101
|
Vascular Endothelial Growth Factor Is Regulated by the Canonical and Noncanonical Transforming Growth Factor- β Pathway in Synovial Fibroblasts Derived from Osteoarthritis Patients. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6959056. [PMID: 31662989 PMCID: PMC6791277 DOI: 10.1155/2019/6959056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/27/2019] [Accepted: 09/06/2019] [Indexed: 01/28/2023]
Abstract
Background Previous studies suggest the presence of an association of vascular endothelial growth factor (VEGF) with osteoarthritis (OA) severity and pain in patients with knee OA. VEGF expression in human synovial fibroblasts (SFs) is induced by transforming growth factor-beta (TGFβ). However, the signaling pathway governing TGFβ-mediated regulation of VEGF in SFs has not been identified. Methods OA patients who underwent total knee arthroplasty had their synovial tissue (SYT) extracted and the constituent SFs cultured. The cells were stimulated with culture medium (control), human recombinant TGFβ (hrTGFβ), hrTGFβ + ALK5 inhibitor SB505124, hrTGFβ + transforming growth factor activating kinase 1 (TAK1) inhibitor (5Z)-7-oxozeaenol, or hrTGFβ + p38 inhibitor SB203580 for 6 h. VEGF mRNA expression in SFs was examined using real-time polymerase chain reaction and VEGF protein production in the cell supernatant was examined using enzyme-linked immunosorbent assay. Additionally, phosphorylated levels of SMAD2 and p38 were examined using western blotting. Results ALK5 (SB505124) and TAK1 (5Z-oxozeaenol) inhibitors completely suppressed TGFβ-induced VEGF mRNA expression and VEGF protein production. Both SB505124 and 5Z-oxozeaenol also suppressed SMAD2 and p38 phosphorylation. The p38 inhibitor (SB203580) partially inhibited TGFβ-mediated VEGF mRNA and VEGF protein production. Conclusion TGFβ-mediated regulation of VEGF expression and VEGF protein production in the SYT of OA patients occurs through both the canonical and noncanonical pathway.
Collapse
|
102
|
Zhang B, Chen H, Ouyang J, Xie Y, Chen L, Tan Q, Du X, Su N, Ni Z, Chen L. SQSTM1-dependent autophagic degradation of PKM2 inhibits the production of mature IL1B/IL-1β and contributes to LIPUS-mediated anti-inflammatory effect. Autophagy 2019; 16:1262-1278. [PMID: 31500508 DOI: 10.1080/15548627.2019.1664705] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Synovitis is implicated in the pathology of osteoarthritis (OA) and significantly contributes to the development of OA. As a noninvasive physical therapy, low-intensity pulsed ultrasound (LIPUS) has been reported to possess anti-inflammatory effect in recent years. However, the role of LIPUS on synovitis of OA and the underlying mechanisms are little known. The present study showed that LIPUS ameliorated synovial inflammation in destabilization of the medial meniscus (DMM) mouse model and air pouch model, and alleviated pain gait patterns of DMM mouse. LIPUS dramatically inhibited the production of mature IL1B/IL-1β (interleukin 1 beta) in vitro and in vivo. In addition, LIPUS upregulated the macroautophagy/autophagy level as well as accelerated the formation of an SQSTM1 (sequestosome1)-PKM (pyruvate kinase, muscle) complex in the lipopolysaccharide (LPS)-adenosine triphosphate (ATP)-treated macrophages. Besides, LIPUS downregulated the level of PKM2 in LPS-ATP-treated macrophages, which could be reversed by SQSTM1 knockdown. In brief, the present study for the first time demonstrates that LIPUS inhibits the production of mature IL1B partially via SQSTM1-dependent autophagic degradation of PKM2 in LPS-ATP-treated macrophages, which may further ameliorate the synovial inflammation and gait patterns in animal models. Our data provide new clues for the treatments of synovitis and other inflammatory diseases using LIPUS. ABBREVIATIONS 3-MA: 3-methyladenene; ATG7: autophagy-related 7; ATP: adenosine triphosphate; BafA1: bafilomycin A1; BMDMs: bone marrow derived macrophages; CHX: cycloheximide; DMM: destabilization of the medial meniscus; ELISA: enzyme-linked immunosorbent assay; GFP: green fluorescent protein; IL1B/IL-1β: interleukin 1 beta; LIPUS: low-intensity pulsed ultrasound; LIR: LC3-interacting region; LPS: lipopolysaccharide; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MDP: muramyl dipeptide; NFKB/NF-κB: nuclear factor kappa B; NLRP3: NLR family, pyrin domain containing 3; OA: osteoarthritis; PKM/PKM2: pyruvate kinase M1/2; PMA: phorbol-12-myristate-13-acetate; PYCARD/ASC; PYD and CARD domain containing; RFP: red fluorescent protein; siRNAs: small interfering RNAs; SQSTM1: sequestosome 1; TEM: transmission electron microscopy.
Collapse
Affiliation(s)
- Bin Zhang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Hangang Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Junjie Ouyang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Liang Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Qiaoyan Tan
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Nan Su
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Zhenhong Ni
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army medical University (Third Military Medical University) , Chongqing, China
| |
Collapse
|
103
|
Wang Q, Onuma K, Liu C, Wong H, Bloom MS, Elliott EE, Cao RR, Hu N, Lingampalli N, Sharpe O, Zhao X, Sohn DH, Lepus CM, Sokolove J, Mao R, Cisar CT, Raghu H, Chu CR, Giori NJ, Willingham SB, Prohaska SS, Cheng Z, Weissman IL, Robinson WH. Dysregulated integrin αVβ3 and CD47 signaling promotes joint inflammation, cartilage breakdown, and progression of osteoarthritis. JCI Insight 2019; 4:128616. [PMID: 31534047 PMCID: PMC6795293 DOI: 10.1172/jci.insight.128616] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the leading cause of joint failure, yet the underlying mechanisms remain elusive, and no approved therapies that slow progression exist. Dysregulated integrin function was previously implicated in OA pathogenesis. However, the roles of integrin αVβ3 and the integrin-associated receptor CD47 in OA remain largely unknown. Here, transcriptomic and proteomic analyses of human and murine osteoarthritic tissues revealed dysregulated expression of αVβ3, CD47, and their ligands. Using genetically deficient mice and pharmacologic inhibitors, we showed that αVβ3, CD47, and the downstream signaling molecules Fyn and FAK are crucial to OA pathogenesis. MicroPET/CT imaging of a mouse model showed elevated ligand-binding capacities of integrin αVβ3 and CD47 in osteoarthritic joints. Further, our in vitro studies demonstrated that chondrocyte breakdown products, derived from articular cartilage of individuals with OA, induced αVβ3/CD47-dependent expression of inflammatory and degradative mediators, and revealed the downstream signaling network. Our findings identify a central role for dysregulated αVβ3 and CD47 signaling in OA pathogenesis and suggest that activation of αVβ3 and CD47 signaling in many articular cell types contributes to inflammation and joint destruction in OA. Thus, the data presented here provide a rationale for targeting αVβ3, CD47, and their signaling pathways as a disease-modifying therapy.
Collapse
Affiliation(s)
- Qian Wang
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Kazuhiro Onuma
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Changhao Liu
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Heidi Wong
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Eileen E. Elliott
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Richard R.L. Cao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nick Hu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nithya Lingampalli
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Orr Sharpe
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Xiaoyan Zhao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Dong Hyun Sohn
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do, South Korea
| | - Christin M. Lepus
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Jeremy Sokolove
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Rong Mao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Cecilia T. Cisar
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Harini Raghu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Constance R. Chu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Nicholas J. Giori
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Stephen B. Willingham
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Susan S. Prohaska
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
104
|
Zhao L, Xiao E, He L, Duan D, He Y, Chen S, Zhang Y, Gan Y. Reducing macrophage numbers alleviates temporomandibular joint ankylosis. Cell Tissue Res 2019; 379:521-536. [PMID: 31522279 DOI: 10.1007/s00441-019-03087-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/05/2019] [Indexed: 01/08/2023]
Abstract
Temporomandibular joint (TMJ) ankylosis is a severe joint disease mainly caused by trauma that leads to a series of oral and maxillofacial function disorders and psychological problems. Our series of studies indicate that TMJ ankylosis development is similar to fracture healing and that severe trauma results in bony ankylosis instead of fibrous ankylosis. Macrophages are early infiltrating inflammatory cells in fracture and play a critical role in initiating fracture repair. We hypothesize that the large numbers of macrophages in the early phase of TMJ ankylosis trigger ankylosed bone mass formation and that the depletion of these macrophages in the early phase could inhibit the development of TMJ ankylosis. By analysing human TMJ ankylosis specimens, we found large numbers of infiltrated macrophages in the less-than-1-year ankylosis samples. A rabbit model of TMJ bony ankylosis was established and large numbers of infiltrated macrophages were found at 4 days post-operation. Local clodronate liposome (CLD-lip) injection, which depleted macrophages, alleviated the severity of ankylosis compared with local phosphate-buffered saline (PBS)-loaded liposome (PBS-lip) injection (macrophage number, PBS-lips vs. CLD-lips: 626.03 ± 164.53 vs. 341.4 ± 108.88 n/mm2; ankylosis calcification score, PBS-lips vs. CLD-lips: 2.11 ± 0.78 vs. 0.78 ± 0.66). Histological results showed that the cartilage area was reduced in the CLD-lip-treated side (PBS-lips vs. CLD-lips: 6.82 ± 4.42% vs. 2.71 ± 2.78%) and that the Wnt signalling regulating cartilage formation was disrupted (Wnt5a expression decreased 60% and Wnt4 expression decreased 73%). Thus, our study showed that large numbers of macrophages infiltrated during the early phase of ankylosis and that reducing macrophage numbers alleviated ankylosis development by reducing cartilage formation.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.,Laboratory of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China
| | - E Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.,Laboratory of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China
| | - Linhai He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.,Peking University Hospital of Stomatology First Clinical Division, 37A Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Denghui Duan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.,Laboratory of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.,Laboratory of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China
| | - Shuo Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.,Laboratory of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China. .,Laboratory of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| | - Yehua Gan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China. .,Central Laboratory, Peking University School and Hospital of Stomatology, #22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| |
Collapse
|
105
|
Ni Z, Kuang L, Chen H, Xie Y, Zhang B, Ouyang J, Wu J, Zhou S, Chen L, Su N, Tan Q, Luo X, Chen B, Chen S, Yin L, Huang H, Du X, Chen L. The exosome-like vesicles from osteoarthritic chondrocyte enhanced mature IL-1β production of macrophages and aggravated synovitis in osteoarthritis. Cell Death Dis 2019; 10:522. [PMID: 31285423 PMCID: PMC6614358 DOI: 10.1038/s41419-019-1739-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023]
Abstract
Synovitis, a common clinical symptom for osteoarthritis (OA) patients, is highly related to OA pathological progression and pain manifestation. The activated synovial macrophages have been demonstrated to play an important role in synovitis, but the mechanisms about macrophage activation are still not clear. In this study, we found that the exosome-like vesicles from osteoarthritic chondrocytes could be a new biological factor to stimulate inflammasome activation and increase mature IL-1β production in macrophages. The degraded cartilage explants produced more exosome-like vesicles than the nondegraded ones, while the exosome-like vesicles from chondrocytes could enter into joint synovium tissue and macrophages. Moreover, the exosome-like vesicles from osteoarthritic chondrocytes enhanced the production of mature IL-1β in macrophages. These vesicles could inhibit ATG4B expression via miR-449a-5p, leading to inhibition of autophagy in LPS-primed macrophages. The decreased autophagy promoted the production of mitoROS, which further enhanced the inflammasome activation and subsequent IL-1β processing. Ultimately, the increase of mature IL-1β may aggravate synovial inflammation and promote the progression of OA disease. Our study provides a new perspective to understand the activation of synovial macrophages and synovitis in OA patients, which may be beneficial for therapeutic intervention in synovitis-related OA patients.
Collapse
Affiliation(s)
- Zhenhong Ni
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Liang Kuang
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Hangang Chen
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Yangli Xie
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Bin Zhang
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Junjie Ouyang
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Jiangyi Wu
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Siru Zhou
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Liang Chen
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Nan Su
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - QiaoYan Tan
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Xiaoqing Luo
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Bo Chen
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Shuai Chen
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Liangjun Yin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, 400010, Chongqing, China
| | - Haiyang Huang
- Department of Orthopedic Surgery, Qianjiang Nationality Hospital, 409000, Chongqing, China
| | - Xiaolan Du
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Lin Chen
- Laboratory for the Rehabilitation of Traumatic Injuries, Laboratory of Trauma, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Laboratory for Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China.
| |
Collapse
|
106
|
Haraden CA, Huebner JL, Hsueh MF, Li YJ, Kraus VB. Synovial fluid biomarkers associated with osteoarthritis severity reflect macrophage and neutrophil related inflammation. Arthritis Res Ther 2019; 21:146. [PMID: 31196179 PMCID: PMC6567574 DOI: 10.1186/s13075-019-1923-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/22/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND To identify a synovial fluid (SF) biomarker profile characteristic of individuals with an inflammatory osteoarthritis (OA) endotype. METHODS A total of 48 knees (of 25 participants) were characterized for an extensive array of SF biomarkers quantified by Rules Based Medicine using the high-sensitivity multiplex immunoassay, Myriad Human InflammationMAP® 1.0, which included 47 different cytokines, chemokines, and growth factors related to inflammation. Multivariable regression with generalized estimating equations (GEE) and false discovery rate (FDR) correction was used to assess associations of SF RBM biomarkers with etarfolatide imaging scores reflecting synovial inflammation; radiographic knee OA severity (based on Kellgren-Lawrence (KL) grade, joint space narrowing, and osteophyte scores); knee joint symptoms; and SF biomarkers associated with activated macrophages and knee OA progression including CD14 and CD163 (shed by activated macrophages) and elastase (shed by activated neutrophils). RESULTS Significant associations of SF biomarkers meeting FDR < 0.05 included soluble (s)VCAM-1 and MMP-3 with synovial inflammation (FDR-adjusted p = 0.025 and 1.06 × 10-7); sVCAM-1, sICAM-1, TIMP-1, and VEGF with radiographic OA severity (p = 1.85 × 10-5 to 3.97 × 10-4); and VEGF, MMP-3, TIMP-1, sICAM-1, sVCAM-1, and MCP-1 with OA symptoms (p = 2.72 × 10-5 to 0.050). All these SF biomarkers were highly correlated with macrophage markers CD163 and CD14 in SF (r = 0.43 to 0.90, FDR < 0.05); all but MCP-1 were also highly correlated with neutrophil elastase in SF (r = 0.62 to 0.89, FDR < 0.05). CONCLUSIONS A subset of six SF biomarkers was related to synovial inflammation in OA, as well as radiographic and symptom severity. These six OA-related SF biomarkers were specifically linked to indicators of activated macrophages and neutrophils. These results attest to an inflammatory OA endotype that may serve as the basis for therapeutic targeting of a subset of individuals at high risk for knee OA progression. TRIAL REGISTRATION Written informed consent was received from participants prior to inclusion in the study; the study was registered at ClinicalTrials.gov ( NCT01237405 ) on November 9, 2010, prior to enrollment of the first participant.
