1
|
Traweger A, Scott A, Kjaer M, Wezenbeek E, Scattone Silva R, Kennedy JG, Butler JJ, Gomez-Florit M, Gomes ME, Snedeker JG, Dakin SG, Wildemann B. Achilles tendinopathy. Nat Rev Dis Primers 2025; 11:20. [PMID: 40148342 DOI: 10.1038/s41572-025-00602-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/29/2025]
Abstract
Achilles tendon pathologies are prevalent, impacting ~6% of the general population and up to 50% of elite endurance runners over their lifetimes. These conditions substantially affect quality of life and work productivity, leading to substantial societal costs. Achilles tendinopathy (AT) is a condition marked by localized pain and functional impairment related to mechanical loading. AT can considerably impair participation and potentially also performance in sports and daily activities. The aetiology of AT is multifactorial and repetitive overloading of the tendon is often observed as the inciting factor by health professionals. However, AT can also be associated with adverse effects of certain medication, ageing and various comorbidities. Characteristic tendon changes include proteoglycan accumulation, fluid accumulation with swelling and hypervascularization. Tissue disorganization advances as pathological changes in matrix structure are driven by altered cellular function and makeup, often accompanied by persistent inflammation. Treatment strategies include various interventions, although these can be protracted and challenging for both patients and health-care providers, often with high failure rates. Current research focuses on understanding the pathological processes at the cellular and molecular levels to distinguish between disease categories and to investigate the role of inflammation, metabolic maladaptation and mechanical stress. Emerging therapeutic approaches need to be developed to address these underlying mechanisms. These approaches focus on optimizing rehabilitation protocols and advancing the development of adjunct therapies, such as advanced therapy medicinal products, alongside the integration of precision medicine to improve treatment outcomes.
Collapse
Affiliation(s)
- Andreas Traweger
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria.
| | - Alex Scott
- Department of Physical Therapy, Centre for Aging SMART, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Evi Wezenbeek
- MOVANT Research Group, Department of Rehabilitation Sciences and Physiotherapy (REVAKI), University of Antwerp, Wilrijk, Belgium
- Department of Rehabilitation Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Rodrigo Scattone Silva
- Health Sciences College of Trairi, Federal University of Rio Grande do Norte, Santa Cruz, Brazil
| | - John G Kennedy
- NYU Langone Health, NYU Langone Orthopedic Hospital, New York, NY, USA
| | - James J Butler
- NYU Langone Health, NYU Langone Orthopedic Hospital, New York, NY, USA
| | - Manuel Gomez-Florit
- Health Research Institute of the Balearic Islands (IdISBa), Research Unit Son Espases University Hospital (HUSE), Palma, Spain
- Group of Cell Therapy and Tissue Engineering (TERCIT), Research Institute on Health Sciences (IUNICS), University of the Balearic Islands (UIB), Palma, Spain
| | - Manuela E Gomes
- School of Medicine and Biomedical Sciences (ICBAS), Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
| | - Jess G Snedeker
- Department of Orthopaedics, Balgrist, University of Zurich, Zurich, Switzerland
- Institute for Biomechanics, ETH Zurich, Zürich, Switzerland
| | - Stephanie G Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Britt Wildemann
- Experimental Trauma Surgery, Department of Trauma, Hand and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
2
|
Xu Z, Hou W, Zhang T, Chen R, Skutella T. Exploring molecular and cellular signaling pathways: Unraveling the pathogenesis of tendinopathy. J Orthop Translat 2025; 51:298-311. [PMID: 40201708 PMCID: PMC11978293 DOI: 10.1016/j.jot.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/09/2025] [Accepted: 02/05/2025] [Indexed: 04/10/2025] Open
Abstract
Despite the long healing duration of tendon injuries, the outcomes of repairs are frequently suboptimal, resulting in persistent pain and reduced functionality. Current clinical approaches to tendinopathy are primarily symptomatic, encompassing nonsteroidal anti-inflammatory drugs (NSAIDs), corticosteroid injections, physical therapies, surgical interventions, loading programs, and pain management. Yet, these treatments have protracted timelines and their efficacy remains uncertain. This uncertainty stems largely from an incomplete understanding of tendinopathy's pathogenesis. Unraveling the mechanisms behind tendinopathy is essential for devising novel therapeutic strategies. In this context, this review systematic reviewed more recent cellular and molecular literature in tendinopathy, in order to summarize the up-to-date advancements including the structure and composition of healthy tendons, the pathophysiological changes in tendinopathy, the molecular pathways implicated in various forms of the condition, and current effective treatment methods. This review not only aims to offer insights but also to inspire further investigation into the mechanisms and clinical management of tendinopathy. The translational potential of this article A deficient understanding of the molecular mechanisms hampers the advancement of therapeutic strategies and drug development. Consequently, an in-depth examination of these molecular mechanisms is essential for comprehending the etiology of tendinopathy and for devising effective clinical management strategies.
Collapse
Affiliation(s)
- Zihan Xu
- Department of Neuroanatomy, Group for Regeneration and Reprogramming, Institute for Anatomy and Cell Biology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
| | - Wenjing Hou
- Department of Neuroanatomy, Group for Regeneration and Reprogramming, Institute for Anatomy and Cell Biology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
| | - Tao Zhang
- Department of Neuroanatomy, Group for Regeneration and Reprogramming, Institute for Anatomy and Cell Biology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
| | - Rui Chen
- Department of Reproductive Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Thomas Skutella
- Department of Neuroanatomy, Group for Regeneration and Reprogramming, Institute for Anatomy and Cell Biology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
| |
Collapse
|
3
|
Millar NL, McInnes IB, Kolbinger F, Raulf F, Akbar M, Li Y, Beckmann N, Accart N, Leupin O, Calonder C, Schieker M, Kneissel M, Bruns C, Siegel RM, Weber E. Targeting the IL-17A pathway for therapy in early-stage tendinopathy. RMD Open 2025; 11:e004729. [PMID: 39988349 PMCID: PMC11881027 DOI: 10.1136/rmdopen-2024-004729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 02/03/2025] [Indexed: 02/25/2025] Open
Abstract
OBJECTIVES Tendinopathy is a frequent clinical problem and represents an extraordinary health economic and socioeconomic burden with high unmet medical needs. Recent clinical evidence suggests blockade of interleukin 17A (IL-17A) for tendinopathy therapy. The present preclinical study elucidates the biological mechanisms of IL-17A pathway stimulation and blockade in tendinopathy. METHODS We explored whether IL-17A and other IL-17 family members are differentially expressed in biopsies of healthy, early-stage and late-stage tendinopathic human rotator cuff tendons using RT-qPCR. IL-17 pathway signature genes in healthy human tendon-derived cells were identified following IL-17A stimulation using AmpliSeq RNA. The molecular, structural and functional consequences of IL-17A pathway stimulation were explored in healthy human tendon-derived cells and in a rat tendon fascicle model ex vivo. The effects of IL-17A pathway blockade were investigated in a rat model of rotator cuff tendinopathy in vivo. RESULTS We provide evidence of differential expression of IL-17A mRNA (IL17A) versus other IL-17 family members in human rotator cuff early-stage tendinopathy. In human tendon-derived cells, stimulation with IL-17A induced the expression of the selected IL-17A pathway signature genes NFKBIZ, ZC3H12A, CXCL1, IL6, MMP3. Expression was inhibited by IL-17A blockade. In the rat ex vivo and in vivo models, IL-17A blockade alleviated inflammatory immune effector release, tendon structural degeneration, tendon inflammation and impaired tendon function. CONCLUSION Our data provide evidence that IL-17A is a key contributor to the pathogenesis of tendinopathy by promoting tendon inflammation and degeneration and that IL-17A blockade may represent a potential therapy in early-stage tendinopathy.
Collapse
Affiliation(s)
- Neal L Millar
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | | | | | - Moeed Akbar
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Yufei Li
- Novartis Pharma AG, Biomedical Research, Basel, Switzerland
| | | | | | - Olivier Leupin
- Novartis Pharma AG, Biomedical Research, Basel, Switzerland
| | | | | | | | | | | | - Eckhard Weber
- Novartis Pharma AG, Biomedical Research, Basel, Switzerland
| |
Collapse
|
4
|
Ling SKK, Liang Z, Lui PPY. High-fat diet-induced obesity exacerbated collagenase-induced tendon injury with upregulation of interleukin-1beta and matrix metalloproteinase-1. Connect Tissue Res 2024; 65:447-457. [PMID: 39364694 DOI: 10.1080/03008207.2024.2409751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024]
Abstract
AIMS Obesity increases tendinopathy's risk, but its mechanisms remain unclear. This study examined the effect of high-fat diet (HFD)-induced obesity on the outcomes and inflammation of collagenase-induced (CI) tendon injury. METHODS Mice were fed with standard chow (SC) or HFD for 12 weeks. Bacterial collagenase I or saline was injected over the patellar tendons of each mouse. At weeks 2 and 8 post-injection, the patellar tendons were harvested for histology, immunohistochemical staining, and gait analysis. The difference (Δ) of limb-idleness index (LII) at the time of post-injury and pre-injury states was calculated. Biomechanical test of tendons was also performed at week 8 post-injection. RESULTS HFD aggravated CI tendon injury with an increase in vascularity and cellularity compared to SC treatment. The histopathological score (week 2: p = 0.025; week 8: p = 0.013) and ΔLII (week 2: p = 0.012; week 8: p = 0.005) were significantly higher in the HFD group compared to those in the SC group after CI tendon injury. Stiffness (saline: p = 0.003; CI: p = 0.010), ultimate stress (saline: p < 0.001; CI: p = 0.006), and Young's modulus (saline: p = 0.017; CI: p = 0.007) were significantly lower in the HFD group compared to the SC group at week 8 after saline or collagenase injection. HFD induced higher expression of IL-1β (week 2: p = 0.010; week 8: p = 0.025) and MMP-1 (week 2: p = 0.010; week 8: p = 0.004) compared to SC treatment after CI tendon injury at both time points. CONCLUSIONS HFD-induced obesity exacerbated histopathological, functional, and biomechanical changes in the CI tendon injury model, which was associated with an upregulation of IL-1β and MMP-1.
