synoviocytes with forskolin (10 μM) increased intracellular cAMP levels and reduced TGF-β1-stimulated COL1A1, α-SMA, and TIMP-1 expression, with no change in PLOD2 expression. Forskolin also reduced TGF-β1-stimulated procollagen I and α-SMA content as well as synoviocyte migration and proliferation. Forskolin (10 μM) increased HA secretion and PRG4 expression and production. A cell-permeant cAMP analog reduced COL1A1 and α-SMA expression and enhanced HA and PRG4 secretion by OA synoviocytes. HA and PRG4 reduced α-SMA expression and content, and PRG4 reduced COL1A1 expression and procollagen I content in OA synoviocytes. Prg4-/- synovium exhibited increased α-SMA, COL1A1, and TIMP-1 expression compared with Prg4+/+ synovium. Prg4-/- synoviocytes demonstrated strong α-SMA and collagen type I staining, whereas these were undetected in Prg4+/+ synoviocytes and were reduced with PRG4 treatment. We conclude that increasing intracellular cAMP levels in synoviocytes mitigates synovial fibrosis through enhanced production of HA and PRG4, possibly representing a novel approach for treatment of OA synovial fibrosis.
Collapse
Affiliation(s)
- Marwa M Qadri
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University , Irvine, California
| | - Gregory D Jay
- Department of Emergency Medicine, Rhode Island Hospital , Providence, Rhode Island.,Department of Biomedical Engineering, Brown University , Providence, Rhode Island
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University , Irvine, California
| | - Ling X Zhang
- Department of Emergency Medicine, Rhode Island Hospital , Providence, Rhode Island
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University , Irvine, California
| |
Collapse
|
572
|
Macchi V, Stocco E, Stecco C, Belluzzi E, Favero M, Porzionato A, De Caro R. The infrapatellar fat pad and the synovial membrane: an anatomo-functional unit. J Anat 2018; 233:146-154. [PMID: 29761471 PMCID: PMC6036933 DOI: 10.1111/joa.12820] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2018] [Indexed: 01/15/2023] Open
Abstract
The infrapatellar pad, a fibro-adipose tissue with peculiar microscopic and mechanical features, is gaining wide attention in the field of rheumatological research. The purpose of this descriptive review is to summarize the most recent published evidence on the anatomic, physiologic and biomechanical inter-relationship between the infrapatellar fat pad and the knee synovial membrane. As an extrasynovial tissue, the infrapatellar fat pad does not directly interact with the articular cartilage; based on its location in close contact with the synovial membrane, and due to the metabolic properties of adipose tissue, it may influence the behavior of the synovial membrane. In fact, considering evidence of macroscopic and microscopic anatomy, the infrapatellar fat pad is the site of insertion of the infrapatellar and medial synovial plicae. Also biochemically, there is much evidence highlighting the interaction among these two structures; in the case of inflammation, the mutual interplay is ascribable to the release of pro-inflammatory mediators stimulating the proliferation of inflammatory cells and promoting tissue modifications in both. All these assumptions could support the emerging idea that the infrapatellar fat pad and the synovial membrane may be considered a morpho-functional unit.
Collapse
Affiliation(s)
- Veronica Macchi
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Elena Stocco
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Carla Stecco
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Elisa Belluzzi
- Musculoskeletal Pathology and Oncology Laboratory, Department of Orthopaedics and Orthopaedics Oncology, University of Padova, Padova, Italy.,Rheumatology Unit, Department of Medicine-DIMED, University Hospital of Padova, Padova, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine-DIMED, University Hospital of Padova, Padova, Italy
| | - Andrea Porzionato
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Raffaele De Caro
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| |
Collapse
|
573
|
McAllister MJ, Chemaly M, Eakin AJ, Gibson DS, McGilligan VE. NLRP3 as a potentially novel biomarker for the management of osteoarthritis. Osteoarthritis Cartilage 2018; 26:612-619. [PMID: 29499288 DOI: 10.1016/j.joca.2018.02.901] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 02/09/2018] [Accepted: 02/20/2018] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) was previously thought of as 'wear and tear' as humans age, however there is increasing evidence to support an inflammatory theory. The nucleotide-binding and oligomerization domain-like receptor containing protein 3 (NLRP3) inflammasome has been implicated in the pathogenesis of a number of arthritic disorders, producing proinflammatory cytokines and degradative enzymes such as Interleukin-1 beta (IL-1β), Tumour necrosis factor alpha (TNF-α) and Matrix metalloproteinase-3 (MMP-3) which drive cartilage degeneration and synovial inflammation. This review aims to summarise the evidence of NLRP3 involvement in OA. Currently, treatment options focus on management of the disease and to date there is no cure. The development of novel biomarkers for OA could improve diagnosis, treatment and management. Importantly, this review provides detail on the involvement of the NLRP3 inflammasome in OA pathology and how its members could act as potential biomarkers to assist clinical decisions.
