1
|
Mertz M, Hetzel T, Alex K, Braun K, Camenzind S, Dodaro R, Jörgensen S, Linder E, Capas-Peneda S, Reihs EI, Tiwari V, Todorović Z, Kahrass H, Selter F. Interdisciplinary Animal Research Ethics-Challenges, Opportunities, and Perspectives. Animals (Basel) 2024; 14:2896. [PMID: 39409845 PMCID: PMC11475729 DOI: 10.3390/ani14192896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Can nonhuman animals be used for the benefit of humans in a scientifically and morally justified manner and, if yes, how? Based on our own experiences as scholars from various academic backgrounds, we argue that this question can only be answered as an interdisciplinary and international endeavor, considering insights from research ethics and animal ethics as well as scientific and legal aspects. The aim of this article is to contribute to the foundation of the emerging field of animal research ethics. In doing so, we describe the following seven phases of animal research experiments: ethical, legal and social presumptions (phase 0), planning (phase I), review (phase II), conduct of experiments (phase III), publication/dissemination (phase IV), further exploitation of results (phase V), and evaluation (phase VI). In total, 20 key ethical, legal, and practical challenges that an ethical framework for the use of animals in research needs to address are identified and analyzed. Finally, we characterize the following four meta-challenges and opportunities associated with animal research ethics as a field: (1) moral pluralism, (2) the integration of views and positions outside the laboratory, (3) international plurality of conduct, standards, and legal norms, and (4) interdisciplinary education.
Collapse
Affiliation(s)
- Marcel Mertz
- Institute for Ethics, History and Philosophy of Medicine, Hannover Medical School, 30625 Hannover, Germany (H.K.); (F.S.)
| | - Tatiana Hetzel
- Institute for Ethics, History and Philosophy of Medicine, Hannover Medical School, 30625 Hannover, Germany (H.K.); (F.S.)
| | - Karla Alex
- Section Translational Medical Ethics, National Center for Tumor Diseases (NCT) Heidelberg, Department of Medical Oncology, Heidelberg University Hospital, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany;
| | - Katharina Braun
- Department of Law, Freie Universität Berlin, 14195 Berlin, Germany;
| | - Samuel Camenzind
- Department of Philosophy, University of Vienna, 1010 Vienna, Austria;
| | - Rita Dodaro
- Department of Philosophy and Humanities, Freie Universität Berlin, 14195 Berlin, Germany
- Humanities Department, Università della Calabria, 87036 Rende, CS, Italy
| | - Svea Jörgensen
- Department of Applied Animal Science and Welfare, Swedish University of Agricultural Sciences, 750 07 Uppsala, Sweden;
| | - Erich Linder
- Messerli Research Institute, University of Veterinary Medicine, 1210 Vienna, Austria;
- Vienna Doctoral School of Philosophy, Department of Philosophy, University of Vienna, 1010 Vienna, Austria
| | - Sara Capas-Peneda
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Eva Ingeborg Reihs
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| | - Vini Tiwari
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany;
- German Center for Neurodegenerative Diseases, 81377 Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, 82152 Planegg, Germany
| | - Zorana Todorović
- Department of Philosophy, University of Belgrade, 11000 Belgrade, Serbia;
- Center for the Study of Bioethics, 11000 Belgrade, Serbia
| | - Hannes Kahrass
- Institute for Ethics, History and Philosophy of Medicine, Hannover Medical School, 30625 Hannover, Germany (H.K.); (F.S.)
| | - Felicitas Selter
- Institute for Ethics, History and Philosophy of Medicine, Hannover Medical School, 30625 Hannover, Germany (H.K.); (F.S.)
| |
Collapse
|
2
|
Gupta A, Mishra SK, Lascelles BDX. Emerging evidence of artemin/GFRα3 signaling in musculoskeletal pain. Osteoarthritis Cartilage 2024:S1063-4584(24)01404-3. [PMID: 39374825 DOI: 10.1016/j.joca.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
Chronic musculoskeletal pain is highly prevalent and poses a significant personal, societal, and economic burden. Management of chronic musculoskeletal pain remains a challenge. Long-term use of common analgesic medications such as nonsteroidal anti-inflammatory drugs and opioids is associated with adverse events, and in the case of opioids, drug addiction. Additionally, many individuals do not experience sufficient pain relief with these therapeutic approaches. Thus, there is an urgent need to develop clinically efficacious and safe therapeutics for musculoskeletal pain. Recent advances in our understanding of musculoskeletal pain neurobiology have helped identify the role of neurotrophic factors, specifically, the glial cell line-derived neurotrophic factor family of ligands (GFL) and their associated signaling pathways. This review outlines our current understanding of the GFL signaling systems, discusses their role in inflammatory and chronic musculoskeletal pain and sensitivity, and comments on the analgesic therapeutic potential of targeting the GFL signaling system.
Collapse
Affiliation(s)
- Ankita Gupta
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Santosh K Mishra
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - B Duncan X Lascelles
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, USA; Thurston Arthritis Center, UNC School of Medicine, Chapel Hill, NC, USA; Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, NC, USA.
| |
Collapse
|
3
|
Oláh T, Cucchiarini M, Madry H. Temporal progression of subchondral bone alterations in OA models involving induction of compromised meniscus integrity in mice and rats: A scoping review. Osteoarthritis Cartilage 2024; 32:1220-1234. [PMID: 38876436 DOI: 10.1016/j.joca.2024.06.002] [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: 01/17/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVE To categorize the temporal progression of subchondral bone alterations induced by compromising meniscus integrity in mouse and rat models of knee osteoarthritis (OA). METHOD Scoping review of investigations reporting subchondral bone changes with appropriate negative controls in the different mouse and rat models of OA induced by compromising meniscus integrity. RESULTS The available literature provides appropriate temporal detail on subchondral changes in these models, covering the entire spectrum of OA with an emphasis on early and mid-term time points. Microstructural changes of the subarticular spongiosa are comprehensively described; those of the subchondral bone plate are not. In mouse models, global subchondral bone alterations are unidirectional, involving an advancing sclerosis of the trabecular structure over time. In rats, biphasic subchondral bone alterations begin with an osteopenic degeneration and loss of subchondral trabeculae, progressing to a late sclerosis of the entire subchondral bone. Rat models, independently from the applied technique, relatively faithfully mirror the early bone loss detected in larger animals, and the late subchondral bone sclerosis observed in human advanced OA. CONCLUSION Mice and rats allow us to study the microstructural consequences of compromising meniscus integrity at high temporal detail. Thickening of the subchondral bone plate, an early loss of thinner subarticular trabecular elements, followed by a subsequent sclerosis of the entire subchondral bone are all important and reliable hallmarks that occur in parallel with the advancing articular cartilage degeneration. Thoughtful decisions on the study design, laterality, selection of controls and volumes of interest are crucial to obtain meaningful data.
Collapse
Affiliation(s)
- Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany; Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany.