Collapse
Affiliation(s)
- Collin A Haraden
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA
| | - Janet L Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA
| | - Ming-Feng Hsueh
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA
| | - Yi-Ju Li
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA.,Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Box 104775, 300 North Duke St, Durham, NC, 27701, USA. .,Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
107
|
Yarnall BW, Chamberlain CS, Hao Z, Muir P. Proinflammatory polarization of stifle synovial macrophages in dogs with cruciate ligament rupture. Vet Surg 2019; 48:1005-1012. [PMID: 31190376 DOI: 10.1111/vsu.13261] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/19/2019] [Accepted: 05/21/2019] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To determine polarization of synovial macrophages during development of cruciate ligament rupture (CR) and determine whether differences in synovial macrophage polarization in CR, osteoarthritis (OA), and healthy joints exist. STUDY DESIGN Prospective case-controlled study. ANIMALS Client-owned dogs with unstable stifles with CR (n = 22), paired stable contralateral stifles with partial CR (pCR; n = 7), joints with OA not related to CR (n = 6), and clinically normal (Normal; n = 7) joints. METHODS Synovial fluid samples were collected. Smears were made for differential cytology counts and estimated total nucleated cell counts. Cytospin preparations were made, and immunocytochemical staining was performed with the pan-macrophage marker CD68, M1 macrophage markers inducible nitric oxide synthase (iNOS) and chemokine (C-C motif) receptor 7 (CCR7), and M2 macrophage markers arginase 1 and CD163. Positively stained cells were counted. RESULTS Numbers of lymphocytes were increased in the CR group compared with the OA and Normal groups (P < .05). Numbers of CD68+ , CCR7+ , and iNOS+ cells in the CR and OA groups were increased compared with the Normal group (P < .05). Globally, the ratio of positively stained M1 polarized CD68+ cells to M2 polarized CD68+ cells was highest for the OA group (2.49), followed by the pCR (2.1), CR (1.63), and Normal (0.7) groups. CONCLUSION Polarization of synovial macrophages toward an M1 proinflammatory phenotype is an early event in the development of canine CR. CLINICAL SIGNIFICANCE M1 polarization in pCR stifles provides evidence of a possible role for macrophages in progressive development of cruciate ligament fiber damage. Lymphocytes may play a role in the synovitis found in CR joints. Our findings provide evidence that these cells are therapeutic targets.
Collapse
Affiliation(s)
- Benjamin W Yarnall
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Connie S Chamberlain
- Department of Orthopedics & Rehabilitation, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Zhengling Hao
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Peter Muir
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
108
|
Teng P, Liu Y, Dai Y, Zhang H, Liu WT, Hu J. Nicotine Attenuates Osteoarthritis Pain and Matrix Metalloproteinase-9 Expression via the α7 Nicotinic Acetylcholine Receptor. THE JOURNAL OF IMMUNOLOGY 2019; 203:485-492. [PMID: 31152077 DOI: 10.4049/jimmunol.1801513] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/08/2019] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that causes chronic disability among the elderly. Despite recent advances in symptomatic management of OA by pharmacological and surgical approaches, there remains a lack of optimal approaches to manage inflammation in the joints, which causes cartilage degradation and pain. In this study, we investigated the efficacy and underlying mechanisms of nicotine exposure in attenuating joint inflammation, cartilage degradation, and pain in a mouse model of OA. A mouse model of OA was induced by injection of monosodium iodoacetate into the knee joint. Cell culture models were also used to study the efficacy and underlying mechanisms of nicotine treatment in attenuating symptoms of OA. Nicotine treatment reduced mechanical allodynia, cartilage degradation, and the upregulation of matrix metalloproteinase-9 (MMP-9), a hallmark of joint inflammation in OA, in mice treated with monosodium iodoacetate. The effects of nicotine were abolished by the selective α7 nicotinic acetylcholine receptor (nAChR) blocker, methyllycaconitine . In RAW264.7 cells and murine primary bone marrow-derived macrophages, nicotine significantly inhibited MMP-9 production induced by LPS. In addition, nicotine significantly enhanced PI3K/Akt and inhibited NF-κB translocation from the cytosol to the nucleus in an α7-nAChR-dependent manner, suggesting that nicotine acts on α7-nAChRs to inhibit MMP-9 production by macrophages through modulation of the PI3K/Akt-NF-κB pathway. Our results provide novel evidence that nicotine can attenuate joint inflammation and pain in experimental OA via α7-nAChRs. α7-nAChR could thus serve as a highly promising target to manage joint inflammation and pain in OA.
Collapse
Affiliation(s)
- Peng Teng
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - Yuan Liu
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; and
| | - Yan Dai
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; and
| | - Haijun Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China; .,Department of Pharmacy, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Jun Hu
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China;
| |
Collapse
|
109
|
van Dalen SCM, Blom AB, Walgreen B, Slöetjes AW, Helsen MMA, Geven EJW, Ter Huurne M, Vogl T, Roth J, van de Loo FAJ, Koenders MI, Casteilla L, van der Kraan PM, van den Bosch MHJ, van Lent PLEM. IL-1β-Mediated Activation of Adipose-Derived Mesenchymal Stromal Cells Results in PMN Reallocation and Enhanced Phagocytosis: A Possible Mechanism for the Reduction of Osteoarthritis Pathology. Front Immunol 2019; 10:1075. [PMID: 31191517 PMCID: PMC6545928 DOI: 10.3389/fimmu.2019.01075] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/26/2019] [Indexed: 01/15/2023] Open
Abstract
Background: Injection of adipose-derived mesenchymal stromal cells (ASCs) into murine knee joints after induction of inflammatory collagenase-induced osteoarthritis (CiOA) reduces development of joint pathology. This protection is only achieved when ASCs are applied in early CiOA, which is characterized by synovitis and high S100A8/A9 and IL-1β levels, suggesting that inflammation is a prerequisite for the protective effect of ASCs. Our objective was to gain more insight into the interplay between synovitis and ASC-mediated amelioration of CiOA pathology. Methods: CiOA was induced by intra-articular collagenase injection. Knee joint sections were stained with hematoxylin/eosin and immunolocalization of polymorphonuclear cells (PMNs) and ASCs was performed using antibodies for NIMP-R14 and CD271, respectively. Chemokine expression induced by IL-1β or S100A8/A9 was assessed with qPCR and Luminex. ASC-PMN co-cultures were analyzed microscopically and with Luminex for inflammatory mediators. Migration of PMNs through transwell membranes toward conditioned medium of non-stimulated ASCs (ASCNS-CM) or IL-1β-stimulated ASCs (ASCIL-1β-CM) was examined using flow cytometry. Phagocytic capacity of PMNs was measured with labeled zymosan particles. Results: Intra-articular saline injection on day 7 of CiOA increased synovitis after 6 h, characterized by PMNs scattered throughout the joint cavity and the synovium. ASC injection resulted in comparable numbers of PMNs which clustered around ASCs in close interaction with the synovial lining. IL-1β-stimulation of ASCs in vitro strongly increased expression of PMN-attracting chemokines CXCL5, CXCL7, and KC, whereas S100A8/A9-stimulation did not. In agreement, the number of clustered PMNs per ASC was significantly increased after 6 h of co-culturing with IL-1β-stimulated ASCs. Also migration of PMNs toward ASCIL-1β-CM was significantly enhanced (287%) when compared to ASCNS-CM. Interestingly, association of PMNs with ASCs significantly diminished KC protein release by ASCs (69% lower after 24 h), accompanied by reduced release of S100A8/A9 protein by the PMNs. Moreover, phagocytic capacity of PMNs was strongly enhanced after priming with ASCIL-1β-CM. Conclusions: Local application of ASCs in inflamed CiOA knee joints results in clustering of attracted PMNs with ASCs in the synovium, which is likely mediated by IL-1β-induced up-regulation of chemokine release by ASCs. This results in enhanced phagocytic capacity of PMNs, enabling the clearance of debris to attenuate synovitis.
Collapse
Affiliation(s)
- Stephanie C M van Dalen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Birgitte Walgreen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Annet W Slöetjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Monique M A Helsen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Edwin J W Geven
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Menno Ter Huurne
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Fons A J van de Loo
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marije I Koenders
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Peter M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Martijn H J van den Bosch
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
110
|
Wang Q, Lepus CM, Raghu H, Reber LL, Tsai MM, Wong HH, von Kaeppler E, Lingampalli N, Bloom MS, Hu N, Elliott EE, Oliviero F, Punzi L, Giori NJ, Goodman SB, Chu CR, Sokolove J, Fukuoka Y, Schwartz LB, Galli SJ, Robinson WH. IgE-mediated mast cell activation promotes inflammation and cartilage destruction in osteoarthritis. eLife 2019; 8:39905. [PMID: 31084709 PMCID: PMC6516833 DOI: 10.7554/elife.39905] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 04/10/2019] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis is characterized by articular cartilage breakdown, and emerging evidence suggests that dysregulated innate immunity is likely involved. Here, we performed proteomic, transcriptomic, and electron microscopic analyses to demonstrate that mast cells are aberrantly activated in human and murine osteoarthritic joint tissues. Using genetic models of mast cell deficiency, we demonstrate that lack of mast cells attenuates osteoarthritis in mice. Using genetic and pharmacologic approaches, we show that the IgE/FcεRI/Syk signaling axis is critical for the development of osteoarthritis. We find that mast cell-derived tryptase induces inflammation, chondrocyte apoptosis, and cartilage breakdown. Our findings demonstrate a central role for IgE-dependent mast cell activation in the pathogenesis of osteoarthritis, suggesting that targeting mast cells could provide therapeutic benefit in human osteoarthritis. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Qian Wang
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Christin M Lepus
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Harini Raghu
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
| | - Mindy M Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
| | - Heidi H Wong
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Ericka von Kaeppler
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Nithya Lingampalli
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Michelle S Bloom
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Nick Hu
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Eileen E Elliott
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Francesca Oliviero
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Leonardo Punzi
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Nicholas J Giori
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, United States
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, United States
| | - Constance R Chu
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, United States
| | - Jeremy Sokolove
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Yoshihiro Fukuoka
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
| | - Lawrence B Schwartz
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, United States.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, United States
| | - William H Robinson
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
111
|
Haltmayer E, Ribitsch I, Gabner S, Rosser J, Gueltekin S, Peham J, Giese U, Dolezal M, Egerbacher M, Jenner F. Co-culture of osteochondral explants and synovial membrane as in vitro model for osteoarthritis. PLoS One 2019; 14:e0214709. [PMID: 30939166 PMCID: PMC6445514 DOI: 10.1371/journal.pone.0214709] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 03/16/2019] [Indexed: 01/15/2023] Open
Abstract
The purpose of the current study was to establish an in vitro model for osteoarthritis (OA) by co-culture of osteochondral and synovial membrane explants. Osteochondral explants were cultured alone (control-1) or in co-culture with synovial membrane explants (control-2) in standard culture medium or with interleukin-1β (IL1β) and tumor necrosis factor (TNFα) added to the culture medium (OA-model-1 = osteochondral explant; OA-model-2 = osteochondroal-synovial explant). In addition, in OA-model groups a 2-mm partial-thickness defect was created in the centre of the cartilage explant. Changes in the expression of extracellular matrix (ECM) genes (collagen type-1 (Col1), Col2, Col10 and aggrecan) as well as presence and quantity of inflammatory marker genes (IL6, matrix metalloproteinase-1 (MMP1), MMP3, MMP13, a disintegrin and metalloproteinase with-thrombospondin-motif-5 (ADAMTS5) were analysed by immunohistochemistry, qPCR and ELISA. To monitor the activity of classically-activated pro-inflammatory (M1) versus alternatively-activated anti-inflammatory/repair (M2) synovial macrophages, the nitric oxide/urea ratio in the supernatant of osteochondral-synovial explant co-cultures was determined. In both OA-model groups immunohistochemistry and qPCR showed a significantly increased expression of MMPs and IL6 compared to their respective control group. ELISA results confirmed a statistically significant increase in MMP1and MMP3 production over the culturing period. In the osteochondral-synovial explant co-culture OA-model the nitric oxide/urea ratio was increased compared to the control group, indicating a shift toward M1 synovial macrophages. In summary, chemical damage (TNFα, IL1β) in combination with a partial-thickness cartilage defect elicits an inflammatory response similar to naturally occurring OA in osteochondral explants with and without osteochondral-synovial explant co-cultures and OA-model-2 showing a closer approximation of OA due to the additional shift of synovial macrophages toward the pro-inflammatory M1 phenotype.