Collapse
Affiliation(s)
- M J McAllister
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom.
| | - M Chemaly
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom
| | - A J Eakin
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom
| | - D S Gibson
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom.
| | - V E McGilligan
- Northern Ireland Centre for Stratified Medicine, Ulster University, Altnagelvin Hospital, Glenshane Road, Londonderry, United Kingdom
| |
Collapse
|
574
|
Lee MC, Saleh R, Achuthan A, Fleetwood AJ, Förster I, Hamilton JA, Cook AD. CCL17 blockade as a therapy for osteoarthritis pain and disease. Arthritis Res Ther 2018; 20:62. [PMID: 29622035 PMCID: PMC5887260 DOI: 10.1186/s13075-018-1560-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/06/2018] [Indexed: 01/12/2023] Open
Abstract
Background Granulocyte macrophage-colony stimulating factor (GM-CSF) has been implicated in the pathogenesis of a number of inflammatory diseases and in osteoarthritis (OA). We identified previously a new GM-CSF→Jmjd3→interferon regulatory factor 4 (IRF4)→chemokine (c-c motif) ligand 17 (CCL17) pathway, which is important for the development of inflammatory arthritis pain and disease. Tumour necrosis factor (TNF) can also be linked with this pathway. Here we investigated the involvement of the pathway in OA pain and disease development using the GM-CSF-dependent collagenase-induced OA (CiOA) model. Methods CiOA was induced in C57BL/6 wild-type (WT), Irf4−/−, Ccl17E/E, Ccr4−/−, Tnf−/− and GM-CSF−/− mice. Additionally, therapeutic targeting of CCL17, Jmjd3 and cyclooxygenase 2 (COX-2) was evaluated. Development of pain (assessment of weight distribution) and OA disease (histologic scoring of synovitis, cartilage destruction and osteophyte size) were assessed. Synovial joint cells, including neutrophils, macrophages, fibroblasts and endothelial cells, were isolated (cell sorting) and gene expression analyzed (quantitative PCR). Results Studies in the gene-deficient mice indicated that IRF4, CCL17 and the CCL17 receptor, CCR4, but not TNF, were required for CiOA pain and optimal cartilage destruction and osteophyte size. Therapeutic neutralization of CCL17 and Jmjd3 ameliorated both pain and disease, whereas the COX-2 inhibitor only ameliorated pain. In the synovium Ccl17 mRNA was expressed only in the macrophages in a GM-CSF-dependent and IRF4-dependent manner. Conclusions The GM-CSF→Jmjd3→IRF4→CCL17 pathway is important for the development of CiOA, with CCL17 thus being a potential therapeutic target for the treatment of both OA pain and disease. Electronic supplementary material The online version of this article (10.1186/s13075-018-1560-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ming-Chin Lee
- The University of Medicine, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Reem Saleh
- The University of Medicine, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Adrian Achuthan
- The University of Medicine, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Andrew J Fleetwood
- The University of Medicine, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences Institute, University of Bonn, 53115, Bonn, Germany
| | - John A Hamilton
- The University of Medicine, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Andrew D Cook
- The University of Medicine, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.
| |
Collapse
|
575
|
Bergström B, Carlsten H, Ekwall AKH. Methotrexate inhibits effects of platelet-derived growth factor and interleukin-1β on rheumatoid arthritis fibroblast-like synoviocytes. Arthritis Res Ther 2018; 20:49. [PMID: 29554943 PMCID: PMC5859417 DOI: 10.1186/s13075-018-1554-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/25/2018] [Indexed: 11/23/2022] Open
Abstract
Background A key feature of joints in rheumatoid arthritis (RA) is the formation of hyperplastic destructive pannus tissue, which is orchestrated by activated fibroblast-like synoviocytes (FLS). We have demonstrated that the RA risk gene and tumor suppressor Limb bud and heart development (LBH) regulates cell cycle progression in FLS. Methotrexate (MTX) is the first-line treatment for RA, but its mechanisms of action remain incompletely understood. Here, we studied the effects of MTX on mitogen-induced FLS proliferation and expression of cell cycle regulators in vitro. Methods Primary FLS from patients with RA or osteoarthritis were stimulated with the mitogen platelet-derived growth factor (PDGF) and the cytokine interleukin-1β (IL-1β) in the presence or absence of MTX. Cells were then subjected to qPCR for gene expression and cell cycle analysis by flow cytometry. Results Stimulation with PDGF and IL-1β increased the percentage of FLS in the G2/M phase and shifted the cell morphology to a dendritic shape. These effects were inhibited by MTX. Furthermore, PDGF + IL-1β reduced LBH mRNA expression. However, MTX treatment yielded significantly higher transcript levels of LBH, and of CDKN1A (p21) and TP53 (p53), compared to untreated samples upon mitogen stimulation. The expression of DNA methyltransferase-1 (DNMT1) was also higher in the presence of MTX and there was strong correlation between DNMT1 and LBH expression. Conclusions Therapeutic concentrations of MTX abolish the effects of PDGF and IL-1β on tumor suppressor expression and inhibit mitogen-promoted FLS proliferation. These data demonstrate novel and important effects of MTX on pathogenic effector cells in the joint, which might involve epigenetic mechanisms.