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany.
| |
Collapse
|
4
|
Palma C, Piazza S, Visone R, Ringom R, Björklund U, Bermejo Gómez A, Rasponi M, Occhetta P. An Advanced Mechanically Active Osteoarthritis-on-Chip Model to Test Injectable Therapeutic Formulations: The SYN321 Case Study. Adv Healthc Mater 2024:e2401187. [PMID: 39318108 DOI: 10.1002/adhm.202401187] [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: 03/29/2024] [Revised: 09/10/2024] [Indexed: 09/26/2024]
Abstract
Current treatments for osteoarthritis (OA) often fail to address the underlying pathophysiology and may have systemic side effects, particularly associated with long-term use of non-steroidal anti-inflammatory drugs (NSAIDs). Thus, researchers are currently directing their efforts toward innovative polymer-drug combinations, such as mixtures of hyaluronic acid viscoelastic hydrogels and NSAIDs like diclofenac, to ensure sustained release of the NSAID within the joint following intra-articular injection. However, the progress of novel injectable therapies for OA is hindered by the absence of preclinical models that accurately represent the pathology of the disease. The uBeat® MultiCompress platform is here presented as a novel approach for studying anti-OA injectable therapeutics on human mechanically-damaged OA cartilage microtissues, in a physiologically relevant environment. This platform can accommodate injectable therapeutic formulations and is successfully tested with SYN321, a novel diclofenac-sodium hyaluronate conjugate under development as a treatment for knee OA. Results indicate the platform's effectiveness in evaluating therapeutic potential, showing downregulation of inflammatory markers and reduction in matrix degradation in OA cartilage micro-tissues treated with SYN321. The uBeat® MultiCompress platform thus represents a valuable tool for OA research, offering a bridge between traditional in vitro studies and potential clinical applications, with implications for future drug discovery.
Collapse
Affiliation(s)
- Cecilia Palma
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
| | - Stefano Piazza
- BiomimX Srl, Viale Decumano 41, MIND - Milano Innovation District, Milan, 20157, Italy
| | - Roberta Visone
- BiomimX Srl, Viale Decumano 41, MIND - Milano Innovation District, Milan, 20157, Italy
| | - Rune Ringom
- Recipharm OT Chemistry AB, Virdings allé 18, Uppsala, 754 50, Sweden
| | - Ulf Björklund
- UB-consulting AB, Trädgårdsgatan 7A, Uppsala, 753 09, Sweden
| | | | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
| | - Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
- BiomimX Srl, Viale Decumano 41, MIND - Milano Innovation District, Milan, 20157, Italy
| |
Collapse
|
5
|
Geng N, Fan M, Kuang B, Zhang F, Xian M, Deng L, Chen C, Pan Y, Chen J, Feng N, Liang L, Ye Y, Liu K, Li X, Du Y, Guo F. 10-hydroxy-2-decenoic acid prevents osteoarthritis by targeting aspartyl β hydroxylase and inhibiting chondrocyte senescence in male mice preclinically. Nat Commun 2024; 15:7712. [PMID: 39231947 PMCID: PMC11375154 DOI: 10.1038/s41467-024-51746-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/15/2024] [Indexed: 09/06/2024] Open
Abstract
Osteoarthritis is a degenerative joint disease with joint pain as the main symptom, caused by fibrosis and loss of articular cartilage. Due to the complexity and heterogeneity of osteoarthritis, there is a lack of effective individualized disease-modifying osteoarthritis drugs in clinical practice. Chondrocyte senescence is reported to participate in occurrence and progression of osteoarthritis. Here we show that small molecule 10-hydroxy-2-decenoic acid suppresses cartilage degeneration and relieves pain in the chondrocytes, cartilage explants from osteoarthritis patients, surgery-induced medial meniscus destabilization or naturally aged male mice. We further confirm that 10-hydroxy-2-decenoic acid exerts a protective effect by targeting the glycosylation site in the Asp_Arg_Hydrox domain of aspartyl β-hydroxylase. Mechanistically, 10-hydroxy-2-decenoic acid alleviate cellular senescence through the ERK/p53/p21 and GSK3β/p16 pathways in the chondrocytes. Our study uncovers that 10-hydroxy-2-decenoic acid modulate cartilage metabolism by targeting aspartyl β-hydroxylase to inhibit chondrocyte senescence in osteoarthritis. 10-hydroxy-2-decenoic acid may be a promising therapeutic drug against osteoarthritis.
Collapse
Affiliation(s)
- Nana Geng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Mengtian Fan
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Biao Kuang
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengmei Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Menglin Xian
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Lin Deng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Cheng Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiming Pan
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Jianqiang Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Naibo Feng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Li Liang
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yuanlan Ye
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Kaiwen Liu
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xiaoli Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yu Du
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengjin Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Ichinose H, Natsume T, Yano M, Awaga Y, Hanada M, Takamatsu H, Matsuyama Y. Evaluation of brain activation related to resting pain using functional magnetic resonance imaging in cynomolgus macaques undergoing knee surgery. J Orthop 2024; 52:12-16. [PMID: 38404703 PMCID: PMC10881445 DOI: 10.1016/j.jor.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Functional magnetic resonance imaging (fMRI) visualizes hemodynamic responses associated with brain and spinal cord activation. Various types of pain have been objectively assessed using fMRI as considerable brain activations. This study aimed to develop a pain model in cynomolgus macaques undergoing knee surgery and confirm brain activation due to resting pain after knee surgery. Methods An osteochondral graft surgery on the femoral condyle in the unilateral knee was performed on four cynomolgus macaques (Macaca fascicularis). Resting pain was evaluated as changes in brain fMRI findings with a 3.0-T MRI scanner preoperatively, postoperatively, and after postoperative administration of morphine. In the fMRI analysis, Z-values >1.96 were considered statistically significant. Results Brain activation without stimulation after surgery in the cingulate cortex (3.09) and insular cortex (3.06) on the opposite side of the surgery was significantly greater than that before surgery (1.05 and 1.03, respectively) according to fMRI. After the administration of morphine, activation due to resting pain decreased in the cingulate cortex (1.38) and insular cortex (1.21). Conclusion Osteochondral graft surgery on the femoral condyle can lead to postoperative resting pain. fMRI can reveal activation in pain-related brain areas and evaluate resting pain due to knee surgery.
Collapse
Affiliation(s)
- Hatsumi Ichinose
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takahiro Natsume
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Mizuho Yano
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Yuji Awaga
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Mitsuru Hanada
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hiroyuki Takamatsu
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Yukihiro Matsuyama
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
7
|
Chapman JH, Ghosh D, Attari S, Ude CC, Laurencin CT. Animal Models of Osteoarthritis: Updated Models and Outcome Measures 2016-2023. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:127-146. [PMID: 38983776 PMCID: PMC11233113 DOI: 10.1007/s40883-023-00309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2024]
Abstract
Purpose Osteoarthritis (OA) is a global musculoskeletal disorder that affects primarily the knee and hip joints without any FDA-approved disease-modifying therapies. Animal models are essential research tools in developing therapies for OA; many animal studies have provided data for the initiation of human clinical trials. Despite this, there is still a need for strategies to recapitulate the human experience using animal models to better develop treatments and understand pathogenesis. Since our last review on animal models of osteoarthritis in 2016, there have been exciting updates in OA research and models. The main purpose of this review is to update the latest animal models and key features of studies in OA research. Method We used our existing classification method and screened articles in PubMed and bibliographic search for animal OA models between 2016 and 2023. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Recent studies were analyzed and classified. We also identified ex vivo models as an area of ongoing research. Each animal model offers its own benefit in the study of OA and there are a full range of outcome measures that can be assessed. Despite the vast number of models, each has its drawbacks that have limited translating approved therapies for human use. Conclusion Depending on the outcome measures and objective of the study, researchers should pick the best model for their work. There have been several exciting studies since 2016 that have taken advantage of regenerative engineering techniques to develop therapies and better understand OA. Lay Summary Osteoarthritis (OA) is a chronic debilitating disease without any cure that affects mostly the knee and hip joints and often results in surgical joint replacement. Cartilage protects the joint from mechanical forces and degrades with age or in response to injury. The many contributing causes of OA are still being investigated, and animals are used for preclinical research and to test potential new treatments. A single consensus OA animal model for preclinical studies is non-existent. In this article, we review the many animal models for OA and provide a much-needed update on studies and model development since 2016.