Collapse
Affiliation(s)
- Eva Haltmayer
- Department for Companion Animals and Horses, University Equine Hospital, Equine Surgery, University of Veterinary Medicine, Vienna, Austria
- * E-mail:
| | - Iris Ribitsch
- Department for Companion Animals and Horses, University Equine Hospital, Equine Surgery, University of Veterinary Medicine, Vienna, Austria
| | - Simone Gabner
- Department of Pathobiology, Histology and Embryology, University of Veterinary Medicine, Vienna, Austria
| | - Julie Rosser
- Institute of Applied Synthetic Chemistry, Technical University, Vienna, Austria
| | - Sinan Gueltekin
- Department for Companion Animals and Horses, University Equine Hospital, Equine Surgery, University of Veterinary Medicine, Vienna, Austria
| | - Johannes Peham
- Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology, Vienna, Austria
| | - Ulrich Giese
- Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology, Vienna, Austria
| | - Marlies Dolezal
- Department of Biomedical Sciences, Bioinformatics and Biostatistics Platform, University of Veterinary Medicine, Vienna, Austria
| | - Monika Egerbacher
- Department of Pathobiology, Histology and Embryology, University of Veterinary Medicine, Vienna, Austria
| | - Florien Jenner
- Department for Companion Animals and Horses, University Equine Hospital, Equine Surgery, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
112
|
Jaswal AP, Bandyopadhyay A. Re-examining osteoarthritis therapy from a developmental biologist's perspective. Biochem Pharmacol 2019; 165:17-23. [PMID: 30922620 DOI: 10.1016/j.bcp.2019.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/13/2019] [Indexed: 01/25/2023]
Abstract
Osteoarthritis is the most prevalent musculoskeletal disorder and one for which there is no disease modifying therapy available at present. Our current understanding of the disease mechanism of osteoarthritis is limited owing to a lacuna of knowledge about the development and maintenance of articular cartilage that is affected during osteoarthritis. All current therapeutic strategies aim at countering inflammation which though mitigates pain but does not arrest the progressive degeneration of articular cartilage. During osteoarthritis, articular cartilage expresses markers for transient cartilage differentiation. Moreover, blocking transient cartilage differentiation is sufficient for halting the progression of experimental osteoarthritis. A developmental biology inspired approach that combines restoration of tissue microenvironment, supplementation with engineered cartilage and built in mechanism to prevent transient cartilage differentiation could be an avenue for developing a disease modifying therapy for osteoarthritis.
Collapse
Affiliation(s)
- Akrit Pran Jaswal
- Lab 10, Department of Biological Sciences and Bio-engineering, IIT, Kanpur, India.
| | | |
Collapse
|
113
|
Xie J, Huang Z, Yu X, Zhou L, Pei F. Clinical implications of macrophage dysfunction in the development of osteoarthritis of the knee. Cytokine Growth Factor Rev 2019; 46:36-44. [PMID: 30910350 DOI: 10.1016/j.cytogfr.2019.03.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritic disease, leading to disability and impaired quality of life and no curative treatments exist. Increasing evidence indicates that low-grade inflammation plays a pivotal role in the onset and progression of OA. In this review, we summarize emerging findings on the pathological roles of synovial macrophages, adipose tissue macrophages, and osteoclasts in OA and their potential clinical implications from cell biology to preclinical and preliminary clinical trials. The failure of synovial macrophages to transition from pro-inflammatory M1 to anti-inflammatory M2 subtypes may contribute to the initiation and maintenance of synovitis in OA. M1 macrophages promote the inflammatory microenvironment and progression of OA through interactions with synovial fibroblasts and chondrocytes, thus increasing the secretion of matrix metalloproteinases. Direct inhibition of M1 or promotion of M2 polarization may be useful therapeutic interventions. Adipose tissue macrophages present in the infrapatella fat pad (IPFP) were involved in the progression of obesity-induced OA, which contributed to changes in the integrity of the IPFP. Furthermore, macrophages and osteoclasts in the subchondral bone were involved in bone remodeling and pain through uncoupled osteoclast/osteoblast activity and increased nociceptive signaling. Growing evidence has indicated an important role for macrophage-mediated low-grade inflammation in OA. Fully understanding the link between macrophages and other cells in joints will provide new insights into OA disease modification.
Collapse
Affiliation(s)
- Jinwei Xie
- Department of Orthopaedics Surgery, Laboratory of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Zeyu Huang
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Li Zhou
- Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Fuxing Pei
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
114
|
Xie J, Huang Z, Pei F. [Role and progress of innate immunity in pathogenesis of osteoarthritis]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:370-376. [PMID: 30874397 PMCID: PMC8337921 DOI: 10.7507/1002-1892.201810068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/09/2019] [Indexed: 02/05/2023]
Abstract
Objective To review and summarize the role and progress of innate immunity in the pathogenesis of osteoarthritis (OA). Methods The domestic and foreign literature in recent years was reviewed. The role of innate immune-mediated inflammation, macrophages, T cells, and complement systems in the pathogenesis of OA, potential therapeutic targets, and the latest research progress were summarized. Results With the deepening of research, OA is gradually considered as a low-grade inflammation, in which innate immunity plays an important role. The polarization of synovial macrophage subpopulation in OA has been studied extensively. Current data shows that the failure of transformation from M1 subtype to M2 subtype is a key link in the progression of OA. T cells and complement system are also involved in the pathological process of OA. Conclusion At present, the role of innate immunity in the progress of OA has been played in the spotlight, whereas the specific mechanism has not been clear. The macrophage subtype polarization is a potential therapeutic target for early prevention and treatment of OA.
Collapse
Affiliation(s)
- Jinwei Xie
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Zeyu Huang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Fuxing Pei
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041,
| |
Collapse
|
115
|
Ross CL, Ang DC, Almeida-Porada G. Targeting Mesenchymal Stromal Cells/Pericytes (MSCs) With Pulsed Electromagnetic Field (PEMF) Has the Potential to Treat Rheumatoid Arthritis. Front Immunol 2019; 10:266. [PMID: 30886614 PMCID: PMC6409305 DOI: 10.3389/fimmu.2019.00266] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/31/2019] [Indexed: 01/14/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of synovium (synovitis), with inflammatory/immune cells and resident fibroblast-like synoviocytes (FLS) acting as major players in the pathogenesis of this disease. The resulting inflammatory response poses considerable risks as loss of bone and cartilage progresses, destroying the joint surface, causing joint damage, joint failure, articular dysfunction, and pre-mature death if left untreated. At the cellular level, early changes in RA synovium include inflammatory cell infiltration, synovial hyperplasia, and stimulation of angiogenesis to the site of injury. Different angiogenic factors promote this disease, making the role of anti-angiogenic therapy a focus of RA treatment. To control angiogenesis, mesenchymal stromal cells/pericytes (MSCs) in synovial tissue play a vital role in tissue repair. While recent evidence reports that MSCs found in joint tissues can differentiate to repair damaged tissue, this repair function can be repressed by the inflammatory milieu. Extremely-low frequency pulsed electromagnetic field (PEMF), a biophysical form of stimulation, has an anti-inflammatory effect by causing differentiation of MSCs. PEMF has also been reported to increase the functional activity of MSCs to improve differentiation to chondrocytes and osteocytes. Moreover, PEMF has been demonstrated to accelerate cell differentiation, increase deposition of collagen, and potentially return vascular dysfunction back to homeostasis. The aim of this report is to review the effects of PEMF on MSC modulation of cytokines, growth factors, and angiogenesis, and describe its effect on MSC regeneration of synovial tissue to further understand its potential role in the treatment of RA.
Collapse
Affiliation(s)
- Christina L Ross
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States.,Wake Forest Center for Integrative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Dennis C Ang
- Department of Rheumatology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| |
Collapse
|
116
|
Janssens LAA, Verheijen IKA, Serangeli J, van Kolfschoten T. Shoulder osteoarthritis in a European saber-toothed cat (Homotherium latidens) from the Lower Palaeolithic site of Schöningen (Germany). INTERNATIONAL JOURNAL OF PALEOPATHOLOGY 2019; 24:279-285. [PMID: 30777196 DOI: 10.1016/j.ijpp.2018.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/06/2018] [Accepted: 06/09/2018] [Indexed: 06/09/2023]
Abstract
Evaluation of a right ventral scapula fragment from a mature Homotherium latidens from Schöningen, Germany (337-300 ka before present - MIS 9) revealed lesions consisting of an osteophyte at the caudal border of the glenoid cavity, and a large, multilobular, cystic feature in the medio-caudal glenoid cavity. Based on the type of lesions, their localization, their severity, and exclusion of several nutritional and other etiologies such as immune mediated disease, joint infection (septic arthritis), and joint tumors, we conclude that the lesion was caused by trauma or age-related shoulder osteoarthritis (or possibly both). We cannot speculate whether the condition was symptomatic, but if it was, the animal must have functioned well enough to hunt or scavenge, since it survived a significant period of lesion development.
Collapse
Affiliation(s)
- Luc A A Janssens
- Faculty of Archaeology, Leiden University, Einsteinweg 2, 2333 CC Leiden, The Netherlands; Evidensia Clinic for Referral Surgery of Companion Animals, Eerste Zeine 112, 5144AM, Waalwijk, The Netherlands; Faculty of Archaeology, Ghent University, Sint-Pietersnieuwstraat, 9000 Ghent, Belgium.
| | - Ivo K A Verheijen
- Faculty of Archaeology, Leiden University, Einsteinweg 2, 2333 CC Leiden, The Netherlands; Universität Tübingen/Senckenberg HEP, paläon 1, 38364 Schöningen, Germany
| | - Jordi Serangeli
- Universität Tübingen/Senckenberg HEP, paläon 1, 38364 Schöningen, Germany
| | - Thijs van Kolfschoten
- Faculty of Archaeology, Leiden University, Einsteinweg 2, 2333 CC Leiden, The Netherlands
| |
Collapse
|
117
|
Luo Q, Ji S, Li Z, Huang T, Fan S, Xi Q. Effects of ultrasound therapy on the synovial fluid proteome in a rabbit surgery-induced model of knee osteoarthritis. Biomed Eng Online 2019; 18:18. [PMID: 30795769 PMCID: PMC6387552 DOI: 10.1186/s12938-019-0637-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 02/15/2019] [Indexed: 01/07/2023] Open
Abstract
Background Ultrasound (US) therapy may improve osteoarthritis symptoms. We investigated the effects of US on the synovial fluid (SF) proteome in a rabbit knee osteoarthritis (KOA) model to explore its therapeutic mechanisms. Methods Sixteen healthy 6-month-old New Zealand white rabbits (eight male, eight female), weighing 2.5–3.0 kg, were randomly divided into groups A and B with eight rabbits per group. Both groups were subjected to right anterior cruciate ligament transaction. Six weeks after surgery, we treated the operated knee joint of group A rabbits with US and of group B rabbits with sham US for 2 weeks. The proteomes of knee joint SF from groups A and B rabbits were then analyzed using a label-free mass spectrometry (MS) quantification method. Results We identified 19 protein sequences annotated by 361 Gene Ontology (GO) items. According to the Kyoto Encyclopedia of Genes and Genomes (KEGG) database of rabbit protein sequences, we then annotated the KO numbers of homologous/similar proteins to 32 relevant KEGG pathways. We extracted 10 significantly differentially expressed proteins among the 32 relevant KEGG messages/metabolism pathways. The proteins whose levels were decreased were apolipoprotein A-I (AopA-1), transferrin (TF), carboxypeptidase B2 (CBP2), arylesterase/paraoxonase (PON), fibrinogen alpha chain, and alpha-2-macroglobulin (A2M). The proteins whose levels were increased were molecular chaperone HtpG/heat shock proteins (htpG, HSP90A), decorin (DCN), pyruvate kinase (PK, pyk), and fatty acid-binding protein 4/adipocyte (FABP4, aP2). Conclusions US therapy can alter protein levels in SF, which can decrease AopA-1, TF, CBP2, PON, fibrinogen alpha chain and A2M protein levels, and increase HtpG/HSP90A, DCN, PK/PKY, and FABP4/aP2 protein levels in SF of KOA, suggesting that the therapeutic mechanisms of US therapy on KOA may occur through changes in the SF proteome.