Collapse
Affiliation(s)
- Beatrice Bergström
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Box 480, 405 30, Gothenburg, Sweden.,Centre for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden
| | - Hans Carlsten
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Box 480, 405 30, Gothenburg, Sweden.,Centre for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Hultgård Ekwall
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Box 480, 405 30, Gothenburg, Sweden. .,Centre for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
576
|
Bhattaram P, Muschler G, Wixler V, Lefebvre V. Inflammatory Cytokines Stabilize SOXC Transcription Factors to Mediate the Transformation of Fibroblast-Like Synoviocytes in Arthritic Disease. Arthritis Rheumatol 2018; 70:371-382. [PMID: 29193895 PMCID: PMC5826855 DOI: 10.1002/art.40386] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/16/2017] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLS) produce key synovial fluid and tissue components to ensure joint integrity under healthy conditions, whereas they become cancer-like and aggressively contribute to joint degeneration in inflammatory arthritis. The aim of this study was to determine whether the SOXC transcription factors SOX4 and SOX11, whose functions are critical in joint development and many cancer types, contribute to FLS activities under normal and inflammatory conditions. METHODS We inactivated the SOXC genes in FLS from adult mice and studied the effect on joint homeostasis and tumor necrosis factor (TNF)-induced arthritis. We used primary cells and synovial biopsy specimens from arthritis patients to analyze the interactions between inflammatory signals and SOXC proteins. RESULTS Postnatal inactivation of the SOXC genes had no major effect on joint integrity in otherwise healthy mice. However, it hampered synovial hyperplasia and joint degeneration in transgenic mice expressing human TNF. These effects were explained by the ability of SOX4/11 to amplify the pathogenic impact of TNF on FLS by increasing their survival and migration. SOXC RNA levels were not changed by TNF and other proinflammatory cytokines, but SOXC proteins were strongly stabilized and able to potentiate the TNF-induced up-regulation of genes involved in FLS transformation. Substantiating the relevance of these findings in human disease, SOXC protein levels, but not RNA levels, were significantly higher in inflamed synovium than in noninflamed synovium from arthritis patients. CONCLUSION SOXC proteins are targets and pivotal mediators of proinflammatory cytokines during FLS transformation in arthritic diseases. Targeting of these proteins could thus improve current strategies to treat arthritic diseases and possibly other inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Viktor Wixler
- Muenster University Hospital Medical School, Muenster, Germany
| | | |
Collapse
|
577
|
Suzuki T, Takakubo Y, Oki H, Liu X, Honma R, Naganuma Y, Goodman SB, Kaneko MK, Kato Y, Takagi M. Immunohistochemical Analysis of Inflammatory Rheumatoid Synovial Tissues Using Anti-Human Podoplanin Monoclonal Antibody Panel. Monoclon Antib Immunodiagn Immunother 2018; 37:12-19. [PMID: 29377768 DOI: 10.1089/mab.2017.0047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Podoplanin (PDPN) is a transmembrane sialoglycoprotein, which is expressed in several normal tissues and malignant tumors. Although PDPN expression in rheumatoid arthritis (RA) has been reported, the role of PDPN in RA and other arthritic conditions has not been fully elucidated. In this study, we examined PDPN expression in inflammatory synovial tissues using an anti-human PDPN (hPDPN) monoclonal antibody (mAb) panel to select the most useful one for evaluation of synovitis. Synovial tissue samples were obtained from 11 RA patients and 9 osteoarthritis (OA) patients undergoing joint surgery. PDPN-positive cells were immunostained by a panel of PDPN mAbs (NZ-1, LpMab-3, LpMab-7, LpMab-10, LpMab-12, LpMab-13, and LpMab-17), followed by cell grading of inflammation and cell counting of PDPN-positivity by a quantitative analyzer. Immunohistochemistry showed that PDPN was markedly expressed in both macrophage-like type A and fibroblast-like type B lining cells of the hyperplastic synovial lining cell layer, and macrophages and fibroblasts in the stroma of RA. Among anti-PDPN mAbs, LpMab-12 showed the highest score. In inflammatory OA synovium, PDPN expression was also detectable. Although LpMab-12 also showed the highest score in OA, the difference was not statistically significant. The inflammatory synovitis score of RA was significantly higher than that of OA. PDPN was expressed in inflammatory lining cells and sublining stroma of RA and OA synovium. In the seven anti-hPDPN antibodies examined, LpMab-12 was the most stainable antibody for PDPN in RA synovitis. Thus, LpMab-12 for PDPN has a possible and promising specific biomarker for evaluating synovitis in RA and inflammatory OA.
Collapse
Affiliation(s)
- Tomoto Suzuki
- 1 Department of Orthopaedic Surgery, Faculty of Medicine, Yamagata University , Yamagata, Japan
| | - Yuya Takakubo
- 1 Department of Orthopaedic Surgery, Faculty of Medicine, Yamagata University , Yamagata, Japan
| | - Hiroharu Oki
- 1 Department of Orthopaedic Surgery, Faculty of Medicine, Yamagata University , Yamagata, Japan
| | - Xing Liu
- 1 Department of Orthopaedic Surgery, Faculty of Medicine, Yamagata University , Yamagata, Japan
| | - Ryusuke Honma
- 1 Department of Orthopaedic Surgery, Faculty of Medicine, Yamagata University , Yamagata, Japan
| | - Yasushi Naganuma
- 1 Department of Orthopaedic Surgery, Faculty of Medicine, Yamagata University , Yamagata, Japan
| | - Stuart B Goodman
- 2 Department of Orthopaedic Surgery, Stanford University , Stanford, California
| | - Mika K Kaneko
- 3 Department of Antibody Drug Development, Tohoku University Graduate School of Medicine , Sendai, Japan
| | - Yukinari Kato
- 3 Department of Antibody Drug Development, Tohoku University Graduate School of Medicine , Sendai, Japan .,4 New Industry Creation Hatchery Center, Tohoku University , Sendai, Japan
| | - Michiaki Takagi
- 1 Department of Orthopaedic Surgery, Faculty of Medicine, Yamagata University , Yamagata, Japan
| |
Collapse
|
578
|
Identification of genes and pathways in the synovia of women with osteoarthritis by bioinformatics analysis. Mol Med Rep 2018; 17:4467-4473. [PMID: 29344651 PMCID: PMC5802222 DOI: 10.3892/mmr.2018.8429] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/05/2017] [Indexed: 01/12/2023] Open
Abstract
Osteoarthritis (OA) has a high prevalence in female patients and sex may be a key factor affecting the progression of OA. The aim of the present study was to identify genetic signatures in the synovial membranes of female patients with OA and to elucidate the potential associated molecular mechanisms. The gene expression profiles of the GSE55457 and GSE55584 datasets were obtained from the Gene Expression Omnibus database. Data of two synovial membranes from normal female individuals (GSM1337306 and GSM1337310) and two synovial membranes from female patients affected by OA (GSM1337327 and GSM1337330) were obtained from the dataset GSE55457, and those of three synovial membranes from female patients affected by OA (GSM1339628, GSM1339629 and GSM1339632) were obtained from the dataset GSE55584. Differentially expressed genes (DEGs) were identified by using Morpheus software. Protein-protein interaction (PPI) networks of the DEGs were constructed by using Cytoscape software. Subsequently, Gene Ontology (GO) function and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analyses of the top module of the PPI network were performed by using ClueGo. A total of 377 DEGs were identified in the synovial membranes of OA patients compared with those of normal individuals, including 164 upregulated and 213 downregulated genes. The top 10 hub genes were ubiquitin (UB)C, ribosomal protein (RP) L23A, mammalian target of rapamycin, heat shock protein 90 α family class A member 1, RPS28, RPL37A, RPS24, RPS4X, RPS18 and UBB. The results of the GO analysis indicated that the DEGs included in the top module of the PPI were mainly enriched in the terms ‘nuclear-transcribed mRNA catabolic process’, ‘nonsense mediated decay’, and ‘cytoplasmic translation and ribosomal small subunit biogenesis’. KEGG pathway analysis indicated that the DEGs included in the top one module were mainly enriched in the ‘ribosome’ pathway. The present study provides a systematic, molecular-level understanding of the degeneration of the synovial membrane in the progression of OA in female patients. The hub genes and molecules associated with the synovial membrane may be used as biomarkers and therapeutic targets for the treatment of OA in female patients with OA.