Collapse
Affiliation(s)
- James H. Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Seyyedmorteza Attari
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
8
|
Song X, Liu Y, Chen S, Zhang L, Zhang H, Shen X, Du H, Sun R. Knee osteoarthritis: A review of animal models and intervention of traditional Chinese medicine. Animal Model Exp Med 2024; 7:114-126. [PMID: 38409942 PMCID: PMC11079151 DOI: 10.1002/ame2.12389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/10/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) characterized by degeneration of knee cartilage and subsequent bone hyperplasia is a prevalent joint condition primarily affecting aging adults. The pathophysiology of KOA remains poorly understood, as it involves complex mechanisms that result in the same outcome. Consequently, researchers are interested in studying KOA and require appropriate animal models for basic research. Chinese herbal compounds, which consist of multiple herbs with diverse pharmacological properties, possess characteristics such as multicomponent, multipathway, and multitarget effects. The potential benefits in the treatment of KOA continue to attract attention. PURPOSE This study aims to provide a comprehensive overview of the advantages, limitations, and specific considerations in selecting different species and methods for KOA animal models. This will help researchers make informed decisions when choosing an animal model. METHODS Online academic databases (e.g., PubMed, Google Scholar, Web of Science, and CNKI) were searched using the search terms "knee osteoarthritis," "animal models," "traditional Chinese medicine," and their combinations, primarily including KOA studies published from 2010 to 2023. RESULTS Based on literature retrieval, this review provides a comprehensive overview of the methods of establishing KOA animal models; introduces the current status of advantages and disadvantages of various animal models, including mice, rats, rabbits, dogs, and sheep/goats; and presents the current status of methods used to establish KOA animal models. CONCLUSION This study provides a review of the animal models used in recent KOA research, discusses the common modeling methods, and emphasizes the role of traditional Chinese medicine compounds in the treatment of KOA.
Collapse
Affiliation(s)
- Xuyu Song
- Orthopaedic trauma surgeryThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Ying Liu
- Academy of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Siyi Chen
- Academy of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Lei Zhang
- Department of Traditional Chinese MedicineThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Huijie Zhang
- College of pharmacyShandong University of Traditional Chinese MedicineJinanShandongChina
| | - Xianhui Shen
- The Second Clinical College of Shandong UniversityShandong UniversityJinanShandongChina
| | - Hang Du
- The Second Clinical College of Shandong UniversityShandong UniversityJinanShandongChina
| | - Rong Sun
- Advanced Medical Research InstituteShandong UniversityJinanShandongChina
- The Second Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
9
|
Martínez-Ramos S, García S. An update of murine models and their methodologies in immune-mediated joint damage and pain research. Int Immunopharmacol 2024; 128:111440. [PMID: 38176343 DOI: 10.1016/j.intimp.2023.111440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Murine models have played an indispensable role in the understanding of rheumatic and musculoskeletal disorders (RMD), elucidating the genetic, endocrine and biomechanical pathways involved in joint pathology and associated pain. To date, the available models in RMD can be classified as induced or spontaneous, both incorporating transgenic alternatives that improve specific insights. It is worth noting that the selection of the most appropriate model together with the evaluation of their specific characteristics and technical capabilities are crucial when designing the experiments. Furthermore, it is also imperative to consistently adhere to the ethical standards concerning animal experimentation. Recognizing the inherent limitation that any model can entirely encapsulates the complexity of the pathophysiology of these conditions, the aim of this review is to provide an updated overview on the methodology of current murine models in major arthropathies and their immune-mediated pathways, addressing to basic, translational and pharmacological research in joint damage and pain.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain.
| | - Samuel García
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| |
Collapse
|
10
|
Fernández-Moreno M, Hermida-Gómez T, Larkins N, Reynolds A, Blanco FJ. Anti-Inflammatory Activity of APPA (Apocynin and Paeonol) in Human Articular Chondrocytes. Pharmaceuticals (Basel) 2024; 17:118. [PMID: 38256951 PMCID: PMC10818286 DOI: 10.3390/ph17010118] [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: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease leading to cartilage loss and reduction in the joint space which results in pain. The current pharmacological treatment of OA is inadequate and pharmacological interventions focus on symptom management. APPA, a combination of apocynin (AP) and paeonol (PA), is a potential drug for treating OA. The aim of this study was to analyze the effects of APPA on the modulation of the inflammatory response in chondrocytes. Samples were incubated with IL-1β and APPA, and their responses to proinflammatory cytokines, catabolic mediators and redox responses were then measured. The effect of APPA on mitogenesis was also evaluated. Results show that APPA attenuated the expression of IL-8, TNF-α, MMP-3, MMP-13, SOD-2 and iNOS, resulting in the protection of human articular cartilage. APPA decreased PGC-1α gene expression induced by IL-1β. APPA did not modulate the gene expression of Mfn2, Sirt-1 or Sirt-3. The overall findings indicate that APPA may be an effective treatment for OA by targeting several of the pathways involved in OA pathogenesis.
Collapse
Affiliation(s)
- Mercedes Fernández-Moreno
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade de A Coruña (UDC), 15071 A Coruña, Spain;
- Grupo de Investigación en Reumatología y Salud (GIR-S), Centro Interdisciplinar de Química y Biología (CICA), Universidade de A Coruña (UDC), Campus de Elviña, 15071 A Coruña, Spain
| | - Tamara Hermida-Gómez
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade de A Coruña (UDC), 15071 A Coruña, Spain;
- Grupo de Investigación en Reumatología y Salud (GIR-S), Centro Interdisciplinar de Química y Biología (CICA), Universidade de A Coruña (UDC), Campus de Elviña, 15071 A Coruña, Spain
- Centro de Investigación Biomédica en Red, Bioingenieria, Biomatereial y Nanomedicina (CIBER-BBN), 50018 Zaragoza, Spain
| | - Nicholas Larkins
- AKL Therapeutics Ltd., Stevenage Bioscience, Gunnels Wood Rd, Stevenage SG1 2FX, UK; (N.L.); (A.R.)
| | - Alan Reynolds
- AKL Therapeutics Ltd., Stevenage Bioscience, Gunnels Wood Rd, Stevenage SG1 2FX, UK; (N.L.); (A.R.)