Collapse
Affiliation(s)
- Qinglu Luo
- The Fifth Affiliated Hospital of Guangzhou Medicine University, No. 621, GangWan Road, HuangPu District, Guangzhou, 510700, Guangdong Province, China.,College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Shuangquan Ji
- The Fifth Affiliated Hospital of Guangzhou Medicine University, No. 621, GangWan Road, HuangPu District, Guangzhou, 510700, Guangdong Province, China
| | - Zhimi Li
- The Fifth Affiliated Hospital of Guangzhou Medicine University, No. 621, GangWan Road, HuangPu District, Guangzhou, 510700, Guangdong Province, China
| | - Tao Huang
- The Fifth Affiliated Hospital of Guangzhou Medicine University, No. 621, GangWan Road, HuangPu District, Guangzhou, 510700, Guangdong Province, China
| | - Siqin Fan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Qin Xi
- The Fifth Affiliated Hospital of Guangzhou Medicine University, No. 621, GangWan Road, HuangPu District, Guangzhou, 510700, Guangdong Province, China.
| |
Collapse
|
118
|
Wood MJ, Leckenby A, Reynolds G, Spiering R, Pratt AG, Rankin KS, Isaacs JD, Haniffa MA, Milling S, Hilkens CM. Macrophage proliferation distinguishes 2 subgroups of knee osteoarthritis patients. JCI Insight 2019; 4:125325. [PMID: 30674730 DOI: 10.1172/jci.insight.125325] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a leading cause of disability, globally. Despite an emerging role for synovial inflammation in OA pathogenesis, attempts to target inflammation therapeutically have had limited success. A better understanding of the cellular and molecular processes occurring in the OA synovium is needed to develop novel therapeutics. We investigated macrophage phenotype and gene expression in synovial tissue of OA and inflammatory-arthritis (IA) patients. Compared with IA, OA synovial tissue contained higher but variable proportions of macrophages (P < 0.001). These macrophages exhibited an activated phenotype, expressing folate receptor-2 and CD86, and displayed high phagocytic capacity. RNA sequencing of synovial macrophages revealed 2 OA subgroups. Inflammatory-like OA (iOA) macrophages are closely aligned to IA macrophages and are characterized by a cell proliferation signature. In contrast, classical OA (cOA) macrophages display cartilage remodeling features. Supporting these findings, when compared with cOA, iOA synovial tissue contained higher proportions of macrophages (P < 0.01), expressing higher levels of the proliferation marker Ki67 (P < 0.01). These data provide new insight into the heterogeneity of OA synovial tissue and suggest distinct roles of macrophages in pathogenesis. Our findings could lead to the stratification of OA patients for suitable disease-modifying treatments and the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Matthew J Wood
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom
| | - Adam Leckenby
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom
| | - Gary Reynolds
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom.,NIHR Newcastle Biomedical Research Centre and
| | - Rachel Spiering
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom
| | - Arthur G Pratt
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom.,NIHR Newcastle Biomedical Research Centre and
| | - Kenneth S Rankin
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,NIHR Newcastle Biomedical Research Centre and
| | - John D Isaacs
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom.,NIHR Newcastle Biomedical Research Centre and
| | - Muzlifah A Haniffa
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,NIHR Newcastle Biomedical Research Centre and.,Department of Dermatology, Newcastle Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon Milling
- Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom.,Institute of Infection, Immunity and Inflammation, Glasgow University, United Kingdom
| | - Catharien Mu Hilkens
- Institute of Cellular Medicine, Newcastle University, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence, Glasgow, Birmingham, Newcastle, United Kingdom.,NIHR Newcastle Biomedical Research Centre and
| |
Collapse
|
119
|
Kozijn AE, Tartjiono MT, Ravipati S, van der Ham F, Barrett DA, Mastbergen SC, Korthagen NM, Lafeber FPJG, Zuurmond AM, Bobeldijk I, Weinans H, Stoop R. Human C-reactive protein aggravates osteoarthritis development in mice on a high-fat diet. Osteoarthritis Cartilage 2019; 27:118-128. [PMID: 30248505 DOI: 10.1016/j.joca.2018.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/17/2018] [Accepted: 09/13/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE C-reactive protein (CRP) levels can be elevated in osteoarthritis (OA) patients. In addition to indicating systemic inflammation, it is suggested that CRP itself can play a role in OA development. Obesity and metabolic syndrome are important risk factors for OA and also induce elevated CRP levels. Here we evaluated in a human CRP (hCRP)-transgenic mouse model whether CRP itself contributes to the development of 'metabolic' OA. DESIGN Metabolic OA was induced by feeding 12-week-old hCRP-transgenic males (hCRP-tg, n = 30) and wild-type littermates (n = 15) a 45 kcal% high-fat diet (HFD) for 38 weeks. Cartilage degradation, osteophytes and synovitis were graded on Safranin O-stained histological knee joint sections. Inflammatory status was assessed by plasma lipid profiling, flow cytometric analyses of blood immune cell populations and immunohistochemical staining of synovial macrophage subsets. RESULTS Male hCRP-tg mice showed aggravated OA severity and increased osteophytosis compared with their wild-type littermates. Both classical and non-classical monocytes showed increased expression of CCR2 and CD86 in hCRP-tg males. HFD-induced effects were evident for nearly all lipids measured and indicated a similar low-grade systemic inflammation for both genotypes. Synovitis scores and synovial macrophage subsets were similar in the two groups. CONCLUSIONS Human CRP expression in a background of HFD-induced metabolic dysfunction resulted in the aggravation of OA through increased cartilage degeneration and osteophytosis. Increased recruitment of classical and non-classical monocytes might be a mechanism of action through which CRP is involved in aggravating this process. These findings suggest interventions selectively directed against CRP activity could ameliorate metabolic OA development.
Collapse
Affiliation(s)
- A E Kozijn
- Metabolic Health Research, TNO, Leiden, the Netherlands; Department of Orthopaedics, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - M T Tartjiono
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - S Ravipati
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - F van der Ham
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - D A Barrett
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - S C Mastbergen
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - N M Korthagen
- Department of Orthopaedics, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - F P J G Lafeber
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - A M Zuurmond
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - I Bobeldijk
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - H Weinans
- Department of Orthopaedics, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| | - R Stoop
- Metabolic Health Research, TNO, Leiden, the Netherlands.
| |
Collapse
|
120
|
Abstract
The increase in global lifespan has in turn increased the prevalence of osteoarthritis which is now the most common type of arthritis. Cartilage tissue located on articular joints erodes during osteoarthritis which causes pain and may lead to a crippling loss of function in patients. The pathophysiology of osteoarthritis has been understudied and currently no disease modifying treatments exist. The only current end-point treatment remains joint replacement surgery. The primary risk factor for osteoarthritis is age. Clinical and basic research is now focused on understanding the ageing process of cartilage and its role in osteoarthritis. This chapter will outline the physiology of cartilage tissue, the clinical presentation and treatment options for the disease and the cellular ageing processes which are involved in the pathophysiology of the disease.
Collapse
|
121
|
van Dalen SCM, Kruisbergen NNL, Walgreen B, Helsen MMA, Slöetjes AW, Cremers NAJ, Koenders MI, van de Loo FAJ, Roth J, Vogl T, Blom AB, van der Kraan PM, van Lent PLEM, van den Bosch MHJ. The role of NOX2-derived reactive oxygen species in collagenase-induced osteoarthritis. Osteoarthritis Cartilage 2018; 26:1722-1732. [PMID: 30195046 DOI: 10.1016/j.joca.2018.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Synovitis in collagenase-induced osteoarthritis (CiOA) is driven by locally released S100A8/A9 proteins and enhances joint destruction. S100A8/A9 can induce reactive oxygen species (ROS) release by phagocytes in OA synovium via neutrophil cytosolic factor-1 (Ncf1)-regulated NOX2 activation. In the present study we investigated whether NOX2-derived ROS affect joint pathology during CiOA. METHODS CiOA was induced in knee joints of wild type (WT) and Ncf1-deficient (Ncf1**) mice. Synovial gene expression of NOX2-subunits was measured with quantitative real-time polymerase chain reaction (qRT-PCR). Joint pathology was assessed using histology and immunohistochemistry for aggrecan neo-epitope VDIPEN. Levels of inflammatory proteins were measured with Luminex or ELISA. Phagocytes in synovium, blood, bone marrow (BM) and spleen were analyzed with flow cytometry. ROS release by phagocytes was measured with a ROS detection kit. RESULTS CiOA induction in knee joints of WT mice caused significantly increased synovial gene expression of NOX2 subunits. On day 7 of CiOA, cartilage damage and MMP activity, as measured by VDIPEN, were comparable between WT and Ncf1** mice. Synovial thickening, synovial S100A8/A9 levels and percentages of synovial macrophages, polymorphonuclear cells (PMNs), and monocytes were not different, as were levels of inflammatory mediators in serum and phagocyte percentages in blood, BM and spleen. On day 42 of CiOA, synovitis, cartilage damage, and osteophyte formation in Ncf1** mice were unaltered when compared to WT mice. ROS detection confirmed that Ncf1** PMNs lack functional NOX2, but in vitro macrophages showed ROS production, suggesting activation of compensatory mechanisms. CONCLUSIONS Absence of Ncf1-mediated ROS production does not alter joint pathology in CiOA.
Collapse
Affiliation(s)
- S C M van Dalen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - N N L Kruisbergen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - B Walgreen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - M M A Helsen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - A W Slöetjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - N A J Cremers
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - M I Koenders
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - F A J van de Loo
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - J Roth
- Institute of Immunology, University of Münster, Germany.
| | - T Vogl
- Institute of Immunology, University of Münster, Germany.
| | - A B Blom
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - P M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - P L E M van Lent
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - M H J van den Bosch
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
122
|
Sambamurthy N, Zhou C, Nguyen V, Smalley R, Hankenson KD, Dodge GR, Scanzello CR. Deficiency of the pattern-recognition receptor CD14 protects against joint pathology and functional decline in a murine model of osteoarthritis. PLoS One 2018; 13:e0206217. [PMID: 30485272 PMCID: PMC6261538 DOI: 10.1371/journal.pone.0206217] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 10/09/2018] [Indexed: 12/18/2022] Open
Abstract
Objective CD14 is a monocyte/macrophage pattern-recognition receptor that modulates innate inflammatory signaling. Soluble CD14 levels in knee OA synovial fluids are associated with symptoms and progression of disease. Here we investigate the role of this receptor in development of OA using a murine joint injury model of disease. Methods 10-week-old Male C57BL/6 (WT) and CD14-deficient (CD14-/-) mice underwent destabilization of the medial meniscus (DMM) surgery to induce OA. Joint histopathology was used to examine cartilage damage, and microCT to evaluate subchondral bone (SCB) remodeling at 6 and 19 weeks after surgery. Synovial and fat pad expression of macrophage markers (F4/80, CD11c, CD68, iNOS, CCR7, CD163 and CD206) was assessed by flow cytometry and droplet digital (dd)PCR. Changes in locomotive activity indicative of joint pain were evaluated longitudinally up to 16 weeks by automated behavioral analysis. Results Early cartilage damage scores 6 weeks post-DMM were similar in both strains (Mean score ±SEM WT: 4.667±1.38, CD14-/-: 4.6±0.6), but at 19 weeks were less severe in CD14-/- (6.0±0.46) than in WT mice (13.44±2.5, p = 0.0002). CD14-/- mice were protected from both age-related and post-surgical changes in SCB mineral density and trabecular thickness. In addition, CD14-/- mice were protected from decreases in climbing activity (p = 0.015 vs. WT, 8 weeks) observed after DMM. Changes in synovial/fat pad expression of CCR7, a marker of M1 macrophages, were slightly reduced post-DMM in the absence of CD14, while expression of CD68 (pan-macrophage marker) and CD163 (M2 marker) were unchanged. Conclusion CD14 plays an important role in progression of structural and functional features of OA in the DMM model, and may provide a new target for therapeutic development.
Collapse
Affiliation(s)
- Nisha Sambamurthy
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Division of Rheumatology, Philadelphia, Pennsylvania, United States of America
| | - Cheng Zhou
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Division of Rheumatology, Philadelphia, Pennsylvania, United States of America
| | - Vu Nguyen
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Division of Rheumatology, Philadelphia, Pennsylvania, United States of America
| | - Ryan Smalley
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Department of Orthopedic Surgery, Philadelphia, Pennsylvania, United States of America
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - George R. Dodge
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Department of Orthopedic Surgery, Philadelphia, Pennsylvania, United States of America
| | - Carla R. Scanzello
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania, United States of America
- University of Pennsylvania Perelman School of Medicine, Division of Rheumatology, Philadelphia, Pennsylvania, United States of America
- * E-mail: ,
| |
Collapse
|
123
|
van den Bosch MHJ. Inflammation in osteoarthritis: is it time to dampen the alarm(in) in this debilitating disease? Clin Exp Immunol 2018; 195:153-166. [PMID: 30421798 DOI: 10.1111/cei.13237] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2018] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease that strongly reduces the quality of life in patients; However, no disease-modifying therapy is available. For a long time, OA was considered a non-inflammatory disease that was the result of 'wear-and-tear' and abnormal mechanics, and therefore many considered the term 'osteoarthritis' a misnomer. However, during the last decades the notion arose that inflammation is not only present in the majority of OA patients but, rather, actively involved in the progression of the disease. Influx of immune cells is observed in the synovium and a plethora of inflammatory mediators is present in tissues and fluids from OA patients. These mediators cause the production of degrading enzymes that break down the cartilage matrix, which is the main hallmark of OA. Alarmins, which belong to the group of danger signals, have been implicated in many inflammatory diseases. They are among the first factors to be released upon cell stress due to, for example, infection, damage and inflammation. They attract and activate cells of the immune system and therefore lie at the base of the inflammatory reaction. In this narrative review, an overview of the history of OA, the evolving concept of inflammation as important factor in the OA pathogenesis, and particularly the central role that alarmins play in the initiation and maintenance of the low-grade inflammatory response in OA, is provided. Moreover, the targeting of alarmins as a promising approach to dampen the inflammation in OA is highlighted.
Collapse
Affiliation(s)
- M H J van den Bosch
- Experimental Rheumatology, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
124
|
MRI-detected osteophytes of the knee: natural history and structural correlates of change. Arthritis Res Ther 2018; 20:237. [PMID: 30352619 PMCID: PMC6235223 DOI: 10.1186/s13075-018-1734-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 09/25/2018] [Indexed: 01/08/2023] Open
Abstract
Backgroud The natural history of semi-quantitative magnetic resonance imaging (MRI)-detected osteophytes (MRI-detected OPs) has not been described and it is unknown whether knee structural abnormalities can predict MRI-detected OP change over time. Thus, the aim of current study is to describe the natural history of knee MRI-detected OP, and to determine if knee structural abnormalities are associated with change of MRI-detected OP in a longitudinal study of older adults. Methods Randomly selected older adults (n = 837, mean age 63 years) had MRI at baseline and 413 of them had MRI 2.6 years later to measure MRI-detected OP, cartilage defects, cartilage volume, bone marrow lesions (BMLs), meniscal extrusion, infrapatellar fat pad (IPFP) quality score/maximum area and effusion-synovitis. Results Over 2.6 years, average MRI-detected OP score increased significantly in all compartments. The total MRI-detected OP score remained stable in 53% of participants, worsened (≥ 1-point increase) in 46% and decreased in 1%. Baseline cartilage defects (RR, 1.25–1.35), BMLs (RR, 1.16–1.17), meniscal extrusion (RR, 1.22–1.33) and IPFP quality score (RR, 1.08–1.20) site-specifically and independently predicted an increase in MRI-detected OP (p values all ≤ 0.05), after adjustment for covariates. Presence of IPFP abnormality was significantly associated with increased MRI-detected OPs but became non-significant after adjustment for other structural abnormalities. Total (RR, 1.27) and suprapatellar pouch effusion-synovitis (RR, 1.22) were both associated with increased MRI-detected OPs in the lateral compartment only (both p < 0.04). Conclusion Knee MRI-detected OPs are common in older adults and are likely to progress. The association between baseline structural abnormalities and worsening MRI-detected OPs suggest MRI-detected OP could be a consequence of multiple knee structural abnormalities.