Collapse
|
579
|
Silawal S, Triebel J, Bertsch T, Schulze-Tanzil G. Osteoarthritis and the Complement Cascade. CLINICAL MEDICINE INSIGHTS. ARTHRITIS AND MUSCULOSKELETAL DISORDERS 2018; 11:1179544117751430. [PMID: 29434479 PMCID: PMC5805003 DOI: 10.1177/1179544117751430] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022]
Abstract
Accumulating evidence demonstrates that complement activation is involved in the pathogenesis of osteoarthritis (OA). However, the intimate complement regulation and cross talk with other signaling pathways in joint-associated tissues remain incompletely understood. Recent insights are summarized and discussed here, to put together a more comprehensive picture of complement involvement in OA pathogenesis. Complement is regulated by several catabolic and inflammatory mediators playing a key role in OA. It seems to be involved in many processes observed during OA development and progression, such as extracellular cartilage matrix (ECM) degradation, chondrocyte and synoviocyte inflammatory responses, cell lysis, synovitis, disbalanced bone remodeling, osteophyte formation, and stem cell recruitment, as well as cartilage angiogenesis. In reverse, complement can be activated by various ECM components and their cleavage products, which are released during OA-associated cartilage degradation. There are, however, some other cartilage ECM components that can inhibit complement, underlining the diverse effects of ECM on the complement activation. It is hypothesized that complement might also be directly activated by mechanical stress, thereby contributing to OA. The question arises whether keeping the complement activation in balance could represent a future therapeutic strategy in OA treatment and in the prevention of its progression.
Collapse
Affiliation(s)
- Sandeep Silawal
- Department of Anatomy, Paracelsus Medical University, Nuremberg, Germany
- Institute of Anatomy, Paracelsus Medical University, Salzburg, Germany
| | - Jakob Triebel
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Thomas Bertsch
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Gundula Schulze-Tanzil
- Department of Anatomy, Paracelsus Medical University, Nuremberg, Germany
- Institute of Anatomy, Paracelsus Medical University, Salzburg, Germany
| |
Collapse
|
580
|
O’Donnell JM, Devitt BM, Arora M. The role of the ligamentum teres in the adult hip: redundant or relevant? A review. J Hip Preserv Surg 2018; 5:15-22. [PMID: 29423246 PMCID: PMC5798146 DOI: 10.1093/jhps/hnx046] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 08/21/2017] [Accepted: 12/12/2017] [Indexed: 12/14/2022] Open
Abstract
The ligamentum teres (LT) has traditionally been described as a redundant structure with no contribution to hip biomechanics or function. There has been renewed interest in the LT as a source of hip pathology due to the high prevalence of LT pathology observed at the time of hip arthroscopy. The LT acts a secondary stabilizer to supplement the work of the capsular ligaments and works in a sling-like manner to prevent subluxation of the hip at the extremes of motion. The presence of free nerve endings within the LT indicates a definite role in pain generation, with the LT undergoing various mechanical and histological adaptations to hip pathology.
Collapse
Affiliation(s)
- John M O’Donnell
- Hip Arthroscopy Australia, 21 Erin Street, Richmond, VIC 3121, Australia
| | - Brian M Devitt
- Orthosport Victoria, 89 Bridge Road, Richmond, VIC 3121, Australia
| | - Manit Arora
- Hip Arthroscopy Australia, 21 Erin Street, Richmond, VIC 3121, Australia
| |
Collapse
|
581
|
Rohdewald PJ. Review on Sustained Relief of Osteoarthritis Symptoms with a Proprietary Extract from Pine Bark, Pycnogenol. J Med Food 2018; 21:1-4. [PMID: 28836883 PMCID: PMC5775113 DOI: 10.1089/jmf.2017.0015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 07/20/2017] [Indexed: 12/25/2022] Open
Abstract
This review summarizes the effects of the standardized proprietary bark extract of the French maritime pine (Pycnogenol®) in mild osteoarthritis (OA), stage 1 and 2. The extract exerts antioxidative, anti-inflammatory, and chondroprotective effects in vitro and in vivo. Its phenolic acids as well as catechin and taxifolin are quickly absorbed. Active metabolites, produced by gut microbiota in the intestinal tract from oligomeric procyanidins, appear in blood 6 h following ingestion and remain for at least 14 h, providing a long-lasting flow of anti-inflammatory substances for relief of OA symptoms. These constituents of Pycnogenol could be detected in serum, blood cells, and synovial fluid of OA patients. The resulting inhibition of cartilage-destructing proteases and pain-producing cyclo-oxygenases provides the basis for relief from pain, improvement of stiffness, enhanced mobility, and well-being in three clinical studies with the pine bark extract as an adjunct supplement. Sparing the use of nonsteroidal anti-inflammatory drugs, supplementation with the pine bark extract reduced gastric complications and hospital admissions of OA patients. Because of its favorable safety profile and sustained anti-inflammatory action, Pycnogenol represents an option as an add-on supplement for OA patients.