Collapse
Affiliation(s)
- Samuel Ka-Kin Ling
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Zuru Liang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Pauline Po Yee Lui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine Ltd, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
5
|
Nakamichi R, Asahara H. The role of mechanotransduction in tendon. J Bone Miner Res 2024; 39:814-820. [PMID: 38795012 PMCID: PMC11301520 DOI: 10.1093/jbmr/zjae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/04/2024] [Accepted: 05/24/2024] [Indexed: 05/27/2024]
Abstract
Tendons play an important role in the maintenance of motor function by connecting muscles and bones and transmitting forces. Particularly, the role of mechanical stress has primarily focused on the key mechanism of tendon homeostasis, with much research on this topic. With the recent development of molecular biological techniques, the mechanisms of mechanical stress sensing and signal transduction have been gradually elucidated with the identification of mechanosensor in tendon cells and the master regulator in tendon development. This review provides a comprehensive overview of the structure and function of tendon tissue, including the role for physical performance and the detailed mechanism of mechanotransduction in its regulation. An important lesson is that the role of mechanotransduction in tendon tissue is only partially clarified, indicating the complexity of the mechanisms of motor function and fueling increasing interest in uncovering these mechanisms.
Collapse
Affiliation(s)
- Ryo Nakamichi
- Department of Molecular and Cellular Biology, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, United States
- Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
- Department of Orthopaedic Surgery, Okayama University hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama 700-8558, Japan
| | - Hiroshi Asahara
- Department of Molecular and Cellular Biology, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, United States
- Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| |
Collapse
|
6
|
Lu P, Dai G, Shi L, Li Y, Zhang M, Wang H, Rui Y. HMGB1 Modulates High Glucose-Induced Erroneous Differentiation of Tendon Stem/Progenitor Cells through RAGE/ β-Catenin Pathway. Stem Cells Int 2024; 2024:2335270. [PMID: 38633380 PMCID: PMC11022503 DOI: 10.1155/2024/2335270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
The association of tendinopathy with diabetes has been well recognized. Tendon stem/progenitor cells (TSPCs) play critical roles in tendon repair, regeneration, and homeostasis maintenance. Diabetic TSPCs exhibit enhanced erroneous differentiation and are involved in the pathogenesis of diabetic tendinopathy, whereas the underlying mechanism of the erroneous differentiation of TSPCs remains unclear. Here, we showed that high glucose treatment promoted the erroneous differentiation of TSPCs with increased osteogenic differentiation capacity and decreased tenogenic differentiation ability, and stimulated the expression and further secretion of HMGB1 in TSPCs and. Functionally, exogenous HMGB1 significantly enhanced the erroneous differentiation of TSPCs, while HMGB1 knockdown mitigated high glucose-promoted erroneous differentiation of TSPCs. Mechanistically, the RAGE/β-catenin signaling was activated in TSPCs under high glucose, and HMGB1 knockdown inhibited the activity of RAGE/β-catenin signaling. Inhibition of RAGE/β-catenin signaling could ameliorate high glucose-induced erroneous differentiation of TSPCs. These results indicated that HMGB1 regulated high glucose-induced erroneous differentiation of TSPCs through the RAGE/β-catenin signaling pathway. Collectively, our findings suggest a novel essential mechanism of the erroneous differentiation of TSPCs, which might contribute to the pathogenesis of diabetic tendinopathy and provide a promising therapeutic target and approach for diabetic tendinopathy.
Collapse
Affiliation(s)
- Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Guangchun Dai
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Liu Shi
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Yingjuan Li
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Department of Geriatrics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Ming Zhang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| |
Collapse
|
7
|
Zhang J, Brown R, Hogan MV, Wang JHC. Mitigating Scar Tissue Formation in Tendon Injuries: Targeting HMGB1, AMPK Activation, and Myofibroblast Migration All at Once. Pharmaceuticals (Basel) 2023; 16:1739. [PMID: 38139865 PMCID: PMC10748062 DOI: 10.3390/ph16121739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Tendon injuries, while prevalent, present significant challenges regarding their structural and functional restoration. Utilizing alpha-smooth muscle actin (α-SMA)-Ai9-scleraxis (Scx)-green fluorescent protein (GFP) transgenic mice, which exhibit both Scx (a tendon cell marker) and α-SMA (a myofibroblast marker), we explored the effects of metformin (Met) on tendon healing, repair, and its mechanisms of action. Our findings revealed that intraperitoneal (IP) injections of Met, administered before or after injury, as well as both, effectively prevented the release of HMGB1 into the tendon matrix and reduced circulating levels of HMGB1. Additionally, Met treatment increased and activated AMPK and suppressed TGF-β1 levels within the healing tendon. Tendon healing was also improved by blocking the migration of α-SMA+ myofibroblasts, reducing the prevalence of disorganized collagen fibers and collagen type III. It also enhanced the presence of collagen type I. These outcomes highlight Met's anti-fibrotic properties in acutely injured tendons and suggest its potential for repurposing as a therapeutic agent to minimize scar tissue formation in tendon injuries, which could have profound implications in clinical practice.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
| | - Roshawn Brown
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
8
|
Kim SH, Lee SH. Updates on ankylosing spondylitis: pathogenesis and therapeutic agents. JOURNAL OF RHEUMATIC DISEASES 2023; 30:220-233. [PMID: 37736590 PMCID: PMC10509639 DOI: 10.4078/jrd.2023.0041] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023]
Abstract
Ankylosing spondylitis (AS) is an autoinflammatory disease that manifests with the unique feature of enthesitis. Gut microbiota, HLA-B*27, and biomechanical stress mutually influence and interact resulting in setting off a flame of inflammation. In the HLA-B*27 positive group, dysbiosis in the gut environment disrupts the barrier to exogenous bacteria or viruses. Additionally, biomechanical stress induces inflammation through enthesial resident or gut-origin immune cells. On this basis, innate and adaptive immunity can propagate inflammation and lead to chronic disease. Finally, bone homeostasis is regulated by cytokines, by which the inflamed region is substituted into new bone. Agents that block cytokines are constantly being developed to provide diverse therapeutic options for preventing the progression of inflammation. In addition, some antibodies have been shown to distinguish disease selectively, which support the involvement of autoimmune immunity in AS. In this review, we critically analyze the complexity and uniqueness of the pathogenesis with updates on the findings of immunity and provide new information about biologics and biomarkers.
Collapse
Affiliation(s)
- Se Hee Kim
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Hoon Lee
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
9
|
Luo J, Wang Z, Tang C, Yin Z, Huang J, Ruan D, Fei Y, Wang C, Mo X, Li J, Zhang J, Fang C, Li J, Chen X, Shen W. Animal model for tendinopathy. J Orthop Translat 2023; 42:43-56. [PMID: 37637777 PMCID: PMC10450357 DOI: 10.1016/j.jot.2023.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 08/29/2023] Open
Abstract
Background Tendinopathy is a common motor system disease that leads to pain and reduced function. Despite its prevalence, our mechanistic understanding is incomplete, leading to limited efficacy of treatment options. Animal models contribute significantly to our understanding of tendinopathy and some therapeutic options. However, the inadequacies of animal models are also evident, largely due to differences in anatomical structure and the complexity of human tendinopathy. Different animal models reproduce different aspects of human tendinopathy and are therefore suitable for different scenarios. This review aims to summarize the existing animal models of tendinopathy and to determine the situations in which each model is appropriate for use, including exploring disease mechanisms and evaluating therapeutic effects. Methods We reviewed relevant literature in the PubMed database from January 2000 to December 2022 using the specific terms ((tendinopathy) OR (tendinitis)) AND (model) AND ((mice) OR (rat) OR (rabbit) OR (lapin) OR (dog) OR (canine) OR (sheep) OR (goat) OR (horse) OR (equine) OR (pig) OR (swine) OR (primate)). This review summarized different methods for establishing animal models of tendinopathy and classified them according to the pathogenesis they simulate. We then discussed the advantages and disadvantages of each model, and based on this, identified the situations in which each model was suitable for application. Results For studies that aim to study the pathophysiology of tendinopathy, naturally occurring models, treadmill models, subacromial impingement models and metabolic models are ideal. They are closest to the natural process of tendinopathy in humans. For studies that aim to evaluate the efficacy of possible treatments, the selection should be made according to the pathogenesis simulated by the modeling method. Existing tendinopathy models can be classified into six types according to the pathogenesis they simulate: extracellular matrix synthesis-decomposition imbalance, inflammation, oxidative stress, metabolic disorder, traumatism and mechanical load. Conclusions The critical factor affecting the translational value of research results is whether the selected model is matched with the research purpose. There is no single optimal model for inducing tendinopathy, and researchers must select the model that is most appropriate for the study they are conducting. The translational potential of this article The critical factor affecting the translational value of research results is whether the animal model used is compatible with the research purpose. This paper provides a rationale and practical guide for the establishment and selection of animal models of tendinopathy, which is helpful to improve the clinical transformation ability of existing models and develop new models.