Collapse
|
125
|
Ashraf S, Mapp PI, Shahtaheri SM, Walsh DA. Effects of carrageenan induced synovitis on joint damage and pain in a rat model of knee osteoarthritis. Osteoarthritis Cartilage 2018; 26:1369-1378. [PMID: 30031926 DOI: 10.1016/j.joca.2018.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Knee osteoarthritis (OA) is associated with ongoing pain and joint damage that can be punctuated by acute flares of pain and inflammation. Synovitis in normal knees might resolve without long-term detriment to joint function. We hypothesised that osteoarthritis is associated with impaired resilience to inflammatory flares. DESIGN We induced synovitis by injecting carrageenan into rat knees with or without meniscal transection (MNX)-induced OA, and measured synovitis, weightbearing asymmetry (pain behaviour), and joint damage up to 35 days after OA induction (23 days after carrageenan-injection). RESULTS Carrageenan injection induced weightbearing asymmetry for 1 week, transient increase in knee diameter for 2 days, and a sustained increase in synovial macrophages, endothelial cell proliferation and vascular density compared with naive vehicle-injected controls. MNX surgery induced weightbearing asymmetry and histological evidence of OA. Carrageenan-injection in MNX-operated knees was followed for 2 days by increased weightbearing asymmetry compared either to MNX+vehicle or to sham+carrageenan groups. OA structural damage and synovitis at day 35 were greater in MNX+carrageenan compared to MNX+vehicle and sham+carrageenan groups. Carrageenan injection did not induce OA in Sham-operated knees. CONCLUSION Intra-articular injection of the pro-inflammatory compound carrageenan in OA and sham-operated control knees induced a short term increase in joint pain. Even though pain flares resolved in both groups and damage was not induced in sham-operated knees, carrageen injection exacerbated long-term joint damage in OA knees. OA knees display less resilience to inflammatory episodes. Preventing inflammatory flares may be particularly important in preventing symptoms and long term joint damage in OA.
Collapse
Affiliation(s)
- S Ashraf
- School of Pharmacy, University of Nottingham, Nottingham, UK; Arthritis Research UK Pain Centre and NIHR Nottingham BRC, University of Nottingham, Nottingham, UK.
| | - P I Mapp
- Arthritis Research UK Pain Centre and NIHR Nottingham BRC, University of Nottingham, Nottingham, UK.
| | - S M Shahtaheri
- Arthritis Research UK Pain Centre and NIHR Nottingham BRC, University of Nottingham, Nottingham, UK.
| | - D A Walsh
- Arthritis Research UK Pain Centre and NIHR Nottingham BRC, University of Nottingham, Nottingham, UK.
| |
Collapse
|
126
|
Li S, Cong W, Hakamivala A, Huang Y, Borrelli J, Tang L. Hyaluronic Acid-Based Optical Probe for the Diagnosis of Human Osteoarthritic Cartilage. Nanotheranostics 2018; 2:347-359. [PMID: 30148052 PMCID: PMC6107780 DOI: 10.7150/ntno.26119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/15/2018] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis is typically caused by cartilage injury, followed by localized inflammatory responses and tissue deterioration. Early treatment of osteoarthritis is often impossible due to the lack of diagnostic options. Recent studies have supported that different imaging probes can be used for arthritis detection in mice. However, none of these diagnostic tools have been tested on human articular cartilage. To fill this gap, an optical imaging probe was developed to target activated macrophages and the accumulation of imaging probes on tissue was used to assess the severity of human osteoarthritis. Methods: The probe was fabricated using hyaluronic acid (HA) particles conjugated with near-infrared dye and folic acid (FA). The ability of the FA-HA probes to detect activated macrophages and quantify cartilage injury was evaluated using a cell culture model in vitro and human osteoarthritic cartilage explants ex vivo. Results: Our cell study results supported that the FA-HA probes are cell compatible (up to 0.5mg/mL) and can detect activated macrophages in 30 minutes. Using human articular cartilage, we verified the existence of activated macrophages on osteoarthritic cartilage with highly up-regulated expression of folate receptors (~13 folds by comparison with healthy control). In addition, we found that FA-HA probes had higher binding amounts (~3 folds) to osteoarthritic tissue than healthy ones. Histological analyses confirmed that there was a strong linear relationship (R=0.933) between the fluorescent intensity of tissue-associated probe and the extent of folate receptors on osteoarthritic cartilage. Finally, the co-localization of the imaging probe, folate receptors and cartilage degeneration on the tissue sections indicated the extraordinary accuracy and efficiency of this osteoarthritis diagnostic probe. Conclusions: Our results support the probe as an effective diagnostic tool to detect the area and severity of osteoarthritic human articular cartilage.
Collapse
Affiliation(s)
- Shuxin Li
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Wei Cong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA.,Department of Oral Anatomy, College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Amirhossein Hakamivala
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - YiHui Huang
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Joseph Borrelli
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Liping Tang
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
127
|
Zhang H, Lin C, Zeng C, Wang Z, Wang H, Lu J, Liu X, Shao Y, Zhao C, Pan J, Xu S, Zhang Y, Xie D, Cai D, Bai X. Synovial macrophage M1 polarisation exacerbates experimental osteoarthritis partially through R-spondin-2. Ann Rheum Dis 2018; 77:1524-1534. [DOI: 10.1136/annrheumdis-2018-213450] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/12/2018] [Accepted: 06/17/2018] [Indexed: 12/14/2022]
Abstract
ObjectivesTo investigate the roles and regulatory mechanisms of synovial macrophages and their polarisation in the development of osteoarthritis (OA).MethodsSynovial tissues from normal patients and patients with OA were collected. M1 or M2-polarised macrophages in synovial tissues of patients with OA and OA mice were analysed by immunofluorescence and immunohistochemical staining. Mice with tuberous sclerosis complex 1 (TSC1) or Rheb deletion specifically in the myeloid lineage were generated and subjected to intra-articular injection of collagenase (collagenase-induced osteoarthritis, CIOA) and destabilisation of the medial meniscus (DMM) surgery to induce OA. Cartilage damage and osteophyte size were measured by Osteoarthritis Research Society International score and micro-CT, respectively. mRNA sequencing was performed in M1 and control macrophages. Mice and ATDC5 cells were treated with R-spondin-2 (Rspo2) or anti-Rspo2 to investigate the role of Rspo2 in OA.ResultsM1 but not M2-polarised macrophages accumulated in human and mouse OA synovial tissue. TSC1 deletion in the myeloid lineage constitutively activated mechanistic target of rapamycin complex 1 (mTORC1), increased M1 polarisation in synovial macrophages and exacerbated experimental OA in both CIOA and DMM models, while Rheb deletion inhibited mTORC1, enhanced M2 polarisation and alleviated CIOA in mice. The results show that promoting the macrophage M1 polarisation leads to exacerbation of experimental OA partially through secretion of Rspo2 and activation of β-catenin signalling in chondrocytes.ConclusionsSynovial macrophage M1 polarisation exacerbates experimental CIOA partially through Rspo2. M1 macrophages and Rspo2 are potential therapeutic targets for OA treatment.
Collapse
|
128
|
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
|
129
|
Jeon OH, David N, Campisi J, Elisseeff JH. Senescent cells and osteoarthritis: a painful connection. J Clin Invest 2018; 128:1229-1237. [PMID: 29608139 PMCID: PMC5873863 DOI: 10.1172/jci95147] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Senescent cells (SnCs) are associated with age-related pathologies. Osteoarthritis is a chronic disease characterized by pain, loss of cartilage, and joint inflammation, and its incidence increases with age. For years, the presence of SnCs in cartilage isolated from patients undergoing total knee artificial implants has been noted, but these cells' relevance to disease was unclear. In this Review, we summarize current knowledge of SnCs in the multiple tissues that constitute the articular joint. New evidence for the causative role of SnCs in the development of posttraumatic and age-related arthritis is reviewed along with the therapeutic benefit of SnC clearance. As part of their senescence-associated secretory phenotype, SnCs secrete cytokines that impact the immune system and its response to joint tissue trauma. We present concepts of the immune response to tissue trauma as well as the interactions with SnCs and the local tissue environment. Finally, we discuss therapeutic implications of targeting SnCs in treating osteoarthritis.
Collapse
Affiliation(s)
- Ok Hee Jeon
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
130
|
de Visser HM, Korthagen NM, Müller C, Ramakers RM, Krijger GC, Lafeber FPJG, Beekman FJ, Mastbergen SC, Weinans H. Imaging of Folate Receptor Expressing Macrophages in the Rat Groove Model of Osteoarthritis: Using a New DOTA-Folate Conjugate. Cartilage 2018; 9:183-191. [PMID: 29096521 PMCID: PMC5871123 DOI: 10.1177/1947603517738073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objective To evaluate the presence and localization of folate receptor expressing macrophages in the rat groove model of osteoarthritis and determine the suitability of a new folate conjugate with albumin-binding entity (cm09) for in vivo SPECT (single-photon emission computed tomography) analysis. Design In male Wistar rats, local cartilage damage was induced in addition to a standard ( n = 10) or high-fat diet ( n = 6). After 12 weeks, 111In labeled folate conjugates were administered, and SPECT/CT (computed tomography) imaging was performed after 24 hours. Subsequently, osteoarthritis severity and folate receptor expression were assessed using (immuno)-histological sections. Results In vivo SPECT/CT imaging of the new folate conjugate (cm09) was as useful as a folate conjugate without albumin-binding entity in the groove model of osteoarthritis with less renal accumulation. Induction of cartilage damage on a standard diet resulted in no effect on the amount of folate receptor expressing macrophages compared with the contralateral sham operated joints. In contrast, inducing cartilage damage in the high-fat diet group resulted in 28.4% increase of folate receptor expression as compared with the nondamaged control joints. Folate receptor expressing cells were predominantly present in the synovial lining and in subchondral bone as confirmed by immunohistochemistry. Conclusions Folate receptor expression, and thus macrophage activation, can clearly be demonstrated in vivo, in small animal models of osteoarthritis using the new 111In-folate conjugate with specific binding to the folate receptor. Increased macrophage activity only plays a role in the groove model of osteoarthritis when applied in a high-fat diet induced dysmetabolic condition, which is in line with the higher inflammatory state of that specific model.
Collapse
Affiliation(s)
- Huub M. de Visser
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nicoline M. Korthagen
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Cristina Müller
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Ruud M. Ramakers
- MILabs B.V., Utrecht, The Netherlands,Section of Radiation, Detection & Medical Imaging, Applied Sciences, Delft University of Technology, Delft, The Netherlands,Department for Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard C. Krijger
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Floris P. J. G. Lafeber
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Freek J. Beekman
- MILabs B.V., Utrecht, The Netherlands,Section of Radiation, Detection & Medical Imaging, Applied Sciences, Delft University of Technology, Delft, The Netherlands,Department for Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Simon C. Mastbergen
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands,Simon C. Mastbergen, Department of Rheumatology & Clinical Immunology, UMC Utrecht, F.02.127, PO Box 85500, 3508 GA Utrecht, The Netherlands.
| | - Harrie Weinans
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
131
|
Morgan RE, Clegg PD, Hunt JA, Innes JF, Tew SR. Interaction with macrophages attenuates equine fibroblast-like synoviocyte ADAMTS5 (aggrecanase-2) gene expression following inflammatory stimulation. J Orthop Res 2018; 36:2178-2185. [PMID: 29521434 PMCID: PMC6120467 DOI: 10.1002/jor.23891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 03/02/2018] [Indexed: 02/04/2023]
Abstract
The joint synovium consists of a heterogeneous cell population, chiefly comprised of macrophages, and fibroblast-like synoviocytes (FLS). An inter-species co-culture model was developed to examine interactions between these cells. Equine FLS and the canine macrophage line DH82 were differentially labeled using fluorescent markers and results from direct co-culture compared with those from both indirect co-culture, and conditioned media experiments. The transcript expression of IL-1β, IL-6, ADAMTS4, and ADAMTS5 in each cell type were determined using species-specific qPCR assays. Lipopolysaccharide stimulation of EFLS rapidly increased IL-1β, IL-6, ADAMTS4, and ADAMTS5 mRNAs. The induction of ADAMTS5 was significantly reduced when equine FLS were cultured with DH82 cells directly or indirectly. Exposure of equine FLS to denatured conditioned media also significantly reduced ADAMTS5 induction. DH82 cells increased interleukin-1β expression substantially following LPS stimulation. However, knockdown of interleukin-1β in DH82 cells, or inhibition of NF-κB in equine FLS prior to co-culture did not change the inhibitory effect on equine FLS ADAMTS5 gene expression. This work indicates that macrophages can influence FLS gene expression through a soluble mediator, and modulate the expression of an enzyme critical in osteoarthritis pathology during inflammatory stimulation. © 2018 The Authors. Journal of Orthopaedic Research® Published by WileyPeriodicals, Inc. on behalf of the Orthopaedic Research Society. J Orthop Res 9999:1-8, 2018.