Collapse
Affiliation(s)
- Peter Jörg Rohdewald
- Institute for Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität , Münster, Germany
| |
Collapse
|
582
|
Vonk LA, van Dooremalen SFJ, Liv N, Klumperman J, Coffer PJ, Saris DB, Lorenowicz MJ. Mesenchymal Stromal/stem Cell-derived Extracellular Vesicles Promote Human Cartilage Regeneration In Vitro. Am J Cancer Res 2018; 8:906-920. [PMID: 29463990 PMCID: PMC5817101 DOI: 10.7150/thno.20746] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 10/08/2017] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is a rheumatic disease leading to chronic pain and disability with no effective treatment available. Recently, allogeneic human mesenchymal stromal/stem cells (MSC) entered clinical trials as a novel therapy for OA. Increasing evidence suggests that therapeutic efficacy of MSC depends on paracrine signalling. Here we investigated the role of extracellular vesicles (EVs) secreted by human bone marrow derived MSC (BMMSC) in human OA cartilage repair. Methods: To test the effect of BMMSC-EVs on OA cartilage inflammation, TNF-alpha-stimulated OA chondrocyte monolayer cultures were treated with BMMSC-EVs and pro-inflammatory gene expression was measured by qRT-PCR after 48 h. To assess the impact of BMMSC-EVs on cartilage regeneration, BMMSC-EVs were added to the regeneration cultures of human OA chondrocytes, which were analyzed after 4 weeks for glycosaminoglycan content by 1,9-dimethylmethylene blue (DMMB) assay. Furthermore, paraffin sections of the regenerated tissue were stained for proteoglycans (safranin-O) and type II collagen (immunostaining). Results: We show that BMMSC-EVs inhibit the adverse effects of inflammatory mediators on cartilage homeostasis. When co-cultured with OA chondrocytes, BMMSC-EVs abrogated the TNF-alpha-mediated upregulation of COX2 and pro-inflammatory interleukins and inhibited TNF-alpha-induced collagenase activity. BMMSC-EVs also promoted cartilage regeneration in vitro. Addition of BMMSC-EVs to cultures of chondrocytes isolated from OA patients stimulated production of proteoglycans and type II collagen by these cells. Conclusion: Our data demonstrate that BMMSC-EVs can be important mediators of cartilage repair and hold great promise as a novel therapeutic for cartilage regeneration and osteoarthritis.
Collapse
|
583
|
Labens R, Daniel C, Hall S, Xia XR, Schwarz T. Effect of intra-articular administration of superparamagnetic iron oxide nanoparticles (SPIONs) for MRI assessment of the cartilage barrier in a large animal model. PLoS One 2017; 12:e0190216. [PMID: 29287105 PMCID: PMC5747449 DOI: 10.1371/journal.pone.0190216] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/11/2017] [Indexed: 11/22/2022] Open
Abstract
Early diagnosis of cartilage disease at a time when changes are limited to depletion of extracellular matrix components represents an important diagnostic target to reduce patient morbidity. This report is to present proof of concept for nanoparticle dependent cartilage barrier imaging in a large animal model including the use of clinical magnetic resonance imaging (MRI). Conditioned (following matrix depletion) and unconditioned porcine metacarpophalangeal cartilage was evaluated on the basis of fluorophore conjugated 30 nm and 80 nm spherical gold nanoparticle permeation and multiphoton laser scanning and bright field microscopy after autometallographic particle enhancement. Consequently, conditioned and unconditioned joints underwent MRI pre- and post-injection with 12 nm superparamagnetic iron oxide nanoparticles (SPIONs) to evaluate particle permeation in the context of matrix depletion and use of a clinical 1.5 Tesla MRI scanner. To gauge the potential pro-inflammatory effect of intra-articular nanoparticle delivery co-cultures of equine synovium and cartilage tissue were exposed to an escalating dose of SPIONs and IL-6, IL-10, IFN-γ and PGE2 were assessed in culture media. The chemotactic potential of growth media samples was subsequently assessed in transwell migration assays on isolated equine neutrophils. Results demonstrate an increase in MRI signal following conditioning of porcine joints which suggests that nanoparticle dependent compositional cartilage imaging is feasible. Tissue culture and neutrophil migration assays highlight a dose dependent inflammatory response following SPION exposure which at the imaging dose investigated was not different from controls. The preliminary safety and imaging data support the continued investigation of nanoparticle dependent compositional cartilage imaging. To our knowledge, this is the first report in using SPIONs as intra-articular MRI contrast agent for studying cartilage barrier function, which could potentially lead to a new diagnostic technique for early detection of cartilage disease.