| | - Francisco J. Blanco
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade de A Coruña (UDC), 15071 A Coruña, Spain;
- Grupo de Investigación en Reumatología y Salud (GIR-S), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Centro Interdisciplinar de Química y Biología (CICA), INIBIC-Sergas, Universidade de A Coruña (UDC), Campus de Oza, 15008 A Coruña, Spain
| |
Collapse
|
11
|
Rhee B, Jin C, Shin SH, Choi H, Lee Y, Kim S. Establishment of an image evaluation grading criteria for experimental stifle joint osteoarthritis in dogs: an X-ray and CT imaging study. Lab Anim Res 2023; 39:34. [PMID: 38102726 PMCID: PMC10722845 DOI: 10.1186/s42826-023-00186-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND This study aimed to establish an image evaluation grading criteria for experimental stifle joint osteoarthritis (OA) in anterior cruciate ligament transection induced OA beagle dog models. The severity of OA was assessed using X-ray and computed tomography (CT) imaging. RESULTS A total of 32 dogs (8 controls and 24 OA-induced dogs) were included in the study. The OA-induced group showed significantly higher manual joint palpation, gait analysis, and OA severity scores than the control group. Based on these two results, we calculated correlation coefficients. There was a strong positive correlation between manual joint palpation scores and OA severity on diagnostic imaging and between gait analysis scores and OA severity. CONCLUSIONS The developed grading criteria based on radiographic evaluation correlated with clinical assessments. The study also employed CT imaging to enhance the accuracy and sensitivity of early-stage OA change detection in the stifle joint. However, further studies with larger sample sizes and multiple evaluators are recommended for the validation and generalizability of this grading system. These established image evaluation grading criteria can help evaluate and monitor the efficacy of interventions and changes in OA lesions in canine models.
Collapse
Affiliation(s)
- Beomseok Rhee
- Research Center, HLB BioStep Co., Ltd., Incheon, 22014, Republic of Korea
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Changfan Jin
- Research Center, HLB BioStep Co., Ltd., Incheon, 22014, Republic of Korea
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seo-Hyun Shin
- Research Center, HLB BioStep Co., Ltd., Incheon, 22014, Republic of Korea
| | - Hojung Choi
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Youngwon Lee
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sokho Kim
- Research Center, HLB BioStep Co., Ltd., Incheon, 22014, Republic of Korea.
| |
Collapse
|
12
|
Haubruck P, Heller R, Blaker CL, Clarke EC, Smith SM, Burkhardt D, Liu Y, Stoner S, Zaki S, Shu CC, Little CB. Streamlining quantitative joint-wide medial femoro-tibial histopathological scoring of mouse post-traumatic knee osteoarthritis models. Osteoarthritis Cartilage 2023; 31:1602-1611. [PMID: 37716405 DOI: 10.1016/j.joca.2023.07.013] [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: 08/07/2022] [Revised: 07/03/2023] [Accepted: 07/26/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVES Histological scoring remains the gold-standard for quantifying post-traumatic osteoarthritis (ptOA) in animal models, allowing concurrent evaluation of numerous joint tissues. Available systems require scoring multiple sections/joint making analysis laborious and expensive. We investigated if a single section allowed equivalent quantitation of pathology in different joint tissues and disease stages, in three ptOA models. METHOD Male 10-12-week-old C57BL/6 mice underwent surgical medial-meniscal-destabilization, anterior-cruciate-ligament (ACL) transection, non-invasive-ACL-rupture, or served as sham-surgical, non-invasive-ACL-strain, or naïve/non-operated controls. Mice (n = 12/group) were harvested 1-, 4-, 8-, and 16-week post-intervention. Serial sagittal toluidine-blue/fast-green stained sections of the medial-femoro-tibial joint (n = 7/joint, 84 µm apart) underwent blinded scoring of 40 histology-outcomes. We evaluated agreement between single-slide versus entire slide-set maximum or median scores (weighted-kappa), and sensitivity/specificity of single-slide versus median/maximum to detect OA pathology. RESULTS A single optimal mid-sagittal section showed excellent agreement with median (weighted-kappa 0.960) and maximum (weighted-kappa 0.926) scores. Agreement for individual histology-outcomes was high with only 19/240 median and 15/240 maximum scores having a weighted-kappa ≤0.4, the majority of these (16/19 and 11/15) in control groups. Statistically-significant histology-outcome differences between ptOA models and their controls detected with the entire slide-set were reliably reproduced using a single slide (sensitivity >93.15%, specificity >93.10%). The majority of false-negatives with single-slide scoring were meniscal and subchondral bone histology-outcomes (89%) and occurred in weeks 1-4 post-injury (84%). CONCLUSION A single mid-sagittal slide reduced the time needed to score diverse histopathological changes by 87% without compromising the sensitivity or specificity of the analysis, across a variety of ptOA models and time-points.
Collapse
Affiliation(s)
- Patrick Haubruck
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Raban Heller
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany; Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany; Bundeswehr Hospital Berlin, Clinic of Traumatology and Orthopaedics, D-10115 Berlin, Germany
| | - Carina L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia; Murray Maxwell Biomechanics Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Elizabeth C Clarke
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Susan M Smith
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Daniel Burkhardt
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Yolanda Liu
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Shihani Stoner
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia; Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia
| | - Cindy C Shu
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| |
Collapse
|
13
|
Reina-Mahecha A, Beers MJ, van der Veen HC, Zuhorn IS, van Kooten TG, Sharma PK. A Review of the Role of Bioreactors for iPSCs-Based Tissue-Engineered Articular Cartilage. Tissue Eng Regen Med 2023; 20:1041-1052. [PMID: 37861960 PMCID: PMC10645985 DOI: 10.1007/s13770-023-00573-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common degenerative joint disease without an ultimate treatment. In a search for novel approaches, tissue engineering (TE) has shown great potential to be an effective way for hyaline cartilage regeneration and repair in advanced stages of OA. Recently, induced pluripotent stem cells (iPSCs) have been appointed to be essential stem cells for degenerative disease treatment because they allow a personalized medicine approach. For clinical translation, bioreactors in combination with iPSCs-engineerd cartilage could match patients needs, serve as platform for large-scale patient specific cartilage production, and be a tool for patient OA modelling and drug screening. Furthermore, to minimize in vivo experiments and improve cell differentiation and cartilage extracellular matrix (ECM) deposition, TE combines existing approaches with bioreactors. METHODS This review summarizes the current understanding of bioreactors and the necessary parameters when they are intended for cartilage TE, focusing on the potential use of iPSCs. RESULTS Bioreactors intended for cartilage TE must resemble the joint cavity niche. However, recreating human synovial joints is not trivial because the interactions between various stimuli are not entirely understood. CONCLUSION The use of mechanical and electrical stimulation to differentiate iPSCs, and maintain and test chondrocytes are key stimuli influencing hyaline cartilage homeostasis. Incorporating these stimuli to bioreactors can positively impact cartilage TE approaches and their possibility for posterior translation into the clinics.