Collapse
Affiliation(s)
- Junchao Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Zetao Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Zi Yin
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jiayun Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xianan Mo
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jiajin Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
| | - Jun Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Department of Orthopedics, Longquan People's Hospital, Zhejiang, 323799, China
| | - Cailian Fang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
| | - Jianyou Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Zhejiang University Huzhou Hospital, 313000, Huzhou, Zhejiang, China
| | - Xiao Chen
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Lu P, Li Y, Dai G, Zhang Y, Shi L, Zhang M, Wang H, Rui Y. HMGB1: a potential new target for tendinopathy treatment. Connect Tissue Res 2023; 64:362-375. [PMID: 37032550 DOI: 10.1080/03008207.2023.2199089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/29/2023] [Indexed: 04/11/2023]
Abstract
Tendinopathy describes a complex pathology of the tendon characterized by abnormalities in the microstructure, composition, and cellularity of the tendon, leading to pain, limitation of activity and reduced function. Nevertheless, the mechanism of tendinopathy has not been fully elucidated, and the treatment of tendinopathy remains a challenge. High mobility group box 1 (HMGB1), a highly conserved and multifaceted nuclear protein, exerts multiple roles and high functional variability and is involved in many biological and pathological processes. In recent years, several studies have suggested that HMGB1 is associated with tendinopathy and may play a key role in the pathogenesis of tendinopathy. Therefore, this review summarizes the expression and distribution of HMGB1 in tendinopathy, focuses on the roles of HMGB1 and HMGB1-based potential mechanisms involved in tendinopathy, and finally summarizes the findings on HMGB1-based therapeutic approaches in tendinopathy, probably providing new insight into the mechanism and further potential therapeutic targets of tendinopathy.
Collapse
Affiliation(s)
- Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yingjuan Li
- School of Medicine, Southeast University, Nanjing, China
- Department of Geriatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Guangchun Dai
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yuanwei Zhang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Liu Shi
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ming Zhang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
11
|
Zhang J, Brown R, Hogan MV, Onishi K, Wang JHC. Metformin improves tendon degeneration by blocking translocation of HMGB1 and suppressing tendon inflammation and senescence in aging mice. J Orthop Res 2023; 41:1162-1176. [PMID: 36262012 PMCID: PMC10113400 DOI: 10.1002/jor.25470] [Citation(s) in RCA: 4] [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: 02/27/2022] [Revised: 08/29/2022] [Accepted: 10/18/2022] [Indexed: 02/04/2023]
Abstract
This study aimed to characterize aging-induced tendinopathy in mouse Achilles tendon and also to assess the treatment effects of metformin (Met) on aging tendon. We showed that compared to young tendon, aging tendon was in an inflammatory and senescent state as shown by increased expression of inflammatory disulfide HMGB1 (dsHMGB1), inflammatory macrophage marker CD68, and senescent cell markers SA-β-gal, p53, and p16. Moreover, aging tendon was degenerated marked by accumulation of proteoglycans and lipids in its interior. However, treatment of aging tendon by intraperitoneal (IP) injection of Met, a specific inhibitor of HMGB1, reduced dsHMGB1 levels, decreased the expression of CD68, SA-β-gal, CCN1, and p16 in vitro and in vivo. Furthermore, Met treatment also increased the number of NS, SSEA-1, and CD73 positive stem cells in culture and improved the tendon structure in aging mouse. These findings of this study indicate that Met exerts anti-inflammatory and anti-senescent effects on aging tendon.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Roshawn Brown
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kentaro Onishi
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
12
|
Malmgaard-Clausen NM, Kjaer M, Dakin SG. Pathological Tendon Histology in Early and Chronic Human Patellar Tendinopathy. TRANSLATIONAL SPORTS MEDICINE 2022; 2022:2799665. [PMID: 38655164 PMCID: PMC11022758 DOI: 10.1155/2022/2799665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 04/26/2024]
Abstract
The present pilot study investigated the extent of histological tissue changes in both chronic tendinopathy and in individuals that display early clinical signs of tendinopathy. The study included 8 individuals of whom 3 were healthy without any tendon symptoms, 2 had early symptoms (1-2 months), and 3 had chronic symptoms (>3 months) from their patellar tendons. Percutaneous needle biopsy samples were obtained from the affected tendon tissue region. Biopsy samples were stained with Haematoxylin & Eosin, and multiplex immunofluorescence staining was performed for markers of inflammation and resolution. Both early and chronic stage patellar tendon biopsy samples from this small patient cohort exhibited expansion of the interfascicular matrix (IFM) and endotenon regions together with increased cellularity and vascularity. These histological observations were moderate in early tendinopathy, whereas they were more pronounced and associated with marked disruption of tissue architecture in chronic tendinopathy. Early stage tendinopathic patellar tendons expressed markers associated with an activated phenotype of fibroblasts (CD90, CD34), macrophages (S100A8), and endothelial cells (ICAM1, VCAM1). These tissues also expressed enzymes implicated in inflammation (PTGS2, 15PGDH) and resolution (ALOX12) and the proresolving receptor ERV1. Immunopositive staining for these markers was predominantly located in the IFM regions. These preliminary findings suggest that mild to moderate structural histological changes including expansion of IFM and endotenon regions are pathological features of early tendinopathy, and support inflammatory and resolving processes are active in early-stage disease. Further investigation of the cellular and molecular basis of early-stage tendinopathy is required to inform therapeutic strategies that prevent the development of irreversible chronic tendon disease.
Collapse
Affiliation(s)
- Nikolaj Moelkjaer Malmgaard-Clausen
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Stephanie G Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Abraham AC, Fang F, Golman M, Oikonomou P, Thomopoulos S. The role of loading in murine models of rotator cuff disease. J Orthop Res 2022; 40:977-986. [PMID: 34081350 PMCID: PMC8639823 DOI: 10.1002/jor.25113] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/07/2021] [Accepted: 05/31/2021] [Indexed: 02/04/2023]
Abstract
Rotator cuff disease pathogenesis is associated with intrinsic (e.g., age, joint laxity, muscle weakness) and extrinsic (e.g., mechanical load, fatigue) factors that lead to chronic degeneration of the cuff tissues. However, etiological studies are difficult to perform in patients due to the long duration of disease onset and progression. Therefore, the purpose of this study was to determine the effects of altered joint loading on the rotator cuff. Mice were subjected to one of three load-dependent rotator cuff tendinopathy models: underuse loading, achieved by injecting botulinum toxin-A into the supraspinatus muscle; overuse loading, achieved using downhill treadmill running; destabilization loading, achieved by surgical excision of the infraspinatus tendon. All models were compared to cage activity animals. Whole joint function was assessed longitudinally using gait analysis. Tissue-scale structure and function were determined using microCT, tensile testing, and histology. The molecular response of the supraspinatus tendon and enthesis was determined by measuring the expression of 84 wound healing-associated genes. Underuse and destabilization altered forepaw weight-bearing, decreased tendon-to-bone attachment strength, decreased mineral density of the humeral epiphysis, and reduced tendon strength. Transcriptional activity of the underuse group returned to baseline levels by 4 weeks, while destabilization had significant upregulation of inflammation, growth factors, and extracellular matrix remodeling genes. Surprisingly, overuse activity caused changes in walking patterns, increased tendon stiffness, and primarily suppressed expression of wound healing-related genes. In summary, the tendinopathy models demonstrated how divergent muscle loading can result in clinically relevant alterations in rotator cuff structure, function, and gene expression.