Collapse
Affiliation(s)
- Raphael Labens
- School of Animal and Veterinary Sciences, Faculty of Science, Charles Sturt University, Wagga Wagga, New South Wales, Australia
- * E-mail:
| | - Carola Daniel
- The Roslin Institute, Easter Bush Campus, The University of Edinburgh, Midlothian, United Kingdom
| | - Sarah Hall
- Animal & Veterinary Sciences, Scotland’s Rural College, Easter Bush Campus, Midlothian, United Kingdom
| | - Xin-Rui Xia
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Tobias Schwarz
- Royal (Dick) School of Veterinary Studies, Easter Bush Campus, The University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
584
|
Sarmanova A, Hall M, Fernandes GS, Bhattacharya A, Valdes AM, Walsh DA, Doherty M, Zhang W. Association between ultrasound-detected synovitis and knee pain: a population-based case-control study with both cross-sectional and follow-up data. Arthritis Res Ther 2017; 19:281. [PMID: 29258575 PMCID: PMC5738097 DOI: 10.1186/s13075-017-1486-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/24/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An important role for synovial pathology in the initiation and progression of knee osteoarthritis has been emphasised recently. This study aimed to examine whether ultrasonography-detected synovial changes associate with knee pain (KP) in a community population. METHODS A case-control study was conducted to compare people with early KP (n = 298), established KP (n = 100) or no KP (n = 94) at baseline. Multinomial logistic regression was used to estimate the odds ratio (OR) and 95% confidence interval (CI) between groups adjusted for radiographic osteoarthritis (ROA) severity and other confounding factors. After 1 year, 255 participants with early and established KP completed the follow-up questionnaire for changes in KP. Logistic regression with adjustment was used to determine predictors of KP worsening. RESULTS At baseline, effusion was associated with early KP (OR 2.64, 95% CI 1.57-4.45) and established KP (OR 5.07, 95% CI 2.74-9.38). Synovial hypertrophy was also associated with early KP (OR 5.43, 95% CI 2.12-13.92) and established KP (OR 13.27, 95% CI 4.97-35.43). The association with effusion diminished when adjusted for ROA. Power Doppler signal was uncommon (early KP 3%, established KP 2%, controls 0%). Baseline effusion predicted worsening of KP at 1 year (OR 1.95, 95% CI 1.05-3.64). However, after adjusting for ROA, the prediction was insignificant (adjusted OR 0.95, 95% CI 0.44-2.02). CONCLUSIONS Ultrasound effusion and synovial hypertrophy are associated with KP, but only effusion predicts KP worsening. However, the association/prediction is not independent from ROA. Power Doppler signal is uncommon in people with KP. Further study is needed to understand whether synovitis is directly involved in different types of KP.
Collapse
Affiliation(s)
- Aliya Sarmanova
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK.,Arthritis Research UK Pain Centre, Nottingham, UK
| | - Michelle Hall
- Arthritis Research UK Pain Centre, Nottingham, UK.,School of Health Sciences, University of Nottingham, Nottingham, UK
| | - Gwen S Fernandes
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK.,Arthritis Research UK Pain Centre, Nottingham, UK.,Arthritis Research UK Centre for Sports, Exercise and Osteoarthritis, Nottingham, UK
| | - Archan Bhattacharya
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK.,Arthritis Research UK Centre for Sports, Exercise and Osteoarthritis, Nottingham, UK.,School of Health Sciences, University of Nottingham, Nottingham, UK
| | - Ana M Valdes
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK.,Arthritis Research UK Pain Centre, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| | - David A Walsh
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK.,Arthritis Research UK Pain Centre, Nottingham, UK.,Arthritis Research UK Centre for Sports, Exercise and Osteoarthritis, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| | - Michael Doherty
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK.,Arthritis Research UK Pain Centre, Nottingham, UK.,Arthritis Research UK Centre for Sports, Exercise and Osteoarthritis, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| | - Weiya Zhang
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK. .,Arthritis Research UK Pain Centre, Nottingham, UK. .,Arthritis Research UK Centre for Sports, Exercise and Osteoarthritis, Nottingham, UK. .,NIHR Nottingham Biomedical Research Centre, Nottingham, UK.
| |
Collapse
|
585
|
Tang S, Deng S, Guo J, Chen X, Zhang W, Cui Y, Luo Y, Yan Z, He QY, Shen S, Wang T. Deep Coverage Tissue and Cellular Proteomics Revealed IL-1β Can Independently Induce the Secretion of TNF-Associated Proteins from Human Synoviocytes. THE JOURNAL OF IMMUNOLOGY 2017; 200:821-833. [DOI: 10.4049/jimmunol.1700480] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/31/2017] [Indexed: 01/15/2023]
|
586
|
Abstract
PURPOSE OF THE REVIEW Mounting evidence supports a role of low-grade inflammation in the pathophysiology of osteoarthritis (OA). We review and discuss the role of synovitis, complement activation, cytokines, and immune cell population in OA. RECENT FINDINGS Using newer imaging modalities, synovitis is found in the majority of knees with OA. Complement activation and pro-inflammatory cytokines play a significant role in the development of cartilage destruction and synovitis. Immune cell infiltration of OA synovial tissue by sub-populations of T cells and activated macrophages correlates with OA disease progression and pain. The innate and acquired immune system plays a key role in the low-grade inflammation found associated with OA. Targets of these pathways my hold promise for future disease-modifying osteoarthritis drugs (DMOADs).