Collapse
Affiliation(s)
- Alejandro Reina-Mahecha
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Martine J Beers
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Hugo C van der Veen
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge S Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Theo G van Kooten
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands.
| |
Collapse
|
14
|
Macfarlane E, Cavanagh L, Fong-Yee C, Tuckermann J, Chen D, Little CB, Seibel MJ, Zhou H. Deletion of the chondrocyte glucocorticoid receptor attenuates cartilage degradation through suppression of early synovial activation in murine posttraumatic osteoarthritis. Osteoarthritis Cartilage 2023; 31:1189-1201. [PMID: 37105394 DOI: 10.1016/j.joca.2023.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/31/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVE Disruption of endogenous glucocorticoid signalling in bone cells attenuates osteoarthritis (OA) in aged mice, however, the role of endogenous glucocorticoids in chondrocytes is unknown. Here, we investigated whether deletion of the glucocorticoid receptor, specifically in chondrocytes, also alters OA progression. DESIGN Knee OA was induced by surgical destabilisation of the medial meniscus (DMM) in male 22-week-old tamoxifen-inducible glucocorticoid receptor knockout (chGRKO) mice and their wild-type (WT) littermates (n = 7-9/group). Mice were harvested 2, 4, 8 and 16 weeks after surgery to examine the spatiotemporal changes in molecular, cellular, and histological characteristics. RESULTS At all time points following DMM, cartilage damage was significantly attenuated in chGRKO compared to WT mice. Two weeks after DMM, WT mice exhibited increased chondrocyte and synoviocyte hypoxia inducible factor (HIF)-2α expression resulting in extensive synovial activation characterised by synovial thickening and increased interleukin-1 beta expression. At 2 and 4 weeks after DMM, WT mice displayed pronounced chondrocyte senescence and elevated catabolic signalling (reduced Yes-associated protein 1 (YAP1) and increased matrix metalloprotease [MMP]-13 expression). Contrastingly, at 2 weeks after DMM, HIF-2α expression and synovial activation were much less pronounced in chGRKO than in WT mice. Furthermore, chondrocyte YAP1 and MMP-13 expression, as well as chondrocyte senescence were similar in chGRKO-DMM mice and sham-operated controls. CONCLUSION Endogenous glucocorticoid signalling in chondrocytes promotes synovial activation, chondrocyte senescence and cartilage degradation by upregulation of catabolic signalling through HIF-2α in murine posttraumatic OA. These findings indicate that inhibition of glucocorticoid signalling early after injury may present a promising way to slow osteoarthritic cartilage degeneration.
Collapse
Affiliation(s)
- Eugenie Macfarlane
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| | - Lauryn Cavanagh
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| | - Colette Fong-Yee
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Baden-Württemberg, Germany.
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| | - Christopher B Little
- Raymond Purves Laboratories, Kolling Institute and Institute of Bone and Joint Research, University of Sydney, and Royal North Shore Hospital, St. Leonards, NSW, Australia.
| | - Markus J Seibel
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology and Metabolism, Concord Repatriation General Hospital, Sydney, NSW, Australia.
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
15
|
Somersan-Karakaya S, Turner KC, Cortes-Burgos L, Miller J, LaCroix-Fralish M, Logovinsky V, Patel Y, Torres R, Ganguly S, Breazna A, DeVeaux M, Bhore R, Gao M, Delfino FJ, Rafique A, Fairhurst JL, Hunt C, Babb R, Badithe A, Poueymirou WT, Surowitz R, Rottey S, Murphy AJ, Harari O, Macdonald LE, Croll SD. Monoclonal antibodies against GFRα3 are efficacious against evoked hyperalgesic and allodynic responses in mouse join pain models but, one of these, REGN5069, was not effective against pain in a randomized, placebo-controlled clinical trial in patients with osteoarthritis pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 14:100136. [PMID: 38099276 PMCID: PMC10719528 DOI: 10.1016/j.ynpai.2023.100136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 12/17/2023]
Abstract
The artemin-GFRα3 signaling pathway has been implicated in various painful conditions including migraine, cold allodynia, hyperalgesia, inflammatory bone pain, and mouse knees contain GFRα3-immunoreactive nerve endings. We developed high affinity mouse (REGN1967) and human (REGN5069) GFRα3-blocking monoclonal antibodies and, following in vivo evaluations in mouse models of chronic joint pain (osteoarthritic-like and inflammatory), conducted a first-in-human phase 1 pharmacokinetics (PK) and safety trial of REGN5069 (NCT03645746) in healthy volunteers, and a phase 2 randomized placebo-controlled efficacy and safety trial of REGN5069 (NCT03956550) in patients with knee osteoarthritis (OA) pain. In three commonly used mouse models of chronic joint pain (destabilization of the medial meniscus, intra-articular monoiodoacetate, or Complete Freund's Adjuvant), REGN1967 and REGN5069 attenuated evoked behaviors including tactile allodynia and thermal hyperalgesia without discernably impacting joint pathology or inflammation, prompting us to further evaluate REGN5069 in humans. In the phase 1 study in healthy subjects, the safety profiles of single doses of REGN5069 up to 3000 mg (intravenous) or 600 mg (subcutaneous) were comparable to placebo; PK were consistent with a monoclonal antibody exhibiting target-mediated disposition. In the phase 2 study in patients with OA knee pain, two doses of REGN5069 (100 mg or 1000 mg intravenous every 4 weeks) for 8 weeks failed to achieve the 12-week primary and secondary efficacy endpoints relative to placebo. In addition to possible differences in GFRα3 biology between mice and humans, we highlight here differences in experimental parameters that could have contributed to a different profile of efficacy in mouse models versus human OA pain. Additional research is required to more fully evaluate any potential role of GFRα3 in human pain.
Collapse
Affiliation(s)
| | | | | | - Jutta Miller
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | | | - Yamini Patel
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Richard Torres
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Samit Ganguly
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Aurora Breazna
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Rafia Bhore
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Min Gao
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Ashique Rafique
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Charleen Hunt
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Robert Babb
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Ashok Badithe
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | | | | | | | - Olivier Harari
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Susan D. Croll
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| |
Collapse
|
16
|
Chan KM, Bowe MT, Allen KD. Recommendations for the analysis of rodent gait data to evaluate osteoarthritis treatments. Osteoarthritis Cartilage 2023; 31:425-434. [PMID: 36435413 PMCID: PMC11474404 DOI: 10.1016/j.joca.2022.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/15/2022] [Accepted: 11/10/2022] [Indexed: 11/26/2022]
Abstract
Behavioral assays of animal pain and disability can increase the clinical relevance of a preclinical study. However, pain and symptoms are difficult to measure in preclinical models. Because animals often alter their movement patterns to reduce or avoid joint pain, gait analysis can be an important tool for quantifying OA-related symptoms in rodents. Technologies to measure rodent gait continue to advance and have been the focus of prior reviews. Regardless of the techniques used, the analysis of rodent gait data can be complex due to multiple confounding variables. The goal of this review is to discuss recent advances in the understanding of OA-related gait changes and provide recommendations on the analysis of gait data. Recent studies suggest OA-affected animals reduce vertical loading through their injured limb while walking, indicating dynamic ground reaction forces are important data to collect when possible. Moreover, gait data analysis depends on accurately measuring and accounting for the confounding effects of velocity and other covariates (such as animal size) when interpreting shifts in various gait parameters. Herein, we discuss different statistical techniques to account for covariates and interpret gait shifts. In particular, this review will discuss residualization and linear mixed effects models, including how both techniques can account for inter- and intra-animal variability and the effects of velocity. Furthermore, this review discusses future considerations for using rodent gait analysis, while highlighting the intricacies of gait analysis as a tool to measure joint function and behavioral outcomes.