Collapse
Affiliation(s)
- Adam C. Abraham
- University of Michigan, Department of Orthopaedic Surgery, Biomedical Science Research Building, 109 Zina Pitcher Pl, Ann Arbor, MI 48109, USA,Corresponding author Adam C. Abraham, Ph.D., Research Investigator, University of Michigan, Department of Orthopaedic Surgery, Ann Arbor, MI 48109, USA,
| | - Fei Fang
- Columbia University Irving Medical Center, Department of Orthopaedic Surgery, New York, NY 10032, USA
| | - Mikhail Golman
- Columbia University, Department of Biomedical Engineering, New York, NY 10027
| | | | - Stavros Thomopoulos
- Columbia University Irving Medical Center, Department of Orthopaedic Surgery, New York, NY 10032, USA,Columbia University, Department of Biomedical Engineering, New York, NY 10027
| |
Collapse
|
14
|
Xu X, Ha P, Yen E, Li C, Zheng Z. Small Leucine-Rich Proteoglycans in Tendon Wound Healing. Adv Wound Care (New Rochelle) 2022; 11:202-214. [PMID: 34978952 DOI: 10.1089/wound.2021.0069] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Significance: Tendon injury possesses a high morbidity rate and is difficult to achieve a satisfying prognosis with currently available treatment strategies. Current approaches used for tendon healing always lead to the formation of fibrovascular scar tissue, which significantly compromises the biomechanics of the healed tendon. Moreover, the related functional deficiency deteriorates over time with an increased injury recurrence risk. Small leucine-rich proteoglycans (SLRPs) link and interact with collagen fibrils to regulate tendon structure and biomechanics, which can provide a new and promising method in the field of tendon injury management. Recent Advances: The effect of SLRPs on tendon development has been extensively investigated. SLRP deficiency impairs tendon collagen fibril structure and biomechanic properties, while administration of SLRPs generally benefits tendon wound healing and regains better mechanical properties. Critical Issues: Current knowledge on the role of SLRPs in tendon development and regeneration mostly comes from uninjured knockout mice, and mainly focuses on the morphology description of collagen fibril profile and mechanical properties. Little is known about the regulatory mechanism on the molecular level. Future Directions: This article reviews the current knowledge in this highly translational topic and provides an evidence-based conclusion, thereby encouraging in-depth investigations of SLRPs in tendons and the development of SLRP-based treatments for desired tendon healing.
Collapse
Affiliation(s)
- Xue Xu
- Department of Oral and Maxillofacial Plastic and Traumatic Surgery, Beijing Stomatological Hospital of Capital Medical University, Beijing, People's Republic of China
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Pin Ha
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Emily Yen
- Arcadia High School, Arcadia, California, USA
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
15
|
Russo V, El Khatib M, Prencipe G, Citeroni MR, Faydaver M, Mauro A, Berardinelli P, Cerveró-Varona A, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Stöckl J, Barboni B. Tendon Immune Regeneration: Insights on the Synergetic Role of Stem and Immune Cells during Tendon Regeneration. Cells 2022; 11:434. [PMID: 35159244 PMCID: PMC8834336 DOI: 10.3390/cells11030434] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Tendon disorders represent a very common pathology in today's population, and tendinopathies that account 30% of tendon-related injuries, affect yearly millions of people which in turn cause huge socioeconomic and health repercussions worldwide. Inflammation plays a prominent role in the development of tendon pathologies, and advances in understanding the underlying mechanisms during the inflammatory state have provided additional insights into its potential role in tendon disorders. Different cell compartments, in combination with secreted immune modulators, have shown to control and modulate the inflammatory response during tendinopathies. Stromal compartment represented by tenocytes has shown to display an important role in orchestrating the inflammatory response during tendon injuries due to the interplay they exhibit with the immune-sensing and infiltrating compartments, which belong to resident and recruited immune cells. The use of stem cells or their derived secretomes within the regenerative medicine field might represent synergic new therapeutical approaches that can be used to tune the reaction of immune cells within the damaged tissues. To this end, promising opportunities are headed to the stimulation of macrophages polarization towards anti-inflammatory phenotype together with the recruitment of stem cells, that possess immunomodulatory properties, able to infiltrate within the damaged tissues and improve tendinopathies resolution. Indeed, the comprehension of the interactions between tenocytes or stem cells with the immune cells might considerably modulate the immune reaction solving hence the inflammatory response and preventing fibrotic tissue formation. The purpose of this review is to compare the roles of distinct cell compartments during tendon homeostasis and injury. Furthermore, the role of immune cells in this field, as well as their interactions with stem cells and tenocytes during tendon regeneration, will be discussed to gain insights into new ways for dealing with tendinopathies.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Johannes Stöckl
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
16
|
Lui PPY, Yung PSH. Inflammatory mechanisms linking obesity and tendinopathy. J Orthop Translat 2022; 31:80-90. [PMID: 34976728 PMCID: PMC8666605 DOI: 10.1016/j.jot.2021.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic tendinopathy is a debilitating tendon disorder with disappointing treatment outcomes. This review focuses on the potential roles of chronic low-grade inflammation in promoting tendinopathy in obesity. A systematic literature search was performed to identify all clinical studies supporting the actions of obesity-associated inflammatory mediators in the development of tendinopathy. The mechanisms of obesity-induced chronic inflammation in adipose tissue are firstly reviewed. Common inflammatory mediators potentially linking obesity and the development of tendinopathy, and their association with mechanical overuse, are discussed, along with pre-clinical evidences and a systematic literature search on clinical studies. The potential contribution of local adipose tissues in the promotion of inflammation, pain and tendon degeneration is then discussed. The future research directions are proposed. Translational potential statement Better understanding of the roles of obesity-associated inflammatory mediators on tendons will clarify the pathophysiological drivers of tendinopathy in patients with obesity and identify possible treatment targets. Further studies on the mechanisms of obesity-induced chronic inflammation on tendon are a promising direction for the treatment of tendinopathy.
Collapse
Affiliation(s)
- Pauline Po Yee Lui
- Corresponding author. Room 74037, 5/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, China.
| | | |
Collapse
|
17
|
Zhang X, Wang D, Mak KLK, Tuan RS, Ker DFE. Engineering Musculoskeletal Grafts for Multi-Tissue Unit Repair: Lessons From Developmental Biology and Wound Healing. Front Physiol 2021; 12:691954. [PMID: 34504435 PMCID: PMC8421786 DOI: 10.3389/fphys.2021.691954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022] Open
Abstract
In the musculoskeletal system, bone, tendon, and skeletal muscle integrate and act coordinately as a single multi-tissue unit to facilitate body movement. The development, integration, and maturation of these essential components and their response to injury are vital for conferring efficient locomotion. The highly integrated nature of these components is evident under disease conditions, where rotator cuff tears at the bone-tendon interface have been reported to be associated with distal pathological alterations such as skeletal muscle degeneration and bone loss. To successfully treat musculoskeletal injuries and diseases, it is important to gain deep understanding of the development, integration and maturation of these musculoskeletal tissues along with their interfaces as well as the impact of inflammation on musculoskeletal healing and graft integration. This review highlights the current knowledge of developmental biology and wound healing in the bone-tendon-muscle multi-tissue unit and perspectives of what can be learnt from these biological and pathological processes within the context of musculoskeletal tissue engineering and regenerative medicine. Integrating these knowledge and perspectives can serve as guiding principles to inform the development and engineering of musculoskeletal grafts and other tissue engineering strategies to address challenging musculoskeletal injuries and diseases.
Collapse
Affiliation(s)
- Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - King-Lun Kingston Mak
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
18
|
Tsai SL, Noedl MT, Galloway JL. Bringing tendon biology to heel: Leveraging mechanisms of tendon development, healing, and regeneration to advance therapeutic strategies. Dev Dyn 2021; 250:393-413. [PMID: 33169466 PMCID: PMC8486356 DOI: 10.1002/dvdy.269] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Tendons are specialized matrix-rich connective tissues that transmit forces from muscle to bone and are essential for movement. As tissues that frequently transfer large mechanical loads, tendons are commonly injured in patients of all ages. Following injury, mammalian tendons heal poorly through a slow process that forms disorganized fibrotic scar tissue with inferior biomechanical function. Current treatments are limited and patients can be left with a weaker tendon that is likely to rerupture and an increased chance of developing degenerative conditions. More effective, alternative treatments are needed. However, our current understanding of tendon biology remains limited. Here, we emphasize why expanding our knowledge of tendon development, healing, and regeneration is imperative for advancing tendon regenerative medicine. We provide a comprehensive review of the current mechanisms governing tendon development and healing and further highlight recent work in regenerative tendon models including the neonatal mouse and zebrafish. Importantly, we discuss how present and future discoveries can be applied to both augment current treatments and design novel strategies to treat tendon injuries.
Collapse
Affiliation(s)
- Stephanie L. Tsai
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Marie-Therese Noedl
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Jenna L. Galloway
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
19
|
Ackerman JE, Best KT, Muscat SN, Loiselle AE. Metabolic Regulation of Tendon Inflammation and Healing Following Injury. Curr Rheumatol Rep 2021; 23:15. [PMID: 33569739 DOI: 10.1007/s11926-021-00981-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review seeks to provide an overview of the role of inflammation and metabolism in tendon cell function, tendinopathy, and tendon healing. We have summarized the state of knowledge in both tendon and enthesis. RECENT FINDINGS Recent advances in the field include a substantial improvement in our understanding of tendon cell biology, including the heterogeneity of the tenocyte environment during homeostasis, the diversity of the cellular milieu during in vivo tendon healing, and the effects of inflammation and altered metabolism on tendon cell function in vitro. In addition, the mechanisms by which altered systemic metabolism, such as diabetes, disrupts tendon homeostasis continue to be better understood. A central conclusion of this review is the critical need to better define fundamental cellular and signaling mechanisms of inflammation and metabolism during tendon homeostasis, tendinopathy, and tendon healing in order to identify therapies to enhance or maintain tendon function.