Collapse
Affiliation(s)
- Rhiannon E. Morgan
- The Royal Veterinary CollegeEquine Referral HospitalHawkshead LaneHatfieldAL9 7TA
| | - Peter D. Clegg
- Department of Musculoskeletal BiologyInstitute of Ageing and Chronic DiseaseThe University of LiverpoolWilliam Henry Duncan BuildingWest Derby StreetLiverpoolMerseysideL7 8TX
| | - John A. Hunt
- Department of Musculoskeletal BiologyInstitute of Ageing and Chronic DiseaseThe University of LiverpoolWilliam Henry Duncan BuildingWest Derby StreetLiverpoolMerseysideL7 8TX
| | - John F. Innes
- Institute of Veterinary SciencesThe University of Liverpool, LeahurstChester High RoadNestonCheshireCH64 7TE
| | - Simon R. Tew
- Department of Musculoskeletal BiologyInstitute of Ageing and Chronic DiseaseThe University of LiverpoolWilliam Henry Duncan BuildingWest Derby StreetLiverpoolMerseysideL7 8TX
| |
Collapse
|
132
|
de Visser HM, Mastbergen SC, Kozijn AE, Coeleveld K, Pouran B, van Rijen MH, Lafeber FPJG, Weinans H. Metabolic dysregulation accelerates injury-induced joint degeneration, driven by local inflammation; an in vivo rat study. J Orthop Res 2018; 36:881-890. [PMID: 28840952 DOI: 10.1002/jor.23712] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/19/2017] [Indexed: 02/04/2023]
Abstract
Evidence is growing for the existence of an obesity-related phenotype of osteoarthritis in which low-grade inflammation and a disturbed metabolic profile play a role. The contribution of an obesity-induced metabolic dysbalance to the progression of the features of osteoarthritis upon mechanically induced cartilage damage was studied in a rat in vivo model. Forty Wistar rats were randomly allocated 1:1 to a standard diet or a high-fat diet. After 12 weeks, in 14 out of 20 rats in each group, cartilage was mechanically damaged in the right knee joint. The remaining six animals in each group served as controls. After a subsequent 12 weeks, serum was collected for metabolic state, subchondral bone changes assessed by μCT imaging, osteoarthritis severity determined by histology, and macrophage presence assessed by CD68 staining. The high-fat diet increased statistically all relevant metabolic parameters, resulting in a dysmetabolic state and subsequent synovial inflammation, whereas cartilage degeneration was hardly influenced. The high-fat condition in combination with mechanical cartilage damage resulted in a clear statistically significant progression of the osteoarthritic features, with increased synovitis and multiple large osteophytes. Both the synovium and osteophytes contained numerous CD68 positive cells. It is concluded that a metabolic dysbalance due to a high-fat diet increases joint inflammation without cartilage degeneration. The dysmetabolic state clearly accelerates progression of osteoarthritis upon surgically induced cartilage damage supported by inflammatory responses as demonstrated by histology and increased CD68 expressing cells localized on the synovial membrane and osteophytes. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:881-890, 2018.
Collapse
Affiliation(s)
- Huub M de Visser
- Department of Orthopaedics, University Medical Center Utrecht, F.02.127, 3508 GA, Utrecht, 85500, The Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Simon C Mastbergen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anne E Kozijn
- Department of Orthopaedics, University Medical Center Utrecht, F.02.127, 3508 GA, Utrecht, 85500, The Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Metabolic Health Research, TNO, Leiden, The Netherlands
| | - Katja Coeleveld
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Behdad Pouran
- Department of Orthopaedics, University Medical Center Utrecht, F.02.127, 3508 GA, Utrecht, 85500, The Netherlands.,Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| | - Mattie H van Rijen
- Department of Orthopaedics, University Medical Center Utrecht, F.02.127, 3508 GA, Utrecht, 85500, The Netherlands
| | - Floris P J G Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopaedics, University Medical Center Utrecht, F.02.127, 3508 GA, Utrecht, 85500, The Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
133
|
The Clinical Significance of Osteophytes in Compartments of the Knee Joint With Normal Articular Cartilage. AJR Am J Roentgenol 2018; 210:W164-W171. [PMID: 29470158 DOI: 10.2214/ajr.17.18664] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The purpose of this study is to determine whether marginal osteophytes in compartments with normal cartilage would be more frequently observed in knees with cartilage lesions and osteophytes in other compartments. MATERIALS AND METHODS This retrospective study reviewed 500 consecutive knee MRI examinations performed within 6 months of arthroscopic knee surgery conducted for 497 patients with symptoms (289 male patients and 208 female patients; age range, 17-74 years; median age, 43 years). The highest grade of cartilage lesion detected at MRI and arthroscopy was recorded. Marginal osteophytes were graded on MRI with use of a standardized scoring system, with grade 0 denoting no osteophyte; grade 1, small osteophyte; grade 2, medium-size osteophyte; and grade 3, large osteophyte). The frequency of false-positive osteophytes, defined as osteophytes present in compartments (the patellofemoral, medial tibiofemoral, and lateral tibiofemoral compartments) with normal cartilage observed on MRI and arthroscopy, was calculated. The Goodman and Kruskal gamma statistic was used to test the association of osteophyte size between compartments. Logistic regression was used to test the association between osteophyte size and the severity of the cartilage lesions. RESULTS Marginal osteophytes were seen in compartments with normal cartilage on MRI and arthroscopy in 60.5% of knees (75 of 124) with cartilage lesions and osteophytes in other compartments and accounted for all false-positive grade 2 and grade 3 osteophytes. Marginal osteophytes were seen in 12.7% of knees (13 of 102) that had no cartilage lesions in any compartment on MRI or arthroscopy, and all of these were grade 1 osteophytes. The presence of larger sized osteophytes in the compartments with cartilage lesions was associated with the presence of larger sized osteophytes in the compartments with normal cartilage. More severe cartilage lesions were associated with larger osteophyte size. CONCLUSION Compartments with marginal osteophytes and normal cartilage are commonly seen in knees that have other compartments with osteophytes and cartilage lesions.
Collapse
|
134
|
Loukov D, Karampatos S, Maly MR, Bowdish DME. Monocyte activation is elevated in women with knee-osteoarthritis and associated with inflammation, BMI and pain. Osteoarthritis Cartilage 2018; 26:255-263. [PMID: 29128509 DOI: 10.1016/j.joca.2017.10.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/11/2017] [Accepted: 10/31/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Monocytes contribute to synovitis and disease pathogenesis in osteoarthritis (OA). Low-grade inflammation occurs in OA and correlates with disease severity and progression. Since monocyte development and function is altered by systemic inflammation, we analyzed monocyte numbers and function between individuals with knee OA and healthy age- and sex-matched controls. DESIGN We analyzed markers of soluble and cellular inflammation in peripheral blood of women with knee OA and compared them to healthy age- and sex-matched controls. Soluble inflammatory mediators (TNF, IL-6, IL-10 and CRP) in the serum were measured by high-sensitivity ELISA. Leukocyte numbers, surface expression of monocyte activation markers, and monocyte production of pro-inflammatory mediators (TNF and IL-1β) following stimulation were measured by flow cytometry. RESULTS Women with knee OA (n = 15) had elevated levels of serum c-reactive protein (CRP) and a lower proportion of circulating monocytes. Monocytes from OA participants had elevated expression of the activation markers CD16, CCR2, and HLA-DR and induced greater production of tumor necrosis factor (TNF) and IL-1β compared to healthy controls. Higher serum TNF and BMI were correlated with increased monocyte expression of CCR2. Additionally monocyte CCR2 expression and serum TNF were correlated with worse pain on a validated questionnaire. CONCLUSIONS Our findings suggest monocytes are activated prior to their entry into the synovium. Modulating systemic inflammation and monocyte recruitment to the synovium could be of therapeutic benefit.
Collapse
Affiliation(s)
- D Loukov
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - S Karampatos
- School of Rehabilitation Science, McMaster University, Hamilton, Ontario, Canada
| | - M R Maly
- School of Rehabilitation Science, McMaster University, Hamilton, Ontario, Canada; Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - D M E Bowdish
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
135
|
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
|
136
|
WISP1/CCN4 aggravates cartilage degeneration in experimental osteoarthritis. Osteoarthritis Cartilage 2017; 25:1900-1911. [PMID: 28735021 DOI: 10.1016/j.joca.2017.07.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 07/12/2017] [Accepted: 07/14/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Increased Wisp1 expression was previously reported in experimental and human osteoarthritis (OA). Moreover, adenoviral overexpression of Wisp1 in naïve mouse knee joints resulted in early OA-like cartilage lesions. Here, we determined how the matricellular protein WISP1 is involved in the pathology that occurs in the complex osteoarthritic environment with aging and experimental OA in wild type (WT) and Wisp1-/- mice. METHODS WT and Wisp1-/- mice were aged or experimental OA was induced with intraarticular collagenase injection, destabilization of the medial meniscus (DMM) or anterior cruciate ligament transection (ACLT). Joint pathology was assessed using histology and microCT. Protease expression was evaluated with qRT-PCR and activity was determined by immunohistochemical staining of the aggrecan neoepitope NITEGE. Protease expression in human end-stage OA synovial tissue was determined with qRT-PCR after stimulation with WISP1. RESULTS With aging, spontaneous cartilage degeneration in Wisp1-/- was not decreased compared to their WT controls. However, we observed significantly decreased cartilage degeneration in Wisp1-/- mice after induction of three independent experimental OA models. While the degree of osteophyte formation was comparable between WT and Wisp1-/- mice, increased cortical thickness and reduced trabecular spacing was observed in Wisp1-/- mice. In addition, we observed decreased MMP3/9 and ADAMTS4/5 expression in Wisp1-/- mice, which was accompanied by decreased levels of NITEGE. In line with this, stimulation of human OA synovium with WISP1 increased the expression of various proteases. CONCLUSIONS WISP1 plays an aggravating role in the development of post-traumatic experimental OA.
Collapse
|
137
|
Mesenchymal Stem Cells for Cartilage Regeneration of TMJ Osteoarthritis. Stem Cells Int 2017; 2017:5979741. [PMID: 29123550 PMCID: PMC5662817 DOI: 10.1155/2017/5979741] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/06/2017] [Indexed: 02/05/2023] Open
Abstract
Temporomandibular joint osteoarthritis (TMJ OA) is a degenerative disease, characterized by progressive cartilage degradation, subchondral bone remodeling, synovitis, and chronic pain. Due to the limited self-healing capacity in condylar cartilage, traditional clinical treatments have limited symptom-modifying and structure-modifying effects to restore impaired cartilage as well as other TMJ tissues. In recent years, stem cell-based therapy has raised much attention as an alternative approach towards tissue repair and regeneration. Mesenchymal stem cells (MSCs), derived from the bone marrow, synovium, and even umbilical cord, play a role as seed cells for the cartilage regeneration of TMJ OA. MSCs possess multilineage differentiation potential, including chondrogenic differentiation as well as osteogenic differentiation. In addition, the trophic modulations of MSCs exert anti-inflammatory and immunomodulatory effects under aberrant conditions. Furthermore, MSCs combined with appropriate scaffolds can form cartilaginous or even osseous compartments to repair damaged tissue and impaired function of TMJ. In this review, we will briefly discuss the pathogenesis of cartilage degeneration in TMJ OA and emphasize the potential sources of MSCs and novel approaches for the cartilage regeneration of TMJ OA, particularly focusing on the MSC-based therapy and tissue engineering.
Collapse
|
138
|
Cremers NAJ, van den Bosch MHJ, van Dalen S, Di Ceglie I, Ascone G, van de Loo F, Koenders M, van der Kraan P, Sloetjes A, Vogl T, Roth J, Geven EJW, Blom AB, van Lent PLEM. S100A8/A9 increases the mobilization of pro-inflammatory Ly6C high monocytes to the synovium during experimental osteoarthritis. Arthritis Res Ther 2017; 19:217. [PMID: 28969686 PMCID: PMC5623958 DOI: 10.1186/s13075-017-1426-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/11/2017] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Monocytes are dominant cells present within the inflamed synovium during osteoarthritis (OA). In mice, two functionally distinct monocyte subsets are described: pro-inflammatory Ly6Chigh and patrolling Ly6Clow monocytes. Alarmins S100A8/A9 locally released by the synovium during inflammatory OA for prolonged periods may be dominant proteins involved in stimulating recruitment of Ly6Chigh monocytes from the circulation to the joint. Our objective was to investigate the role of S100A8/A9 in the mobilization of Ly6Chigh and Ly6Clow monocytic populations to the inflamed joint in collagenase-induced OA (CiOA). METHOD S100A8 was injected intra-articularly to investigate monocyte influx. CiOA was induced by injection of collagenase into knee joints of wild-type C57BL/6 (WT), and S100a9-/- mice. Mice were sacrificed together with age-matched saline-injected control mice (n = 6/group), and expression of monocyte markers, pro-inflammatory cytokines, and chemokines was determined in the synovium using ELISA and RT-qPCR. Cells were isolated from the bone marrow (BM), spleen, blood, and synovium and monocytes were identified using FACS. RESULTS S100A8/A9 was highly expressed during CiOA. Intra-articular injection of S100A8 leads to elevated expression of monocyte markers and the monocyte-attracting chemokines CCL2 and CX3CL1 in the synovium. At day 7 (d7) after CiOA induction in WT mice, numbers of Ly6Chigh, but not Ly6Clow monocytes, were strongly increased (7.6-fold) in the synovium compared to saline-injected controls. This coincided with strong upregulation of CCL2, which preferentially attracts Ly6Chigh monocytes. In contrast, S100a9-/- mice showed a significant increase in Ly6Clow monocytes (twofold) within the synovium at CiOA d7, whereas the number of Ly6Chigh monocytes remained unaffected. In agreement with this finding, the Ly6Clow mobilization marker CX3CL1 was significantly higher within the synovium of S100a9-/- mice. Next, we studied the effect of S100A8/A9 on release of Ly6Chigh monocytes from the BM into the circulation. A 14% decrease in myeloid cells was found in WT BM at CiOA d7. No decrease in myeloid cells in S100a9-/- BM was found, suggesting that S100A8/A9 promotes the release of myeloid populations from the BM. CONCLUSION Induction of OA locally leads to strongly elevated S100A8/A9 expression and an elevated influx of Ly6Chigh monocytes from the BM to the synovium.