Collapse
Affiliation(s)
- Kiara M Chan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Markia T Bowe
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Orthopedics and Sports Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
17
|
Karami P, Stampoultzis T, Guo Y, Pioletti DP. A guide to preclinical evaluation of hydrogel-based devices for treatment of cartilage lesions. Acta Biomater 2023; 158:12-31. [PMID: 36638938 DOI: 10.1016/j.actbio.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/19/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
The drive to develop cartilage implants for the treatment of major defects in the musculoskeletal system has resulted in a major research thrust towards developing biomaterial devices for cartilage repair. Investigational devices for the restoration of articular cartilage are considered as significant risk materials by regulatory bodies and therefore proof of efficacy and safety prior to clinical testing represents a critical phase of the multidisciplinary effort to bridge the gap between bench and bedside. To date, review articles have thoroughly covered different scientific facets of cartilage engineering paradigm, but surprisingly, little attention has been given to the preclinical considerations revolving around the validation of a biomaterial implant. Considering hydrogel-based cartilage products as an example, the present review endeavors to provide a summary of the critical prerequisites that such devices should meet for cartilage repair, for successful implantation and subsequent preclinical validation prior to clinical trials. Considerations pertaining to the choice of appropriate animal model, characterization techniques for the quantitative and qualitative outcome measures, as well as concerns with respect to GLP practices are also extensively discussed. This article is not meant to provide a systematic review, but rather to introduce a device validation-based roadmap to the academic investigator, in anticipation of future healthcare commercialization. STATEMENT OF SIGNIFICANCE: There are significant challenges around translation of in vitro cartilage repair strategies to approved therapies. New biomaterial-based devices must undergo exhaustive investigations to ensure their safety and efficacy prior to clinical trials. These considerations are required to be applied from early developmental stages. Although there are numerous research works on cartilage devices and their in vivo evaluations, little attention has been given into the preclinical pathway and the corresponding approval processes. With a focus on hydrogel devices to concretely illustrate the preclinical path, this review paper intends to highlight the various considerations regarding the preclinical validation of hydrogel devices for cartilage repair, from regulatory considerations, to implantation strategies, device performance aspects and characterizations.
Collapse
Affiliation(s)
- Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Theofanis Stampoultzis
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Yanheng Guo
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland.
| |
Collapse
|
18
|
Go EJ, Kim SA, Cho ML, Lee KS, Shetty AA, Kim SJ. A Combination of Surgical and Chemical Induction in a Rabbit Model for Osteoarthritis of the Knee. Tissue Eng Regen Med 2022; 19:1377-1388. [PMID: 36318365 PMCID: PMC9679088 DOI: 10.1007/s13770-022-00488-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/14/2022] [Accepted: 08/28/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Appropriate animal models of osteoarthritis (OA) are essential to develop new treatment modalities for OA. A combination of surgical and chemical induction could be appropriate for OA models. METHODS Rabbit knee OA models developed by surgical induction (anterior cruciate ligament transection [ACLT]), chemical induction (monosodium iodoacetate [MIA] injection), and a combination of both were compared to assess compositional and structural destruction of the knee joint. Twenty-one New Zealand white rabbits were randomly divided into 3 groups to induce OA (group 1: ACLT, n = 3; group 2: MIA [3, 6, 9 mg] injection, n = 9; group 3: ACLT + MIA [3, 6, 9 mg] injection, n = 9). RESULTS In all groups, the Modified Mankin score was significantly higher in the osteoarthritis-induced knee than in the control. Modified Mankin scores were compared by category. The ACLT group was observed to score high in cartilage structure. In the MIA group, chondrocytes and matrix staining showed higher scores, and the ACLT+MIA group scored higher in all categories for cartilage structure, chondrocytes, matrix staining, and tidemark integrity. The ACLT + 3 mg MIA showed definite OA characteristics such as cartilage surface destruction and degeneration of cartilage layers, and the ACLT + 6 mg MIA and ACLT + 9 mg MIA showed more prominent OA characteristics such as cartilage surface destruction, matrix disorganization, and osteophyte formation. CONCLUSION The combination of MIA injection and ACLT could be an appropriate method for OA induction in rabbit models.
Collapse
Affiliation(s)
- Eun Jeong Go
- Department of Orthopaedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Cheonbo-ro, Uijeongbu-si, Gyeonggi-do, 271, Republic of Korea
| | - Seon Ae Kim
- Department of Orthopaedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Cheonbo-ro, Uijeongbu-si, Gyeonggi-do, 271, Republic of Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kwan Soo Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Asode Ananthram Shetty
- Faculty of Health and Social Care, Institute of Medical Sciences, Canterbury Christ Church University, Kent, UK
| | - Seok Jung Kim
- Department of Orthopaedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Cheonbo-ro, Uijeongbu-si, Gyeonggi-do, 271, Republic of Korea.
| |
Collapse
|
19
|
Williams MD, Meyers RC, Braxton LA, Diekman B, Lascelles BDX. Pilot comparison of outcome measures across chemical and surgical experimental models of chronic osteoarthritis in the rat (Rattus norvegicus). PLoS One 2022; 17:e0277943. [PMID: 36409758 PMCID: PMC9678322 DOI: 10.1371/journal.pone.0277943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Relatively little work has evaluated both the disease of osteoarthritis (OA) and clinically-relevant pain outcome measures across different OA models in rats. The objective of this study was to compare sensitivity, pain, and histological disease severity across chemical and surgical models of OA in the rat. Stifle OA was induced in Sprague-Dawley rats via intraarticular injection of monoiodoacetate (MIA) or surgical transection of anterior cruciate ligament and/or destabilization of medial meniscus (ACL+DMM or DMM alone). Reflexive (e.g., mechanical and thermal stimuli) measures of sensitivity and non-reflexive assays (e.g., lameness, static hindlimb weight-bearing asymmetry, dynamic gait analysis) of pain were measured over time. Joint degeneration was assessed histologically. Six-weeks post OA-induction, the ACL+DMM animals had significantly greater visually observed lameness than MIA animals; however, both ACL+DMM and MIA animals showed equal pain as measured by limb use during ambulation and standing. The MIA animals showed increased thermal, but not mechanical, sensitivity compared to ACL+DMM animals. Joint degeneration was significantly more severe in the MIA model at 6 weeks. Our pilot data suggest both the ACL+DMM and MIA models are equal in terms of clinically relevant pain behaviors, but the MIA model is associated with more severe histological changes over time potentially making it more suitable for screening disease modifying agents. Future work should further characterize each model in terms of complex pain behaviors and biochemical, molecular, and imaging analysis of the sensory system and joint tissues, which will allow for more informed decisions associated with model selection and investigative outcomes.