Collapse
Affiliation(s)
- Jessica E Ackerman
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
| | - Katherine T Best
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
| | - Samantha N Muscat
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
| |
Collapse
|
20
|
Rapamycin Treatment of Tendon Stem/Progenitor Cells Reduces Cellular Senescence by Upregulating Autophagy. Stem Cells Int 2021; 2021:6638249. [PMID: 33603790 PMCID: PMC7870298 DOI: 10.1155/2021/6638249] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
The elderly population is prone to tendinopathy due to aging-related tendon changes such as cellular senescence and a decreased ability to modulate inflammation. Aging can render tendon stem/progenitor cells (TSCs) into premature senescence. We investigated the effects of rapamycin, a specific mTOR inhibitor, on the senescence of TSCs. We first showed that after treatment with bleomycin in vitro, rat patellar TSCs (PTSCs) underwent senescence, characterized by morphological alterations, induction of senescence-associated β-galactosidase (SA-β-gal) activity, and an increase in p53, p21, and p62 protein expression. Senescence of PTSCs was also characterized by the elevated expression of MMP-13 and TNF-α genes, both of which are molecular hallmarks of chronic tendinopathy. We then showed that rapamycin treatment was able to reverse the above senescent phenotypes and increase autophagy in the senescent PTSCs. The activation of autophagy and senescence rescue was, at least partly, due to the translocation of HMGB1 from the nucleus to the cytosol that functions as an autophagy promoter. By reducing TSC senescence, rapamycin may be used as a therapeutic to inhibit tendinopathy development in the aging population by promoting autophagy.
Collapse
|
21
|
Millar NL, Silbernagel KG, Thorborg K, Kirwan PD, Galatz LM, Abrams GD, Murrell GAC, McInnes IB, Rodeo SA. Tendinopathy. Nat Rev Dis Primers 2021; 7:1. [PMID: 33414454 DOI: 10.1038/s41572-020-00234-1] [Citation(s) in RCA: 408] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 12/14/2022]
Abstract
Tendinopathy describes a complex multifaceted pathology of the tendon, characterized by pain, decline in function and reduced exercise tolerance. The most common overuse tendinopathies involve the rotator cuff tendon, medial and lateral elbow epicondyles, patellar tendon, gluteal tendons and the Achilles tendon. The prominent histological and molecular features of tendinopathy include disorganization of collagen fibres, an increase in the microvasculature and sensory nerve innervation, dysregulated extracellular matrix homeostasis, increased immune cells and inflammatory mediators, and enhanced cellular apoptosis. Although diagnosis is mostly achieved based on clinical symptoms, in some cases, additional pain-provoking tests and imaging might be necessary. Management consists of different exercise and loading programmes, therapeutic modalities and surgical interventions; however, their effectiveness remains ambiguous. Future research should focus on elucidating the key functional pathways implicated in clinical disease and on improved rehabilitation protocols.
Collapse
Affiliation(s)
- Neal L Millar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | | | - Kristian Thorborg
- Institute of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Paul D Kirwan
- School of Physiotherapy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Leesa M Galatz
- Department of Orthopaedic Surgery, Icahn School of Medicine, Mount Sinai Health System, New York, NY, USA
| | - Geoffrey D Abrams
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
22
|
Mousley JJ, Hill-Buxton LM, Gill SD, McGee SL, Page RS. Polymorphisms and alterations in gene expression associated with rotator cuff tear and healing following surgical repair: a systematic review. J Shoulder Elbow Surg 2021; 30:200-215. [PMID: 32827653 DOI: 10.1016/j.jse.2020.07.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/20/2020] [Accepted: 07/26/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Rotator cuff tears (RCTs) are a common cause of shoulder disability, yet both conservative and surgical treatment strategies can lead to poor results in some patient populations. Enhanced understanding of the genetic processes associated with RCTs can assist in the development of more effective management options and help predict individual responses to surgical treatment. This systematic review analyzes the current literature on the genetic footprint associated with RCTs and interprets these findings to enhance the current understanding of RCT pathogenesis, potential treatment regimens, and prognostic biomarkers of outcomes after surgical repair. METHODS A systematic search of the Embase, PubMed, and Web of Science electronic databases was performed. Medical Subject Headings (MeSH) and Emtree index terms were formulated from the concept terms "rotator cuff tear," "genetics," and "human," and synonyms of these concepts were applied to the Web of Science search. Articles were screened against predefined inclusion and exclusion criteria. Eligible studies compared gene expression patterns and genetic polymorphisms between cases (with RCTs) and controls (without RCTs). Quality assessment was performed with studies being rated as high, moderate, or poor quality. A modified best-evidence synthesis was applied, and studies were determined to be of strong, moderate, or limited evidence. RESULTS The search identified 259 articles. Of these studies, 26 were eligible for review. Two studies were considered poor quality; 15 studies, moderate quality; and 9 studies, high quality. Analysis of these articles found that RCTs were associated with alterations in genes that code for the extracellular matrix, cell apoptosis, immune and inflammatory responses, and growth factor pathways. In particular, there was strong evidence of a significant association between RCTs and the genes MMP3, TNC, and ESRRB. Strong evidence of an association between BMP5 upregulation and successful healing after surgical repair was also found. CONCLUSION This review provides strong evidence of an genetic association with RCTs. The genotype and gene expression patterns detailed within this review can assist in deciphering the biological mechanisms resulting in RCTs, as well as predicting an individual's response to surgical repair. Future research could investigate whether manipulating these genes-or their associated signaling pathways-could assist in RCT healing and whether genetic biomarkers could be used clinically to predict patient outcomes after surgical repair of RCTs.
Collapse
Affiliation(s)
| | - Leaha-Marie Hill-Buxton
- Barwon Centre for Orthopaedic Research & Education (B-CORE), St John of God Hospital and Barwon Health, Geelong, VIC, Australia
| | - Stephen D Gill
- School of Medicine, Deakin University, Geelong, VIC, Australia; Barwon Centre for Orthopaedic Research & Education (B-CORE), St John of God Hospital and Barwon Health, Geelong, VIC, Australia
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard S Page
- School of Medicine, Deakin University, Geelong, VIC, Australia; Barwon Centre for Orthopaedic Research & Education (B-CORE), St John of God Hospital and Barwon Health, Geelong, VIC, Australia
| |
Collapse
|
23
|
Zhang J, Li F, Nie D, Onishi K, Hogan MV, Wang JHC. Effect of Metformin on Development of Tendinopathy Due to Mechanical Overloading in an Animal Model. Foot Ankle Int 2020; 41:1455-1465. [PMID: 33180557 PMCID: PMC7736509 DOI: 10.1177/1071100720966318] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Tendinopathy is a debilitating tendon disorder that affects millions of Americans and costs billions of health care dollars every year. High mobility group box 1 (HMGB1), a known tissue damage signaling molecule, has been identified as a mediator in the development of tendinopathy due to mechanical overloading of tendons in mice. Metformin (Met), a drug approved by the Food and Drug Administration used for the treatment of type 2 diabetes, specifically inhibits HMGB1. This study tested the hypothesis that Met would prevent mechanical overloading-induced tendinopathy in a mouse model of tendinopathy created by intensive treadmill running (ITR). METHODS C57BL/6J mice (female, 3 months old) were equally separated into 4 groups and treated for 24 weeks as follows: group 1 had cage control activities, group 2 received a single intraperitoneal injection of Met (50 mg/kg body weight) daily, group 3 underwent ITR to induce tendinopathy, and group 4 received daily Met injection along with ITR to inhibit HMGB1. Tendinopathic changes were assessed in Achilles tendons of all mice using histology, immunohistochemistry, and enzyme-linked immunosorbent assays. RESULTS ITR induced HMGB1 release into the tendon matrix and developed characteristics of tendinopathy as evidenced by the expression of macrophage marker CD68, proinflammatory molecules (COX-2, PGE2), cell morphological changes from normal elongated cells to round cells, high levels of expression of chondrogenic markers (SOX-9, collagen type II), and accumulation of proteoglycans in tendinopathic tendons. Daily injection of Met inhibited HMGB1 release and decreased these degenerative changes in ITR tendons. CONCLUSIONS Inhibition of HMGB1 by injections of Met prevented tendinopathy development due to mechanical overloading in the Achilles tendon in mice. CLINICAL RELEVANCE Met may be able to be repurposed as a therapeutic option for preventing the development of tendinopathy in high-risk patients.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, 15213
| | - Feng Li
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, 15213
| | - Daibang Nie
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, 15213,Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Kentaro Onishi
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213
| | - MaCalus V Hogan
- Departments of Orthopaedic Surgery, Bioengineering, and Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213
| | - James H-C. Wang
- Departments of Orthopaedic Surgery, Bioengineering, and Physical Medicine and Rehabilitation University of Pittsburgh, Pittsburgh, 15213
| |
Collapse
|
24
|
Zhang Y, Deng XH, Lebaschi AH, Wada S, Carballo CB, Croen B, Ying L, Rodeo SA. Expression of alarmins in a murine rotator cuff tendinopathy model. J Orthop Res 2020; 38:2513-2520. [PMID: 32285963 DOI: 10.1002/jor.24690] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/11/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023]
Abstract
The aim of this study was to investigate the presence of alarmins in a novel murine rotator cuff tendinopathy model. Alarmins have been described as essential early activators of an immune response to tissue damage. Subacromial impingement was induced in both shoulders of 37 male C57Bl/6 mice by placement of a small metal clip in the subacromial space. Animals were allocated to different time points up to 6 weeks. The morphology and cellularity of the supraspinatus tendon were evaluated by hematoxylin-eosin staining, alcian blue, and picrosirius red. The expression and localization of alarmins interleukin-33 (IL-33), c (HMGB1), hypoxia-inducible factor-1 subunit α (HIF1α), and S100A9 were evaluated by immunohistochemical staining and quantitative polymerase chain reaction. The percentage of positively stained cells with HMGB1 and IL-33 was significantly increased in the impingement group at 1w, 4w, and 6w. HIF1α staining was higher in the impingement group at 1w and 6w compared with the control group. HMGB1 gene expression was higher in the 5d impingement group and 6w impingement group. The gene expression of HIF1α was upregulated at all-time points in the impingement group (5d, 2w, 4w, and 6w). The expression of the S100A9 gene was also upregulated in the 5d impingement group. This is the first study to demonstrate the involvement of alarmins in the early phase of tendinopathy using a reproducible animal model. Alarmins may play an important role in the early phases of the development of tendinopathy They may represent potential therapeutic targets for treatment of tendinopathy.