Collapse
Affiliation(s)
| | - Adrian Filiberti
- Department of Medicine, University of Oklahoma Health Sciences Center, 975 N.E. 10th St, BRC 256, Oklahoma City, OK, 73104, USA
| | - Syed Ali Husain
- Department of Medicine, University of Oklahoma Health Sciences Center, 975 N.E. 10th St, BRC 256, Oklahoma City, OK, 73104, USA
| | - Mary Beth Humphrey
- Department of Medicine, University of Oklahoma Health Sciences Center, 975 N.E. 10th St, BRC 256, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs, Oklahoma City, OK, USA.
| |
Collapse
|
587
|
The serine proteinase hepsin is an activator of pro-matrix metalloproteinases: molecular mechanisms and implications for extracellular matrix turnover. Sci Rep 2017; 7:16693. [PMID: 29196708 PMCID: PMC5711915 DOI: 10.1038/s41598-017-17028-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/21/2017] [Indexed: 01/04/2023] Open
Abstract
Increasing evidence implicates serine proteinases in the proteolytic cascades leading to the pathological destruction of extracellular matrices such as cartilage in osteoarthritis (OA). We have previously demonstrated that the type II transmembrane serine proteinase (TTSP) matriptase acts as a novel initiator of cartilage destruction via the induction and activation of matrix metalloproteinases (MMPs). Hepsin is another TTSP expressed in OA cartilage such that we hypothesized this proteinase may also contribute to matrix turnover. Herein, we demonstrate that addition of hepsin to OA cartilage in explant culture induced significant collagen and aggrecan release and activated proMMP-1 and proMMP-3. Furthermore, hepsin directly cleaved the aggrecan core protein at a novel cleavage site within the interglobular domain. Hepsin expression correlated with synovitis as well as tumour necrosis factor α expression, and was induced in cartilage by a pro-inflammatory stimulus. However, a major difference compared to matriptase was that hepsin demonstrated markedly reduced capacity to activate proteinase-activated receptor-2. Overall, our data suggest that hepsin, like matriptase, induces potent destruction of the extracellular matrix whilst displaying distinct efficiencies for the cleavage of specific substrates.
Collapse
|
588
|
Qadri M, Almadani S, Jay GD, Elsaid KA. Role of CD44 in Regulating TLR2 Activation of Human Macrophages and Downstream Expression of Proinflammatory Cytokines. THE JOURNAL OF IMMUNOLOGY 2017; 200:758-767. [PMID: 29196459 DOI: 10.4049/jimmunol.1700713] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 11/05/2017] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is a low-grade chronic inflammatory joint disease. Innate immunity contributes to OA progression, mediated by TLR2 and TLR4. We evaluated the role of cluster determinant 44 (CD44), a transmembrane glycoprotein, in regulating TLR2-linked macrophage activation and resultant proinflammatory responses. TLR2 stimulation was performed on differentiated THP-1 macrophages in the presence or absence of a CD44-specific Ab or hyaluronan (HA). NF-κB nuclear translocation, IL-1 β and TNF-α gene expression, and protein concentrations were determined. Anti-CD44 Ab and HA treatments reduced NF-κB translocation, IL-1β and TNF-α expression, and production (p < 0.001). Inhibition of proinflammatory response in macrophages by HA was mediated by CD44. Protein phosphatase 2A mediated the reduction in NF-κB translocation by HA. CD44 knockdown reduced NF-κB nuclear translocation and downstream IL-1β and TNF-α protein production following TLR2 receptor stimulation (p < 0.001). CD44+/+ murine bone marrow-derived macrophages produced higher TNF-α compared with CD44-/- macrophages following TLR2 stimulation (p < 0.01). HA dose-dependently inhibited TLR2-induced TNF-α production by murine bone marrow-derived macrophages (p < 0.001). OA synovial fluids (SF) stimulated TLR2 and TLR4 receptors and induced NF-κB translocation in THP-1 macrophages. Anti-CD44 Ab treatment significantly reduced macrophage activation by OA SF (p < 0.01). CD44 regulated TLR2 responses in human macrophages, whereby a reduction in CD44 levels or engagement of CD44 by its ligand (HA) or a CD44-specific Ab reduced NF-κB translocation and downstream proinflammatory cytokine production. A CD44-specific Ab reduced macrophage activation by OA SF, and CD44 is a potentially novel target in OA treatment.
Collapse
Affiliation(s)
- Marwa Qadri
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618
| | - Sara Almadani
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115; and
| | - Gregory D Jay
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI 02903
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618;
| |
Collapse
|
589
|
Jeong H, Baek SY, Kim SW, Eun YH, Kim IY, Lee J, Jeon CH, Koh EM, Cha HS. Comorbidities and health-related quality of life in Koreans with knee osteoarthritis: Data from the Korean National Health and Nutrition Examination Survey (KNHANES). PLoS One 2017; 12:e0186141. [PMID: 29045425 PMCID: PMC5646822 DOI: 10.1371/journal.pone.0186141] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022] Open
Abstract
Objectives This study aimed to evaluate the association of knee osteoarthritis (OA) with comorbidities and health-related quality of life (HRQOL). Methods A total of 8,907 (weighted n = 13,687,058) participants aged ≥50 years who had undergone knee radiography were selected from the 2010–2012 Korea National Health and Nutrition Examination Survey. OA was classified into four subgroups based on the presence or absence of pain and radiographic OA (ROA): non-OA (Pain-/ROA-), pain only (Pain+/ROA-), ROA only (Pain-/ROA+), and painful ROA (Pain+/ROA+). ROA was defined as Kellgren–Lawrence grade ≥ 2. HRQOL measurements including EuroQOL visual analogue scale (EQ-VAS) scores and the five dimensions and summary index of the EuroQOL-5 dimension (EQ-5D index) were also analyzed. Multivariable logistic regression and linear regression analyses were performed. Results After adjustment for socioeconomic and lifestyle characteristics, cardiovascular disease, malignancy, and other comorbidities were not significantly associated with OA. Pain only and painful ROA were each significantly associated with limitations in physical activity (odds ratio (OR) 2.66, 95% CI 2.07–3.44, p < 0.001 and OR 2.83, 95% CI 2.25–3.58, p < 0.001, respectively), lower EQ-VAS (β-coefficient = -10.95, p < 0.001 and β-coefficient = -9.75, p < 0.001, respectively), and EQ-5D index (β-coefficient = -0.10, p < 0.001 and β-coefficient = -0.13, p < 0.001) compared with the non-OA group, whereas ROA only was not associated with limitations in physical activity or lower HRQOL score. Conclusions Comorbidities were not significantly associated with knee OA after adjustment. Knee OA was associated with physical activity and HRQOL. Painful knee OA, with or without ROA, was more strongly associated with decreased physical activity and lower quality of life than ROA without pain.