Collapse
Affiliation(s)
- Morika D. Williams
- Division of Comparative Medicine, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Translational Research in Pain Program, North Carolina State University, Raleigh, North Carolina, United States of America
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, North Carolina, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Rachel C. Meyers
- Translational Research in Pain Program, North Carolina State University, Raleigh, North Carolina, United States of America
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Lauryn A. Braxton
- Translational Research in Pain Program, North Carolina State University, Raleigh, North Carolina, United States of America
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, North Carolina, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Brian Diekman
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - B. Duncan X. Lascelles
- Translational Research in Pain Program, North Carolina State University, Raleigh, North Carolina, United States of America
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, North Carolina, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
- Division of Rheumatology, Allergy, and Immunology, Thurston Arthritis Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Center for Translational Pain Medicine, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
20
|
Hargreaves R, Akinsanya K, Ajit SK, Dhruv NT, Driscoll J, Farina P, Gavva N, Gill M, Houghton A, Iyengar S, Jones C, Kavelaars A, Kaykas A, Koroshetz WJ, Laeng P, Laird JM, Lo DC, Luthman J, Munro G, Oshinsky ML, Sittampalam GS, Woller SA, Tamiz AP. Preclinical target validation for non-addictive therapeutics development for pain. Expert Opin Ther Targets 2022; 26:811-822. [DOI: 10.1080/14728222.2022.2147063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | | | - Seena K. Ajit
- Department of Pharmacology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States
| | - Neel T. Dhruv
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Jamie Driscoll
- National Institute of Mental Health, Bethesda, Maryland, United States
| | - Peter Farina
- Canaan Partners, Westport, Connecticut, United States
| | - Narender Gavva
- Drug Discovery Sciences, Takeda Pharmaceuticals, San Diego, California, United States
| | - Marie Gill
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | | | - Smriti Iyengar
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Carrie Jones
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States
| | - Annemieke Kavelaars
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | | | - Walter J. Koroshetz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Pascal Laeng
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Jennifer M. Laird
- Eli Lilly and Company, Windlesham, United Kingdom of Great Britain and Northern Ireland
| | - Donald C. Lo
- National Center for Advancing Translational Sciences, Bethesda, Maryland, United States
| | | | | | - Michael L. Oshinsky
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - G. Sitta Sittampalam
- National Center for Advancing Translational Sciences, Bethesda, Maryland, United States
| | - Sarah A. Woller
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Amir P. Tamiz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| |
Collapse
|
21
|
Wang Y, Wagner ES, Yu D, Chen KJ, Keel TJ, Pownder SL, Koff MF, Cheetham J, Samaroo KJ, Reesink HL. Assessment of osteoarthritis functional outcomes and intra-articular injection volume in the rat anterior cruciate ligament transection model. J Orthop Res 2022; 40:2004-2014. [PMID: 34994469 PMCID: PMC9259760 DOI: 10.1002/jor.25245] [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: 06/01/2021] [Revised: 11/24/2021] [Accepted: 12/13/2021] [Indexed: 02/04/2023]
Abstract
The rat surgical anterior cruciate ligament transection (ACLT) model is commonly used to investigate intra-articular osteoarthritis (OA) therapies, and histological assessment is often the primary outcome measure. However, histological changes do not always correlate well with clinical outcomes. Therefore, this study evaluated functional outcomes in the rat surgical ACLT model and compared intra-articular injection volumes ranging from 20 to 50 μl. Unilateral ACLT was surgically induced and static weight-bearing, mechanical allodynia, motor function, and gait were assessed in four groups of male, Sprague-Dawley rats (n = 6 per group). Intra-articular injections of 20 µl Dulbecco's phosphate-buffered saline (DPBS), 50 µl DPBS, or 50 µl of synthetic biomimetic boundary lubricant were administered once weekly for 3 weeks postoperatively. Structural changes were evaluated histologically at 20 weeks. Rat cadaver knees were injected with 20, 30, 40, or 50 µl of gadolinium solutions and were imaged using magnetic resonance imaging (MRI). Static weight-bearing, mechanical allodynia, and gait parameters in ACLT groups revealed differences from baseline and naïve controls for 4 weeks post-ACLT; however, these differences did not persist beyond 6 weeks. Different intra-articular DPBS injection volumes did not result in functional or histological changes; however, peri-articular leakage was documented via MRI following 50, 40, and 30 µl but not 20 µl gadolinium injections. Statement of clinical significance: Differences in functional parameters were predominantly restricted to early, postoperative changes in the rat surgical ACLT model despite evidence of moderate histologic OA at 20 weeks. Injection volumes of 20-30 µl are more appropriate for investigating intra-articular therapies in the rat knee.
Collapse
Affiliation(s)
- Yuyan Wang
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Emma S. Wagner
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Danqiao Yu
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Kevin J. Chen
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Taidhgin J. Keel
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Sarah L. Pownder
- Magnetic Resonance Imaging Laboratory, Hospital for Special Surgery, New York, NY
| | - Matthew F. Koff
- Magnetic Resonance Imaging Laboratory, Hospital for Special Surgery, New York, NY
| | - Jonathan Cheetham
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | | | - Heidi L. Reesink
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| |
Collapse
|
22
|
Little CB, Zaki S, Blaker CL, Clarke EC. Animal models of osteoarthritis. Bone Joint Res 2022; 11:514-517. [PMID: 35909339 PMCID: PMC9396918 DOI: 10.1302/2046-3758.118.bjr-2022-0217.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Cite this article: Bone Joint Res 2022;11(8):514–517.
Collapse
Affiliation(s)
- Christopher B. Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, Australia
| | - Carina L. Blaker
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| | - Elizabeth C. Clarke
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| |
Collapse
|
23
|
Effects of Intra-Articular Autologous Adipose Micrograft for the Treatment of Osteoarthritis in Dogs: A Prospective, Randomized, Controlled Study. Animals (Basel) 2022; 12:ani12141844. [PMID: 35883392 PMCID: PMC9311928 DOI: 10.3390/ani12141844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/14/2022] [Indexed: 12/12/2022] Open
Abstract
The purpose of this study was to estimate the safety, feasibility, and efficacy of the intra-articular treatment of autologous microfragmented adipose tissue in dogs with spontaneous osteoarthritis (OA) in comparison with hyaluronic acid (HA), the standard intra-articular treatment. Specifically, it clinically evaluated pain and lameness, the radiographic progression of osteoarthritis, and synovial fluid inflammation. This was a prospective, single-center, parallel-group, randomized, controlled, in vivo clinical study. Participants (n = 40) received either a single intra-articular injection of microfragmented adipose tissue or a single intra-articular injection of HA (1:1). Clinical outcomes were determined using a specialistic clinician assessment obtained by the completion of a specific clinical form based on the Vesseur modified lameness classification system, a pain evaluation using the Visual Analogue Scale (VAS), the measurement of the range of motion (ROM) of the affected joint, limb circumference, and the owners' score evaluation using the Canine Brief Pain Inventory (CBPI) for up to 6 months after the time of injection. Patients underwent a radiographic examination to establish the degree of OA in the affected joint, and synovial fluid samples were collected to assess the biochemical environment of the joint and evaluate and quantify the cellular population and the presence of three specific inflammation biomarkers for up to 60 days. The results of this study suggest that microfragmented autologous adipose tissue is safe and can effectively relieve pain and improve function in dogs with spontaneous articular OA. This one-step procedure is simple, timesaving, cost-effective, minimally invasive, and eliminates the need for complex and time-intensive cell culture processing. Furthermore, the clinical evidence and cytological results suggest better long-term pain control, resulting in an improvement in joint function, compared to HA treatment. The canine spontaneous OA model could play a key role in developing successful treatments for human medicine.