Collapse
Affiliation(s)
- Ying Zhang
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York.,School of Public Health, Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiang-Hua Deng
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Amir H Lebaschi
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Susumu Wada
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Camila B Carballo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Brett Croen
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Liang Ying
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Scott A Rodeo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| |
Collapse
|
25
|
Crowe LAN, Garcia Melchor E, Murrell GAC, McInnes IB, Akbar M, Millar NL. Stromal “activation” markers do not confer pathogenic activity in tendinopathy. TRANSLATIONAL SPORTS MEDICINE 2020. [DOI: 10.1002/tsm2.204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Lindsay A. N. Crowe
- Institute of Infection, Immunity and Inflammation College of Medicine, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Emma Garcia Melchor
- Institute of Infection, Immunity and Inflammation College of Medicine, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - George A. C. Murrell
- Orthopaedic Research Institute St George Hospital Campus University of New South Wales Sydney NSW Australia
| | - Iain B. McInnes
- Institute of Infection, Immunity and Inflammation College of Medicine, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Moeed Akbar
- Institute of Infection, Immunity and Inflammation College of Medicine, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Neal L. Millar
- Institute of Infection, Immunity and Inflammation College of Medicine, Veterinary and Life Sciences University of Glasgow Glasgow UK
| |
Collapse
|
26
|
Cho DS, Schmitt RE, Dasgupta A, Ducharme AM, Doles JD. Single-cell deconstruction of post-sepsis skeletal muscle and adipose tissue microenvironments. J Cachexia Sarcopenia Muscle 2020; 11:1351-1363. [PMID: 32643301 PMCID: PMC7567136 DOI: 10.1002/jcsm.12596] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 04/22/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Persistent loss of skeletal muscle mass and function as well as altered fat metabolism are frequently observed in severe sepsis survivors. Studies examining sepsis-associated tissue dysfunction from the perspective of the tissue microenvironment are scarce. In this study, we comprehensively assessed transcriptional changes in muscle and fat at single-cell resolution following experimental sepsis induction. METHODS Skeletal muscle and visceral white adipose tissue from control mice or mice 1 day or 1 month following faecal slurry-induced sepsis were used. Single cells were mechanically and enzymatically prepared from whole tissue, and viable cells were further isolated by fluorescence activated cell sorting. Droplet-based single-cell RNA-sequencing (scRNA-seq; 10× Genomics) was used to generate single-cell gene expression profiles of thousands of muscle and fat-resident cells. Bioinformatics analyses were performed to identify and compare individual cell populations in both tissues. RESULTS In skeletal muscle, scRNA-seq analysis classified 1438 single cells into myocytes, endothelial cells, fibroblasts, mesenchymal stem cells, macrophages, neutrophils, T-cells, B-cells, and dendritic cells. In adipose tissue, scRNA-seq analysis classified 2281 single cells into adipose stem cells, preadipocytes, endothelial cells, fibroblasts, macrophages, dendritic cells, B-cells, T-cells, NK cells, and gamma delta T-cells. One day post-sepsis, the proportion of most non-immune cell populations was decreased, while immune cell populations, particularly neutrophils and macrophages, were highly enriched. Proportional changes of endothelial cells, neutrophils, and macrophages were validated using faecal slurry and cecal ligation and puncture models. At 1 month post-sepsis, we observed persistent enrichment/depletion of cell populations and further uncovered a cell-type and tissue-specific ability to return to a baseline transcriptomic state. Differential gene expression analyses revealed key genes and pathways altered in post-sepsis muscle and fat and highlighted the engagement of infection/inflammation and tissue damage signalling. Finally, regulator analysis identified gonadotropin-releasing hormone and Bay 11-7082 as targets/compounds that we show can reduce sepsis-associated loss of lean or fat mass. CONCLUSIONS These data demonstrate persistent post-sepsis muscle and adipose tissue disruption at the single-cell level and highlight opportunities to combat long-term post-sepsis tissue wasting using bioinformatics-guided therapeutic interventions.
Collapse
Affiliation(s)
- Dong Seong Cho
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| | - Rebecca E. Schmitt
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| | - Aneesha Dasgupta
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| | | | - Jason D. Doles
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| |
Collapse
|
27
|
Zhang C, Gu X, Zhao G, Wang W, Shao J, Zhu J, Yuan T, Sun J, Nie D, Zhou Y. Extracellular HMGB-1 activates inflammatory signaling in tendon cells and tissues. Ther Adv Chronic Dis 2020; 11:2040622320956429. [PMID: 32963751 PMCID: PMC7488923 DOI: 10.1177/2040622320956429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/13/2020] [Indexed: 01/15/2023] Open
Abstract
Background: Increasing evidence indicates that secretion of high mobility group box 1 protein (HMGB-1) is functionally associated with tendinopathy development. However, the underlying effect and mechanism of extracellular HMGB-1 on tendon cells are unclear. Methods: We tested the effect of exogenous HMGB-1 on cell growth, migration, and inflammatory signaling responses with isolated rat Achilles tendon cells. Also, we studied the role of extracellular HMGB-1, when administrated alone or in combination with mechanical overloading induced by intensive treadmill running (ITR), in stimulating inflammatory effects in tendon tissues. Results: By using in vitro and in vivo models, we show for the first time that exogenous HMGB-1 dose-dependently induces inflammatory reactions in tendon cells and tendon tissue. Extracellular HMGB-1 promoted redistribution of HMGB-1 from the nucleus to the cytoplasm, and activated canonical nuclear factor kappa B (NF-κB) signaling and mitogen-activated protein kinase (MAPK) signaling. Short-term administration of HMGB-1 induced hyper-cellularity of rat Achilles tendon tissues, accompanied with enhanced immune cell infiltration. Additional ITR to HMGB-1 treatment worsens these responses, and application of HMGB-1 specific inhibitor glycyrrhizin (GL) completely abolishes such inflammatory effects in tendon tissues. Conclusion: Collectively, these results confirm that HMGB-1 plays key roles in the induction of tendinopathy. Our findings improve the understanding of the molecular and cellular mechanisms during tendinopathy development, and provide essential information for potential targeted treatments of tendinopathy.
Collapse
Affiliation(s)
- Chuanxin Zhang
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xinfeng Gu
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China Department of Bone and Joint, Shuguang Hospital Affiliated to Shanghai University of Tradition Chinese Medicine, Shanghai, China
| | - Guangyi Zhao
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute Pittsburgh, PA, USA
| | - Wang Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Jiahua Shao
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jun Zhu
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ting Yuan
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiuyi Sun
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China Department of Orthropaedics, Navy Medical Center of PLA, Shanghai, China
| | - Daibang Nie
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yiqin Zhou
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
28
|
Gacaferi H, Mimpen JY, Baldwin MJ, Snelling SJB, Nelissen RGHH, Carr AJ, Dakin SG. The potential roles of high mobility group box 1 (HMGB1) in musculoskeletal disease: A systematic review. TRANSLATIONAL SPORTS MEDICINE 2020. [DOI: 10.1002/tsm2.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Hamez Gacaferi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
- Department of Orthopaedics Leiden University Medical Centre Leiden The Netherlands
| | - Jolet Y. Mimpen
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Mathew J. Baldwin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Sarah J. B. Snelling
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | | | - Andrew J. Carr
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Stephanie G. Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| |
Collapse
|
29
|
Tendon and ligament mechanical loading in the pathogenesis of inflammatory arthritis. Nat Rev Rheumatol 2020; 16:193-207. [PMID: 32080619 DOI: 10.1038/s41584-019-0364-x] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Mechanical loading is an important factor in musculoskeletal health and disease. Tendons and ligaments require physiological levels of mechanical loading to develop and maintain their tissue architecture, a process that is achieved at the cellular level through mechanotransduction-mediated fine tuning of the extracellular matrix by tendon and ligament stromal cells. Pathological levels of force represent a biological (mechanical) stress that elicits an immune system-mediated tissue repair pathway in tendons and ligaments. The biomechanics and mechanobiology of tendons and ligaments form the basis for understanding how such tissues sense and respond to mechanical force, and the anatomical extent of several mechanical stress-related disorders in tendons and ligaments overlaps with that of chronic inflammatory arthritis in joints. The role of mechanical stress in 'overuse' injuries, such as tendinopathy, has long been known, but mechanical stress is now also emerging as a possible trigger for some forms of chronic inflammatory arthritis, including spondyloarthritis and rheumatoid arthritis. Thus, seemingly diverse diseases of the musculoskeletal system might have similar mechanisms of immunopathogenesis owing to conserved responses to mechanical stress.