Collapse
Affiliation(s)
- Hyemin Jeong
- Division of Rheumatology, Department of Medicine, Soonchunhyang University Hospital, Bucheon, South Korea
| | - Sun Young Baek
- Biostatic and Clinical Epidemiology Center, Samsung Medical Center, Seoul, South Korea
| | - Seon Woo Kim
- Biostatic and Clinical Epidemiology Center, Samsung Medical Center, Seoul, South Korea
| | - Yeong Hee Eun
- Division of Rheumatology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - In Young Kim
- Division of Rheumatology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jaejoon Lee
- Division of Rheumatology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Chan Hong Jeon
- Division of Rheumatology, Department of Medicine, Soonchunhyang University Hospital, Bucheon, South Korea
| | - Eun-Mi Koh
- Division of Rheumatology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hoon-Suk Cha
- Division of Rheumatology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- * E-mail:
| |
Collapse
|
590
|
Shimura Y, Kurosawa H, Tsuchiya M, Sawa M, Kaneko H, Liu L, Makino Y, Nojiri H, Iwase Y, Kaneko K, Ishijima M. Serum interleukin 6 levels are associated with depressive state of the patients with knee osteoarthritis irrespective of disease severity. Clin Rheumatol 2017; 36:2781-2787. [PMID: 28900748 DOI: 10.1007/s10067-017-3826-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 08/26/2017] [Accepted: 09/04/2017] [Indexed: 01/15/2023]
Abstract
This cross-sectional study investigated the prevalence of depressive state and association between depressive state and serum interleukin (IL)-6 levels in knee osteoarthritis (OA) patients. A total of 115 painful knee OA patients were enrolled and divided into two groups according to the radiographic OA severity. Pain was evaluated using a visual analog scale (VAS). Depressive state was assessed by the self-rating depression scale (SDS). Serum IL-6 levels were also measured. Pearson's correlation coefficient was used to assess the correlation between the variants tested, and logistic regression analysis was used to identify factors associated with the depressive state. Fifty-two percent of the patients had an SDS score of ≥ 40, which is indicative of the depressive state. The pain VAS score (r = 0.22, p = 0.02) and serum IL-6 level (r = 0.31, p < 0.01) were independently associated with the SDS score of all early-stage knee OA patients (Kellgren-Lawrence [K/L] grade 2). However, only the serum IL-6 level was independently associated with the SDS scores of advanced-stage knee OA patients (K/L grades 3 and 4, r = 0.36, p < 0.01). A logistic regression analysis revealed that serum IL-6 level was the variable for the SDS score [odds ratio 1.41 (95% confidence interval 1.03-1.94, p < 0.03)]. Approximately half of the knee OA patients were found to be in the depressive state, and their serum IL-6 levels to be associated with the depressive state, irrespective of OA severity.
Collapse
Affiliation(s)
- Yukio Shimura
- Department of Orthopedics, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan.,Minamikoshigaya Hospital, Koshigaya, Saitama, Japan
| | - Hisashi Kurosawa
- Department of Orthopedics, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Masaru Tsuchiya
- Department of Orthopedics, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Mamiko Sawa
- Department of Orthopedics, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Haruka Kaneko
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Lizu Liu
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuji Makino
- Department of Orthopedics, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Hidetoshi Nojiri
- Department of Orthopedics, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Yoshiyuki Iwase
- Department of Orthopedics, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Kazuo Kaneko
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan. .,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
591
|
Gallo J, Raska M, Kriegova E, Goodman SB. Inflammation and its resolution and the musculoskeletal system. J Orthop Translat 2017; 10:52-67. [PMID: 28781962 PMCID: PMC5541893 DOI: 10.1016/j.jot.2017.05.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 02/08/2023] Open
Abstract
Inflammation, an essential tissue response to extrinsic/intrinsic damage, is a very dynamic process in terms of complexity and extension of cellular and metabolic involvement. The aim of the inflammatory response is to eliminate the pathogenic initiator with limited collateral damage of the inflamed tissue, followed by a complex tissue repair to the preinflammation phenotype. Persistent inflammation is a major contributor to the pathogenesis of many musculoskeletal diseases including ageing-related pathologies such as osteoporosis, osteoarthritis, and sarcopaenia. Understanding the mechanisms of inflammation and its resolution is therefore critical for the development of effective regenerative, and therapeutic strategies in orthopaedics.
Collapse
Affiliation(s)
- Jiri Gallo
- Department of Orthopaedics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, I.P. Pavlova 6, 779 00 Olomouc, Czech Republic
| | - Milan Raska
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 775 15 Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 775 15 Olomouc, Czech Republic
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Pavilion C, Redwood City, CA 94063-6342, USA
| |
Collapse
|