Collapse
Affiliation(s)
- Niels A J Cremers
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Martijn H J van den Bosch
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Stephanie van Dalen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Irene Di Ceglie
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Giuliana Ascone
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Fons van de Loo
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Marije Koenders
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Peter van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Annet Sloetjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Thomas Vogl
- Institute of Immunology, University of Munster, Munster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Munster, Munster, Germany
| | - Edwin J W Geven
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands.
| |
Collapse
|
139
|
Wu CL, McNeill J, Goon K, Little D, Kimmerling K, Huebner J, Kraus V, Guilak F. Conditional Macrophage Depletion Increases Inflammation and Does Not Inhibit the Development of Osteoarthritis in Obese Macrophage Fas-Induced Apoptosis-Transgenic Mice. Arthritis Rheumatol 2017; 69:1772-1783. [PMID: 28544542 PMCID: PMC5611814 DOI: 10.1002/art.40161] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate whether short-term, systemic depletion of macrophages can mitigate osteoarthritis (OA) following injury in the setting of obesity. METHODS CSF-1R-GFP+ macrophage Fas-induced apoptosis (MaFIA)-transgenic mice that allow conditional depletion of macrophages were placed on a high-fat diet and underwent surgery to induce knee OA. A small molecule (AP20187) was administrated to deplete macrophages in MaFIA mice. The effects of macrophage depletion on acute joint inflammation, OA severity, and arthritic bone changes were evaluated using histology and micro-computed tomography. Immunohistochemical analysis was performed to identify various immune cells. The levels of serum and synovial fluid cytokines were also measured. RESULTS Macrophage-depleted mice had significantly fewer M1 and M2 macrophages in the surgically operated joints relative to controls and exhibited decreased osteophyte formation immediately following depletion. Surprisingly, macrophage depletion did not attenuate the severity of OA in obese mice; instead, it induced systemic inflammation and led to a massive infiltration of CD3+ T cells and particularly neutrophils, but not B cells, into the injured joints. Macrophage-depleted mice also demonstrated a markedly increased number of proinflammatory cytokines including granulocyte colony-stimulating factor, interleukin-1β (IL-1β), IL-6, IL-8, and tumor necrosis factor in both serum and joint synovial fluid, although the mice showed a trend toward decreased levels of insulin and leptin in serum after macrophage depletion. CONCLUSION Our findings indicate that macrophages are vital for modulating homeostasis of immune cells in the setting of obesity and suggest that more targeted approaches of depleting specific macrophage subtypes may be necessary to mitigate inflammation and OA in the setting of obesity.
Collapse
Affiliation(s)
- Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis MO, 63110
- Shriners Hospitals for Children – St. Louis, St. Louis MO, 63110
| | - Jenna McNeill
- Department of Orthopaedic Surgery, Washington University, St. Louis MO, 63110
- Shriners Hospitals for Children – St. Louis, St. Louis MO, 63110
| | - Kelsey Goon
- Department of Orthopaedic Surgery, Washington University, St. Louis MO, 63110
- Shriners Hospitals for Children – St. Louis, St. Louis MO, 63110
| | - Dianne Little
- Departments of Basic Medical Sciences and Biomedical Engineering, Purdue University, West Lafayette, IN, 47907
| | - Kelly Kimmerling
- Department of Orthopaedic Surgery, Washington University, St. Louis MO, 63110
- Shriners Hospitals for Children – St. Louis, St. Louis MO, 63110
| | - Janet Huebner
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham NC 27701
| | - Virginia Kraus
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham NC 27701
- Division of Rheumatology, Department of Medicine, Duke University Medical Center, Durham NC 27701
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis MO, 63110
- Shriners Hospitals for Children – St. Louis, St. Louis MO, 63110
| |
Collapse
|
140
|
Narayanan G, Bhattacharjee M, Nair LS, Laurencin CT. Musculoskeletal Tissue Regeneration: the Role of the Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-017-0036-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
141
|
Sieker JT, Ayturk UM, Proffen BL, Weissenberger MH, Kiapour AM, Murray MM. Immediate Administration of Intraarticular Triamcinolone Acetonide After Joint Injury Modulates Molecular Outcomes Associated With Early Synovitis. Arthritis Rheumatol 2017; 68:1637-47. [PMID: 26866935 DOI: 10.1002/art.39631] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 02/04/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To test whether intraarticular corticosteroid injection mitigates injury-induced synovitis and collagen degradation after anterior cruciate ligament transection (ACLT) and to characterize the synovial response using a functional genomics approach in a preclinical model of posttraumatic osteoarthritis. METHODS Yorkshire pigs underwent unilateral ACLT without subsequent corticosteroid injection (the ACLT group; n = 6) or ACLT with immediate injection of 20 mg triamcinolone acetonide (the steroid group; n = 6). A control group of pigs (the intact group; n = 6) did not undergo surgery. Total synovial membrane cellularity and synovial fluid concentration of C1,2C neoepitope-bearing collagen fragments 14 days after injury were primary end points and were compared between the ACLT, steroid, and intact groups. Cells were differentiated by histologic phenotype and counted, while RNA sequencing was used to quantify transcriptome-wide gene expression and monocyte, macrophage, and lymphocyte markers. RESULTS In the intact group, total cellularity was 13% (95% confidence interval [95% CI] 9-16) and the C1,2C concentration was 0.24 μg/ml (95% CI 0.08-0.39). In the ACLT group, significant increases were observed in total cellularity (to 21% [95% CI 16-27]) and C1,2C concentration (to 0.49 μg/ml [95% CI 0.39-0.59]). Compared to values in the ACLT group, total cellularity in the steroid group was nonsignificantly decreased to 17% (95% CI 15-18) (P = 0.26) and C1,2C concentration in the steroid group was significantly decreased to 0.29 μg/ml (95% CI 0.23-0.35) (P = 0.04). A total of 255 protein-coding transcripts were differentially expressed between the ACLT group and the intact group. These genes mainly enriched pathways related to cellular immune response, proteolysis, and angiogenesis. Mononuclear leukocytes were the dominant cell type in cell-dense areas. MARCO, SOCS3, CCR1, IL4R, and MMP2 expression was significantly associated with C1,2C levels. CONCLUSION Early intraarticular immunosuppression mitigated injury-induced increases in collagen fragments, an outcome better predicted by specific marker expression than by histologic measures of synovitis.
Collapse
Affiliation(s)
- Jakob T Sieker
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, and Orthopaedic Clinic König-Ludwig-Haus, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Ugur M Ayturk
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benedikt L Proffen
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Manuela H Weissenberger
- Orthopaedic Clinic König-Ludwig-Haus, Julius Maximilian University of Würzburg, Würzburg, Germany, and Goethe University Hospital Frankfurt, Frankfurt, Germany
| | - Ata M Kiapour
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Martha M Murray
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
142
|
Manferdini C, Paolella F, Gabusi E, Gambari L, Piacentini A, Filardo G, Fleury-Cappellesso S, Barbero A, Murphy M, Lisignoli G. Adipose stromal cells mediated switching of the pro-inflammatory profile of M1-like macrophages is facilitated by PGE2: in vitro evaluation. Osteoarthritis Cartilage 2017; 25:1161-1171. [PMID: 28153787 DOI: 10.1016/j.joca.2017.01.011] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 01/16/2017] [Accepted: 01/22/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To define if adipose mesenchymal stromal cell (ASC) treatment mediated switching of the pro-inflammatory profile of M1-like macrophages as a means to develop a tailored in vitro efficacy/potency test. DESIGN We firstly performed immunohistochemical analysis of CD68, CD80 (M1-like) and CD206 (M2-like) macrophages in osteoarthritic (OA) synovial tissue. ASC were co-cultured in contact and in transwell with activated (GM-CSF + IFNγ)-M1 macrophages. We analyzed IL1β, TNFα, IL6, MIP1α/CCL3, S100A8, S100A9, IL10, CD163 and CD206 by qRT-PCR or immunoassays. Prostaglandin E2 (PGE2) blocking experiments were performed using PGE2 receptor antagonist. RESULTS In moderate grade OA synovium we did not always find a higher percentage of CD80 with respect to CD206. M1-like-activated macrophage factors IL1β, TNFα, IL6, MIP1α/CCL3, S100A8 and S100A9 were down-modulated both in contact and in transwell by ASC. However, in both systems ASC induced the typical M2-like macrophage markers IL10, CD163 and CD206. Activated-M1-like macrophages pre-treated with PGE2 receptor antagonist failed to decrease secretion of TNFα, IL6 and to increase that of IL10, CD163 and CD206 when co-cultured with ASC confirming a PGE2 specific role. CONCLUSIONS We demonstrated that ASC are responsible for the switching of activated-M1-like inflammatory macrophages to a M2-like phenotype, mainly through PGE2. This evidenced that activated-M1-like macrophages may represent a relevant cell model to test the efficacy/potency of ASC and suggests a specific role of ASC as important determinants in therapeutic dampening of synovial inflammation in OA.
Collapse
Affiliation(s)
- C Manferdini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - F Paolella
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - E Gabusi
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - L Gambari
- SD Laboratorio RAMSES, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - A Piacentini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - G Filardo
- Laboratorio NABI, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | | | - A Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - M Murphy
- Regenerative Medicine Institute, Galway, Ireland.
| | - G Lisignoli
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| |
Collapse
|
143
|
Sample SJ, Racette MA, Hans EC, Volstad NJ, Holzman G, Bleedorn JA, Schaefer SL, Waller KR, Hao Z, Block WF, Muir P. Radiographic and magnetic resonance imaging predicts severity of cruciate ligament fiber damage and synovitis in dogs with cranial cruciate ligament rupture. PLoS One 2017; 12:e0178086. [PMID: 28575001 PMCID: PMC5456057 DOI: 10.1371/journal.pone.0178086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 05/07/2017] [Indexed: 11/19/2022] Open
Abstract
Cruciate ligament rupture (CR) and associated osteoarthritis (OA) is a common condition in dogs. Dogs frequently develop a second contralateral CR. This study tested the hypothesis that the degree of stifle synovitis and cranial cruciate ligament (CrCL) matrix damage in dogs with CR is correlated with non-invasive diagnostic tests, including magnetic resonance (MR) imaging. We conducted a prospective cohort study of 29 client-owned dogs with an unstable stifle due to complete CR and stable contralateral stifle with partial CR. We evaluated correlation of stifle synovitis and CrCL fiber damage with diagnostic tests including bilateral stifle radiographs, 3.0 Tesla MR imaging, and bilateral stifle arthroscopy. Histologic grading and immunohistochemical staining for CD3+ T lymphocytes, TRAP+ activated macrophages and Factor VIII+ blood vessels in bilateral stifle synovial biopsies were also performed. Serum and synovial fluid concentrations of C-reactive protein (CRP) and carboxy-terminal telopeptide of type I collagen (ICTP), and synovial total nucleated cell count were determined. Synovitis was increased in complete CR stifles relative to partial CR stifles (P<0.0001), although total nucleated cell count in synovial fluid was increased in partial CR stifles (P<0.01). In partial CR stifles, we found that 3D Fast Spin Echo Cube CrCL signal intensity was correlated with histologic synovitis (SR = 0.50, P<0.01) and that radiographic OA was correlated with CrCL fiber damage assessed arthroscopically (SR = 0.61, P<0.001). Taken together, results of this study show that clinical diagnostic tests predict severity of stifle synovitis and cruciate ligament matrix damage in stable partial CR stifles. These data support use of client-owned dogs with unilateral complete CR and contralateral partial CR as a clinical trial model for investigation of disease-modifying therapy for partial CR.
Collapse
Affiliation(s)
- Susannah J. Sample
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Molly A. Racette
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Eric C. Hans
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nicola J. Volstad
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Gerianne Holzman
- UW Veterinary Care Hospital, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jason A. Bleedorn
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Susan L. Schaefer
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kenneth R. Waller
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Zhengling Hao
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Walter F. Block
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Peter Muir
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
144
|
Hermeto LC, Rossi RD, Jardim PHDA, Santana AE, Rinaldi JDC, Justulin LA. Comparison between two different experimental models of osteoarthritis in rabbits. Intra-articular collagenase injection and anterior cruciate ligament transection. Acta Cir Bras 2017; 31:602-607. [PMID: 27737345 DOI: 10.1590/s0102-865020160090000005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/23/2016] [Indexed: 11/22/2022] Open
Abstract
PURPOSE: To compare two different experimental models of osteoarthritis in rabbits: intra-articular collagenase injection and anterior cruciate ligament transection. METHODS: Ten adult rabbits were randomly divided in two groups: COLL (collagenase group) and ACLT (anterior cruciate ligament transection). The COLL group was treated with 0.5 ml collagenase solution (2mg collagenase/0.5 ml sterile PBS), and the ACTL group was subjected to anterior cruciate ligament. After six and twelve weeks, respectively, the animals in the COLL and ACTL groups were euthanized. The gross appearance and histological examinations conducted in the cartilage articular surface was blindly scored according to the criteria developed by Yoshimi et al. (1994) and Mankin et al. (1971), respectively. RESULTS: The gross morphologic observation, macroscopic score and histological examinations have demonstrated that the ACTL group presented the highest scores, and lesions more severe than those in the COLL group. CONCLUSIONS: Both methods, anterior cruciate ligament transection and collagenase, applied to the stifle joint of the rabbits have effectively induced degenerative changes in the cartilage tissue, through statistically significant analysis (p≤0.05). The ACTL method has presented more severe lesions.