Collapse
|
30
|
Kumar D, Lee B, Puan KJ, Lee W, Luis BS, Yusof N, Andiappan AK, Del Rosario R, Poschmann J, Kumar P, DeLibero G, Singhal A, Prabhakar S, De Yun W, Poidinger M, Rötzschke O. Resistin expression in human monocytes is controlled by two linked promoter SNPs mediating NFKB p50/p50 binding and C-methylation. Sci Rep 2019; 9:15245. [PMID: 31645609 PMCID: PMC6811637 DOI: 10.1038/s41598-019-51592-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Resistin is a key cytokine associated with metabolic and inflammatory diseases. Especially in East Asian populations, the expression levels are strongly influenced by genetic polymorphisms. Mechanisms and functional implications of this genetic control are still unknown. By employing reporter assays, EMSA, inhibition studies, bisulphite sequencing, ChIP-Seq and gene-editing we show that the p50/p50 homodimer known to act as repressor for a number of pro-inflammatory genes plays a central role in the genetic regulation of resistin in monocytes along with promoter methylation. In the common RETN haplotype p50/p50 constitutively dampens the expression by binding to the promoter. In an Asian haplotype variant however this interaction is disrupted by the A allele of rs3219175. The SNP is in very close linkage to rs34861192, a CpG SNP, located 280 bp upstream which provides an allele-specific C-methylation site. rs34861192 is located in a 100 bp region found to be methylated in the common but not in the Asian haplotype, resulting in the latter having a higher basal expression, which also associates with elevated histone acetylation (H3K27ac). Genotype associations within cohort data of 200 East Asian individuals revealed significant associations between this haplotype and the plasma levels of factors such as TGF-b, S100B, sRAGE and IL-8 as well as with myeloid DC counts. Thus, the common RETN haplotype is tightly regulated by the epigenetic mechanism linked to p50/p50-binding. This control is lost in the Asian haplotype, which may have evolved to balance the antagonistic RETN effects on pathogen protection vs. metabolic and inflammatory disease induction.
Collapse
Affiliation(s)
- Dilip Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| | - Bernett Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Kia Joo Puan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Wendy Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Boris San Luis
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Nurhashikin Yusof
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Ricardo Del Rosario
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA, 02142, USA
| | - Jeremie Poschmann
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Centre de Recherche en Transplantation et Immunologie, Université de Nantes, Nantes, France
| | - Pavanish Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Gennaro DeLibero
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Amit Singhal
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore
| | - Wang De Yun
- Department of Otolaryngology, National University of Singapore, Singapore, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Olaf Rötzschke
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| |
Collapse
|
31
|
Zhao G, Zhang J, Nie D, Zhou Y, Li F, Onishi K, Billiar T, Wang JHC. HMGB1 mediates the development of tendinopathy due to mechanical overloading. PLoS One 2019; 14:e0222369. [PMID: 31560698 PMCID: PMC6764662 DOI: 10.1371/journal.pone.0222369] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/16/2019] [Indexed: 11/30/2022] Open
Abstract
Mechanical overloading is a major cause of tendinopathy, but the underlying pathogenesis of tendinopathy is unclear. Here we report that high mobility group box1 (HMGB1) is released to the tendon extracellular matrix and initiates an inflammatory cascade in response to mechanical overloading in a mouse model. Moreover, administration of glycyrrhizin (GL), a naturally occurring triterpene and a specific inhibitor of HMGB1, inhibits the tendon’s inflammatory reactions. Also, while prolonged mechanical overloading in the form of long-term intensive treadmill running induces Achilles tendinopathy in mice, administration of GL completely blocks the tendinopathy development. Additionally, mechanical overloading of tendon cells in vitro induces HMGB1 release to the extracellular milieu, thereby eliciting inflammatory and catabolic responses as marked by increased production of prostaglandin E2 (PGE2) and matrix metalloproteinase-3 (MMP-3) in tendon cells. Application of GL abolishes the cellular inflammatory/catabolic responses. Collectively, these findings point to HMGB1 as a key molecule that is responsible for the induction of tendinopathy due to mechanical overloading placed on the tendon.
Collapse
Affiliation(s)
- Guangyi Zhao
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Daibang Nie
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yiqin Zhou
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Joint Surgery and Sports Medicine Department, Shanghai Changzheng Hospital, Second Military Medical University, Huangpu, Shanghai, China
| | - Feng Li
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kentaro Onishi
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Timothy Billiar
- Department of Surgery, University of Pittsburgh, Pennsylvania, United States of America
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
32
|
Hansson E, Skiöldebrand E. Low-grade inflammation causes gap junction-coupled cell dysfunction throughout the body, which can lead to the spread of systemic inflammation. Scand J Pain 2019; 19:639-649. [PMID: 31251727 DOI: 10.1515/sjpain-2019-0061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/21/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Gap junction-coupled cells form networks in different organs in the body. These networks can be affected by inflammatory stimuli and become dysregulated. Cell signaling is also changed through connexin-linked gap junctions. This alteration affects the surrounding cells and extracellular matrix in organs. These changes can cause the spread of inflammatory substances, thus affecting other network-linked cells in other organs in the body, which can give rise to systemic inflammation, which in turn can lead to pain that can turn into chronic. METHODS This is a review based on literature search and our own research data of inflammatory stimuli that can affect different organs and particularly gap-junction-coupled cells throughout the body. CONCLUSIONS A remaining question is which cell type or tissue is first affected by inflammatory stimuli. Can endotoxin exposure through the air, water and body start the process and are mast cells the first target cells that have the capacity to alter the physiological status of gap junction-coupled cells, thereby causing breakdown of different barrier systems? IMPLICATIONS Is it possible to address the right cellular and biochemical parameters and restore inflammatory systems to a normal physiological level by therapeutic strategies?
Collapse
Affiliation(s)
- Elisabeth Hansson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 3rd Floor, SE 413 45 Gothenburg, Sweden, Phone: +46-31-786 3363
| | - Eva Skiöldebrand
- Section of Pathology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden.,Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
33
|
Serum levels of the high-mobility group box 1 protein (HMGB1) in children with type 1 diabetes mellitus: case-control study. Cent Eur J Immunol 2019; 44:33-37. [PMID: 31114434 PMCID: PMC6526583 DOI: 10.5114/ceji.2019.84012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 03/15/2018] [Indexed: 01/13/2023] Open
Abstract
Introduction The involvement of the high-mobility group box 1 protein (HMGB1) in various autoimmune and inflammatory diseases has been documented; however, the role of this proinflammatory molecule in children with diabetes type 1 (T1DM) has not been addressed. The aim of this case-control study is to compare the serum level of HMGB1 in children with newly diagnosed T1DM (group 1) and a control group composed of healthy children. Material and methods This case-control study included 136 children: group 1 (n = 96) and a control group (n = 40). Measurements were taken from serum for the following: HMGB1, white blood cell count, C-reactive protein, glucose, haemoglobin A1C, and β-cell autoantibodies (GADA-65, IA-2, ICA). HMGB1 was determined using enzyme-linked immunosorbent assay on a Labsystems iEMS Reader MF analyser (Labsystems Diagnostics Oy, Helsinki, Finland). Results The level (median and interquartile range) of HMGB1 was statistically higher (p < 0.001) in children with T1DM: 8.7 (5.0-9.8) µg/l, in comparison with the control group: 1.0 (0.6-1.4) µg/l. No correlation was found between HMGB1 and HbA1c in group 1, or between HMGB1 and BMI. A statistically higher percentage of positive children for autoantibodies were present in group 1 compared to the control group (p ≤ 0.001). HMGB1 serum levels were also tested and the presence of autoantibodies, and none of those antibodies correlated with the level of HMGB1. Conclusions The higher level of HMGB1 in children with T1DM, compared to the control group, indicates that this proinflammatory molecule is a good candidate marker of inflammation in children with T1DM.
Collapse
|
34
|
Crowe LAN, McLean M, Kitson SM, Melchor EG, Patommel K, Cao HM, Reilly JH, Leach WJ, Rooney BP, Spencer SJ, Mullen M, Chambers M, Murrell GAC, McInnes IB, Akbar M, Millar NL. S100A8 & S100A9: Alarmin mediated inflammation in tendinopathy. Sci Rep 2019; 9:1463. [PMID: 30728384 PMCID: PMC6365574 DOI: 10.1038/s41598-018-37684-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/12/2018] [Indexed: 11/11/2022] Open
Abstract
Alarmins S100A8 and S100A9 are endogenous molecules released in response to environmental triggers and cellular damage. They are constitutively expressed in immune cells such as monocytes and neutrophils and their expression is upregulated under inflammatory conditions. The molecular mechanisms that regulate inflammatory pathways in tendinopathy are largely unknown therefore identifying early immune effectors is essential to understanding the pathology. Based on our previous investigations highlighting tendinopathy as an alarmin mediated pathology we sought evidence of S100A8 & A9 expression in a human model of tendinopathy and thereafter, to explore mechanisms whereby S100 proteins may regulate release of inflammatory mediators and matrix synthesis in human tenocytes. Immunohistochemistry and quantitative RT-PCR showed S100A8 & A9 expression was significantly upregulated in tendinopathic tissue compared with control. Furthermore, treating primary human tenocytes with exogenous S100A8 & A9 significantly increased protein release of IL-6, IL-8, CCL2, CCL20 and CXCL10; however, no alterations in genes associated with matrix remodelling were observed at a transcript level. We propose S100A8 & A9 participate in early pathology by modulating the stromal microenvironment and influencing the inflammatory profile observed in tendinopathy. S100A8 and S100A9 may participate in a positive feedback mechanism involving enhanced leukocyte recruitment and release of pro-inflammatory cytokines from tenocytes that perpetuates the inflammatory response within the tendon in the early stages of disease.
Collapse
Affiliation(s)
- Lindsay A N Crowe
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Michael McLean
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Susan M Kitson
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Emma Garcia Melchor
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Katharina Patommel
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Hai Man Cao
- Orthopaedic Research Institute, Department of Orthopaedic Surgery, St George Hospital Campus, University of New South Wales, New South Wales, Australia
| | - James H Reilly
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - William J Leach
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Brain P Rooney
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Simon J Spencer
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Michael Mullen
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - Max Chambers
- Department of Orthopaedic Surgery, Queen Elizabeth University Hospital Glasgow, Glasgow, Scotland, UK
| | - George A C Murrell
- Orthopaedic Research Institute, Department of Orthopaedic Surgery, St George Hospital Campus, University of New South Wales, New South Wales, Australia
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Moeed Akbar
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK
| | - Neal L Millar
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences University of Glasgow, Glasgow, Scotland, UK.
| |
Collapse
|
35
|
Sikes KJ, Li J, Shen Q, Gao SG, Sandy JD, Plaas A, Wang VM. TGF-b1 or hypoxia enhance glucose metabolism and lactate production via HIF1A signaling in tendon cells. Connect Tissue Res 2018; 59:458-471. [PMID: 29447016 PMCID: PMC6175639 DOI: 10.1080/03008207.2018.1439483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
UNLABELLED Purpose/Aim of the study: Healthy tendons are maintained in homeostasis through controlled usage of glucose for energy and redox equilibrium. Tendon cell stress imposed by overuse injury or vascular insufficiency is accompanied by activation of wound healing pathways which facilitate an adaptive response and the restoration of homeostasis. To understand this response at the gene expression level we have studied the in vivo effects of injected TGF-β1 in a murine model of tendinopathy, as well as treatment of murine tendon explants with either TGF-β1 or hypoxia in vitro. METHODS AND RESULTS We provide evidence (from expression patterns and immunohistochemistry) that both in vivo and in vitro, the stress response in tendon cells may be metabolically controlled in part by glycolytic reprogramming. A major feature of the response to TGF-β1 or hypoxia is activation of the Warburg pathway which generates lactate from glucose under normoxia and thereby inhibits mitochondrial energy production. CONCLUSIONS We discuss the likely outcome of this major metabolic shift in terms of the potential benefits and damage to tendon and suggest how incorporation of this metabolic response into our understanding of initiation and progression of tendinopathies may offer new opportunities for diagnosis and the monitoring of therapies.
Collapse
Affiliation(s)
- Katie J Sikes
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612,Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, Chicago, IL 60607
| | - Jun Li
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612
| | - Quan Shen
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612,Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China 410008
| | - Shu-Guang Gao
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612,Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China 410008
| | - John D Sandy
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612
| | - Anna Plaas
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612
| | - Vincent M Wang
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 339 Kelly Hall, 325 Stanger Street MC 0298, Blacksburg, VA, 24061,Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, Chicago, IL 60607
| |
Collapse
|
36
|
Association of Inflammatory Responses and ECM Disorganization with HMGB1 Upregulation and NLRP3 Inflammasome Activation in the Injured Rotator Cuff Tendon. Sci Rep 2018; 8:8918. [PMID: 29891998 PMCID: PMC5995925 DOI: 10.1038/s41598-018-27250-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/31/2018] [Indexed: 12/25/2022] Open
Abstract
Inflammation and extracellular matrix (ECM) disorganization following the rotator cuff tendon injuries (RCTI) delay the repair and healing process and the molecular mechanisms underlying RCTI pathology are largely unknown. Here, we examined the role of HMGB1 and NLRP3 inflammasome pathway in the inflammation and ECM disorganization in RCTI. This hypothesis was tested in a tenotomy-RCTI rat model by transecting the RC tendon from the humerus. H&E and pentachrome staining revealed significant changes in the morphology, architecture and ECM organization in RC tendon tissues following RCTI when compared with contralateral control. Severity of the injury was high in the first two weeks with improvement in 3–4 weeks following RCTI, and this correlated with the healing response. The expression of proteins associated with increased HMGB-1 and upregulation of NLRP3 inflammasome pathway, TLR4, TLR2, TREM-1, RAGE, ASC, Caspase-1, and IL-1β, in the first two weeks following RCTI followed by decline in 3–4 weeks. These results suggest the association of inflammatory responses and ECM disorganization with HMGB1 upregulation and NLRP3 inflammasome activation in the RC tendons and could provide novel target(s) for development of better therapeutic strategies in the management of RCTI.
Collapse
|
37
|
Tang C, Chen Y, Huang J, Zhao K, Chen X, Yin Z, Heng BC, Chen W, Shen W. The roles of inflammatory mediators and immunocytes in tendinopathy. J Orthop Translat 2018; 14:23-33. [PMID: 30035030 PMCID: PMC6034108 DOI: 10.1016/j.jot.2018.03.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 02/06/2023] Open
Abstract
Tendinopathy is a common disease of the musculoskeletal system, particularly in athletes and sports amateurs. In this review, we will present evidence for the critical role of inflammatory mediators and immunocytes in the pathogenesis of tendinopathy and the efficacy of current antiinflammatory therapy and regenerative medicine in the clinic. We hereby propose a hypothesis that in addition to pulling force there may be compressive forces being exerted on the tendon during physical activities, which may initiate the onset of tendinopathy. We performed literature searches on MEDLINE from the inception of this review to February 2018. No language restrictions were imposed. The search terms were as follows: ("Tendinopathy"[Mesh] OR "Tendon Injuries"[Mesh] OR "Tendinitis"[Mesh] OR "Tendon"[Mesh]) AND (Inflammation OR "Inflammatory mediator*" OR Immunocyte*) OR ("anti inflammatory*" OR "regenerative medicine"). Inclusion criteria included articles that were original and reliable, with the main contents being highly relevant to our review. Exclusion criteria included articles that were not available online or have not been published. We scanned the abstract of these articles first. This was then followed by a careful screening of the articles which might be suitable for our review. Finally, 84 articles were selected as references. This review article is written in the narrative form. The translational potential of this article: Understanding the mechanisms of inflammation and existing antiinflammatory and regenerative therapies is key to the development of therapeutic strategies in tendinopathy.
Collapse
Affiliation(s)
- Chenqi Tang
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang 310009, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang 310000, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Zhejiang 310000, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang 310000, China.,China Orthopaedic Regenerative Medicine (CORMed), Chinese Medical Association, Hangzhou, China
| | - Yangwu Chen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang 310009, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang 310000, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Zhejiang 310000, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang 310000, China.,China Orthopaedic Regenerative Medicine (CORMed), Chinese Medical Association, Hangzhou, China
| | - Jiayun Huang
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang 310009, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang 310000, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Zhejiang 310000, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang 310000, China.,China Orthopaedic Regenerative Medicine (CORMed), Chinese Medical Association, Hangzhou, China
| | - Kun Zhao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang 310000, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang 310000, China.,China Orthopaedic Regenerative Medicine (CORMed), Chinese Medical Association, Hangzhou, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang 310000, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang 310000, China.,China Orthopaedic Regenerative Medicine (CORMed), Chinese Medical Association, Hangzhou, China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang 310000, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang 310000, China
| | - Boon Chin Heng
- Faculty of Dentistry, Department of Endodontology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Weishan Chen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang 310009, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Zhejiang 310000, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang 310009, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang 310000, China.,Orthopaedics Research Institute, Zhejiang Univerisity, Zhejiang 310000, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang 310000, China.,China Orthopaedic Regenerative Medicine (CORMed), Chinese Medical Association, Hangzhou, China
| |
Collapse
|