Collapse
Affiliation(s)
- Larissa Correa Hermeto
- PhD, Postdoctoral Student, Postgraduate Program in Veterinary Sciences, Veterinary Medicine and Animal Science School, Universidade Federal do Mato Grosso do Sul (UFMS), Brazil. Acquisition of data, conception and design of the study, technical procedures, statistical analysis, manuscript preparation and writing, critical revision
| | - Rafael De Rossi
- PhD, Associate Professor, Anesthesiology and Surgery Sector, Department of Veterinary Medicine, Veterinary Medicine and Animal Science School, UFMS, Campo Grande-MS, Brazil. Scientific and intellectual content of the study, analysis and interpretation of data, final approval
| | - Paulo Henrique de Affonseca Jardim
- DVM, Department of Veterinary Medicine, School of Veterinary Medicine and Animal Science School, UFMS, Campo Grande-MS, Brazil. Technical procedures, statistical analysis
| | - Aureo Evangelista Santana
- PhD, Full Professor, Department of Veterinary Clinical Medicine and Surgery, Agrarian and Veterinary Sciences School, Universidade Estadual de São Paulo (UNESP), Jaboticabal-SP, Brazil. Scientific and intellectual content of the study, analysis and interpretation of data
| | - Jaqueline de Carvalho Rinaldi
- PhD, Postdoctoral Student, Institute of Biosciences, Department of Morphology, Extracellular Matrix Laboratory, UNESP, Botucatu-SP, Brazil. Histopathological examinations
| | - Luis Antonio Justulin
- PhD, Associate Professor, Institute of Biosciences, Department of Morphology, Extracellular Matrix Laboratory, UNESP, Botucatu-SP, Brazil. Histopathological examinations
| |
Collapse
|
145
|
Hsia AW, Anderson MJ, Heffner MA, Lagmay EP, Zavodovskaya R, Christiansen BA. Osteophyte formation after ACL rupture in mice is associated with joint restabilization and loss of range of motion. J Orthop Res 2017; 35:466-473. [PMID: 27031945 PMCID: PMC5603200 DOI: 10.1002/jor.23252] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/29/2016] [Indexed: 02/04/2023]
Abstract
Osteophytes are a typical radiographic finding during osteoarthritis (OA). Osteophytes are thought to form in response to joint instability; however, the time course of osteophyte formation and joint stabilization following joint injury is not well understood. In this study, we investigated the time course of osteophyte formation and joint function following non-invasive knee injury in mice. We hypothesized that initial joint instability following knee injury would initiate osteophyte formation, which would in turn restabilize the joint and reduce range of motion (ROM). Mice were subjected to non-invasive anterior cruciate ligament (ACL) rupture. Anterior-posterior (AP) joint laxity, ROM, and chondro/osteophyte formation were measured immediately after injury, and 2, 4, 6, and 8 weeks post-injury. Chondrophyte areas at each time point were measured with histology, while mineralized osteophyte volume was determined using micro-computed tomography. Immediately after ACL rupture, AP joint laxity was increased twofold, while ROM was increased 11.7%. Chondrophytes appeared by 2 weeks post-injury, corresponding with a decrease in AP joint laxity and ROM. By 8 weeks post-injury, considerable osteophyte formation was observed around the joint, AP joint laxity returned to control levels, and joint ROM decreased to 61% of control values. These data support a role for chondro/osteophytes in joint restabilization after injury, and provide crucial insight into the time course and pathology of joint degeneration during OA development in the mouse. Statement of Clinical Significance: Results from this study increase understanding of conditions leading to osteophyte formation.© 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:466-473, 2017.
Collapse
Affiliation(s)
- Allison W. Hsia
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA
| | | | - Mollie A. Heffner
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA
| | - Earl P. Lagmay
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA
| | - Regina Zavodovskaya
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Blaine A. Christiansen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA,Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA
| |
Collapse
|
146
|
de Visser HM, Weinans H, Coeleveld K, van Rijen MHP, Lafeber FPJG, Mastbergen SC. Groove model of tibia-femoral osteoarthritis in the rat. J Orthop Res 2017; 35:496-505. [PMID: 27183198 PMCID: PMC5363339 DOI: 10.1002/jor.23299] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/06/2016] [Indexed: 02/04/2023]
Abstract
Several experimental models of osteoarthritis in rats are used to study the pathophysiology of osteoarthritis. Many mechanically induced models have the limitation that permanent joint instability is induced by, for example, ligament transection or meniscal damage. This permanent instability will counteract the potential beneficial effects of therapy. The groove model of osteoarthritis uses a one-time trigger, surgically induced cartilage damage on the femoral condyles, and has been validated for the canine tibia-femoral compartment. The present study evaluates this model for the rat knee joint. The articular cartilage of the weight bearing surface of both femoral condyles and trochlea were damaged (grooved) without damaging the underlying subchondral bone. Severity of joint degeneration was histologically assessed, in addition to patella cartilage damage, and subchondral bone characteristics by means of (contrast-enhanced) micro-CT. Mild histological degeneration of the surgically untouched tibial plateau cartilage was observed in addition to damage of the femoral condyles, without clear synovial tissue inflammation. Contrast enhanced micro-CT demonstrated proteoglycan loss of the surgically untouched patella cartilage. Besides, a more sclerotic structure of the subchondral bone was observed. The tibia-femoral groove model in a rat results in mild knee joint degeneration, without permanent joint instability and joint inflammation. This makes the rat groove model a useful model to study the onset and progression of post-traumatic non-inflammatory osteoarthritis, creating a relatively sensitive model to study disease modifying osteoarthritic drugs. © 2016 The Authors. Journal of Orthopaedic Research published by Wiley Periodicals, Inc. on behalf of the Orthopaedic Research Society. J Orthop Res 35:496-505, 2017.
Collapse
Affiliation(s)
- Huub M. de Visser
- Department of Rheumatology and Clinical Immunology; University Medical Center Utrecht; UMC Utrecht, F.02.127, P.O. Box 85500 3508 GA Utrecht The Netherlands
- Department of Orthopaedics; University Medical Center Utrecht; Utrecht The Netherlands
| | - Harrie Weinans
- Department of Rheumatology and Clinical Immunology; University Medical Center Utrecht; UMC Utrecht, F.02.127, P.O. Box 85500 3508 GA Utrecht The Netherlands
- Department of Orthopaedics; University Medical Center Utrecht; Utrecht The Netherlands
- Department of Biomechanical Engineering; Delft University of Technology; Delft The Netherlands
| | - Katja Coeleveld
- Department of Rheumatology and Clinical Immunology; University Medical Center Utrecht; UMC Utrecht, F.02.127, P.O. Box 85500 3508 GA Utrecht The Netherlands
| | | | - Floris P. J. G. Lafeber
- Department of Rheumatology and Clinical Immunology; University Medical Center Utrecht; UMC Utrecht, F.02.127, P.O. Box 85500 3508 GA Utrecht The Netherlands
| | - Simon C. Mastbergen
- Department of Rheumatology and Clinical Immunology; University Medical Center Utrecht; UMC Utrecht, F.02.127, P.O. Box 85500 3508 GA Utrecht The Netherlands
| |
Collapse
|
147
|
|
148
|
Paduszyński W, Jeśkiewicz M, Uchański P, Gackowski S, Radkowski M, Demkow U. Hoffa's Fat Pad Abnormality in the Development of Knee Osteoarthritis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1039:95-102. [PMID: 28770522 DOI: 10.1007/5584_2017_77] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past two decades, many hypotheses have been put forward to explain the cause of knee osteoarthritis. Scientific reports bring up the role of adipose tissue in the activation of the inflammatory mechanisms, which is a characteristic feature of osteoarthritis natural history. Adipose tissue produces and releases cytokines, interleukins, and growth factors by means of paracrine, endocrine, and autocrine mechanisms. Hoffa's fat pad (infrapatellar adipose tissue) plays a viable role in the initiation and progression of osteoarthritis due to its role in the activation and release of pro-inflammatory mediators. The degenerative joint disease is considered an inflammatory process. Therefore, in this article we overview the importance of Hoffa's fat pad in the development and progression of osteoarthritis.
Collapse
Affiliation(s)
- Wojciech Paduszyński
- Department of Neurotraumatology and Traumatology, Children's University Hospital, Warsaw Medical University, 63A Żwirki i Wigury Street, 02-091, Warsaw, Poland
| | - Mateusz Jeśkiewicz
- Department of Neurotraumatology and Traumatology, Children's University Hospital, Warsaw Medical University, 63A Żwirki i Wigury Street, 02-091, Warsaw, Poland
| | - Paweł Uchański
- Department of Neurotraumatology and Traumatology, Children's University Hospital, Warsaw Medical University, 63A Żwirki i Wigury Street, 02-091, Warsaw, Poland
| | - Sebastian Gackowski
- Department of Neurotraumatology and Traumatology, Children's University Hospital, Warsaw Medical University, 63A Żwirki i Wigury Street, 02-091, Warsaw, Poland
| | - Marek Radkowski
- Department of Immunopathology of Infectious and Parasitic Diseases, Warsaw Medical University, 3C Pawinskiego Street, 02-106, Warsaw, Poland
| | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Warsaw Medical University, 63A Żwirki i Wigury Street, 02-091, Warsaw, Poland.
| |
Collapse
|
149
|
Utomo L, Bastiaansen-Jenniskens YM, Verhaar JAN, van Osch GJVM. Cartilage inflammation and degeneration is enhanced by pro-inflammatory (M1) macrophages in vitro, but not inhibited directly by anti-inflammatory (M2) macrophages. Osteoarthritis Cartilage 2016; 24:2162-2170. [PMID: 27502245 DOI: 10.1016/j.joca.2016.07.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 07/09/2016] [Accepted: 07/28/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Macrophages play a crucial role in the progression of osteoarthritis (OA). Their phenotype may range from pro-inflammatory to anti-inflammatory. The aim of this study was to evaluate the direct effects of macrophage subtypes on cartilage by culturing macrophage conditioned medium (MCM) on human articular cartilage. DESIGN Human OA cartilage explants were cultured with MCM of pro-inflammatory M(IFNγ+TNFα), or anti-inflammatory M(IL-4) or M(IL-10) human monocyte-derived macrophages. To assess effects of anti-inflammatory macrophages, the cartilage was cultured with a combination of MCM phenotypes as well as pre-stimulated with IFNγ+TNFα cartilage before culture with MCM. The reactions of the explants were assessed by gene expression, nitric oxide (NO) production and release of glycosaminoglycans (GAGs). RESULTS M(IFNγ+TNFα) MCM affected OA cartilage by upregulation of IL1B (Interleukin 1β), IL6, MMP13 (Matrix Metalloproteinase-13) and ADAMTS5 (A Disintegrin And Metalloproteinase with Thrombospondin Motifs-5), while inhibiting ACAN (aggrecan) and COL2A1 (collagen type II). M(IL-10) upregulated IL1B and Suppressor of cytokine signaling 1 (SOCS1). NO production and GAG release by the cartilage was increased when cultured with M(IFNγ+TNFα) MCM. M(IL-4) and M(IL-10) did not inhibit the effects of M(IFNγ+TNFα) MCM of neither phenotype affected IFNγ+TNFα pre-stimulated cartilage, in which an inflammatory gene response was deliberately induced. CONCLUSION M(IFNγ+TNFα) macrophages have a prominent direct effect on OA cartilage, while M(IL-4) and M(IL-10) do not inhibit the effects of M(IFNγ+TNFα), or IFNγ+TNFα induced inflammation of the cartilage. Therapies aiming at inhibiting cartilage degeneration may take this into account by directing suppression of pro-inflammatory macrophages or stimulation of anti-inflammatory macrophages.
Collapse
Affiliation(s)
- L Utomo
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Y M Bastiaansen-Jenniskens
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - J A N Verhaar
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - G J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
150
|
Miron RJ, Zohdi H, Fujioka-Kobayashi M, Bosshardt DD. Giant cells around bone biomaterials: Osteoclasts or multi-nucleated giant cells? Acta Biomater 2016; 46:15-28. [PMID: 27667014 DOI: 10.1016/j.actbio.2016.09.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 09/14/2016] [Accepted: 09/22/2016] [Indexed: 12/31/2022]
Abstract
Recently accumulating evidence has put into question the role of large multinucleated giant cells (MNGCs) around bone biomaterials. While cells derived from the monocyte/macrophage lineage are one of the first cell types in contact with implanted biomaterials, it was originally thought that specifically in bone tissues, all giant cells were bone-resorbing osteoclasts whereas foreign body giant cells (FBGCs) were found associated with a connective tissue foreign body reaction resulting in fibrous encapsulation and/or material rejection. Despite the great majority of bone grafting materials routinely found with large osteoclasts, a special subclass of bone biomaterials has more recently been found surrounded by large giant cells virtually incapable of resorbing bone grafts even years after their implantation. While original hypotheses believed that a 'foreign body reaction' may be taking place, histological data retrieved from human samples years after their implantation have put these original hypotheses into question by demonstrating better and more stable long-term bone volume around certain bone grafts. Exactly how or why this 'special' subclass of giant cells is capable of maintaining long-term bone volume, or methods to scientifically distinguish them from osteoclasts remains extremely poorly studied. The aim of this review article was to gather the current available literature on giant cell markers and differences in expression patterns between osteoclasts and MNGCs utilizing 19 specific markers including an array of CD-cell surface markers. Furthermore, the concept of now distinguishing between pro-inflammatory M1-MNGCs (previously referred to as FBGCs) as well as wound-healing M2-MNGCs is introduced and discussed. STATEMENT OF SIGNIFICANCE This review article presents 19 specific cell-surface markers to distinguish between osteoclasts and MNGCs including an array of CD-cell surface markers. Furthermore, the concept of now distinguishing between pro-inflammatory M1-MNGCs (often previously referred to as FBGCs) as well as wound-healing M2-MNGCs is introduced and discussed. The proposed concepts and guidelines aims to guide the next wave of research facilitating the differentiation between osteoclast/MNGCs formation, as well as provides the basis for increasing our understanding of the exact function of MNGCs in bone tissue/biomaterial homeostasis.
Collapse
|