Collapse
|
24
|
D'Amico D, Olmer M, Fouassier AM, Valdés P, Andreux PA, Rinsch C, Lotz M. Urolithin A improves mitochondrial health, reduces cartilage degeneration, and alleviates pain in osteoarthritis. Aging Cell 2022; 21:e13662. [PMID: 35778837 PMCID: PMC9381911 DOI: 10.1111/acel.13662] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 05/11/2022] [Accepted: 06/07/2022] [Indexed: 01/22/2023] Open
Abstract
Osteoarthritis (OA) is the most common age‐related joint disorder with no effective therapy. According to the World Health Organization, OA affects over 500 million people and is characterized by degradation of cartilage and other joint tissues, severe pain, and impaired mobility. Mitochondrial dysfunction contributes to OA pathology. However, interventions to rescue mitochondrial defects in human OA are not available. Urolithin A (Mitopure) is a natural postbiotic compound that promotes mitophagy and mitochondrial function and beneficially impacts muscle health in preclinical models of aging and in elderly and middle‐aged humans. Here, we showed that Urolithin A improved mitophagy and mitochondrial respiration in primary chondrocytes from joints of both healthy donors and OA patients. Furthermore, Urolithin A reduced disease progression in a mouse model of OA, decreasing cartilage degeneration, synovial inflammation, and pain. These improvements were associated with increased mitophagy and mitochondrial content, in joints of OA mice. These findings indicate that UA promotes joint mitochondrial health, alleviates OA pathology, and supports Urolithin A's potential to improve mobility with beneficial effects on structural damage in joints.
Collapse
Affiliation(s)
- Davide D'Amico
- Amazentis SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Merissa Olmer
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | | | - Pamela Valdés
- Amazentis SA, EPFL Innovation Park, Lausanne, Switzerland
| | | | - Chris Rinsch
- Amazentis SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Martin Lotz
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| |
Collapse
|
25
|
Li Z, Lin Z, Liu S, Yagi H, Zhang X, Yocum L, Romero‐Lopez M, Rhee C, Makarcyzk MJ, Yu I, Li EN, Fritch MR, Gao Q, Goh KB, O'Donnell B, Hao T, Alexander PG, Mahadik B, Fisher JP, Goodman SB, Bunnell BA, Tuan RS, Lin H. Human Mesenchymal Stem Cell-Derived Miniature Joint System for Disease Modeling and Drug Testing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105909. [PMID: 35436042 PMCID: PMC9313499 DOI: 10.1002/advs.202105909] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/04/2022] [Indexed: 05/12/2023]
Abstract
Diseases of the knee joint such as osteoarthritis (OA) affect all joint elements. An in vitro human cell-derived microphysiological system capable of simulating intraarticular tissue crosstalk is desirable for studying etiologies/pathogenesis of joint diseases and testing potential therapeutics. Herein, a human mesenchymal stem cell-derived miniature joint system (miniJoint) is generated, in which engineered osteochondral complex, synovial-like fibrous tissue, and adipose tissue are integrated into a microfluidics-enabled bioreactor. This novel design facilitates different tissues communicating while still maintaining their respective phenotypes. The miniJoint exhibits physiologically relevant changes when exposed to interleukin-1β mediated inflammation, which are similar to observations in joint diseases in humans. The potential of the miniJoint in predicting in vivo efficacy of drug treatment is confirmed by testing the "therapeutic effect" of the nonsteroidal anti-inflammatory drug, naproxen, as well as four other potential disease-modifying OA drugs. The data demonstrate that the miniJoint recapitulates complex tissue interactions, thus providing a robust organ chip model for the study of joint pathology and the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Zhong Li
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Zixuan Lin
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Silvia Liu
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPA15261USA
| | - Haruyo Yagi
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Xiurui Zhang
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Lauren Yocum
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | | | - Claire Rhee
- Department of Orthopaedic SurgeryStanford UniversityStanfordCA94305USA
| | - Meagan J. Makarcyzk
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
| | - Ilhan Yu
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Eileen N. Li
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
| | - Madalyn R. Fritch
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Qi Gao
- Department of Orthopaedic SurgeryStanford UniversityStanfordCA94305USA
| | - Kek Boon Goh
- Institute of PhysicsUniversity of FreiburgFreiburg79104Germany
- School of EngineeringMonash University MalaysiaSelangor47500Malaysia
| | - Benjamen O'Donnell
- Center for Stem Cell Research and Regenerative MedicineTulane University School of MedicineOrleansLA70112USA
| | - Tingjun Hao
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Peter G. Alexander
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Bhushan Mahadik
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - John P. Fisher
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Stuart B. Goodman
- Department of Orthopaedic SurgeryStanford UniversityStanfordCA94305USA
| | - Bruce A. Bunnell
- Center for Stem Cell Research and Regenerative MedicineTulane University School of MedicineOrleansLA70112USA
- Present address:
Department of Microbiology, Immunology, and GeneticsUniversity of North Texas Health Science CenterFort WorthTX76107USA
| | - Rocky S. Tuan
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
- McGowan Institute for Regenerative MedicineUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Present address:
The Chinese University of Hong KongShatinHong Kong SAR999077China
| | - Hang Lin
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
- McGowan Institute for Regenerative MedicineUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| |
Collapse
|
26
|
Alves-Simões M. Rodent models of knee osteoarthritis for pain research. Osteoarthritis Cartilage 2022; 30:802-814. [PMID: 35139423 DOI: 10.1016/j.joca.2022.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease and a leading cause of disability worldwide. Pain is the main symptom, yet no current treatment can halt disease progression or effectively provide symptomatic relief. Numerous animal models have been described for studying OA and some for the associated OA pain. This review aims to update on current models used for studying OA pain, focusing on mice and rats. These models include surgical, chemical, mechanical, and spontaneous OA models. The impact of sex and age will also be addressed in the context of OA modelling. Although no single animal model has been shown ideal for studying OA pain, increased efforts to phenotype OA will likely impact the choice of models for pre-clinical and basic research studies.
Collapse
Affiliation(s)
- M Alves-Simões
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
27
|
Development and characterization of a humanized mouse model of osteoarthritis. Osteoarthritis Cartilage 2022; 30:875-885. [PMID: 35307533 DOI: 10.1016/j.joca.2022.02.620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In light of the role of immune cells in OA pathogenesis, the development of sophisticated animal models closely mimicking the immune dysregulation during the disease development and progression could be instrumental for the preclinical evaluation of novel treatments. Among these models, immunologically humanized mice may represent a relevant system, particularly for testing immune-interacting DMOADs or cell therapies before their transfer to the clinic. Our objective, therefore, was to develop an experimental model of OA by destabilization of the medial meniscus (DMM) in humanized mice. METHOD Irradiated 5-week-old NOD/LtSz-scid IL2Rγnull (NSG) mice were humanized by intravenous injection of CD34+ human hematopoietic stem cells. The engraftment efficiency was evaluated by flow cytometry 17 weeks after the humanization procedure. Humanized and non-humanized NSG mice underwent DMM or sham surgery and OA development was assessed 1, 6, and 12 weeks after the surgery. RESULTS 120 days after the humanization, human T and B lymphocytes, macrophages and NK cells, were present in the blood and spleen of the humanized NSG mice. The DMM surgery induced articular cartilage and meniscal alterations associated with an increase in OA and the meniscal score. Moreover, the surgery triggered an inflammatory response that was sustained at a low grade in the DMM group. CONCLUSIONS Our study shows for the first time the feasibility of inducing OA by DMM in humanized mice. This novel OA model could constitute a useful tool to bridge the gap between the preclinical and clinical evaluation of immune interacting DMOADs and cell-based therapies.
Collapse
|
28
|
Wang G, Xing D, Liu W, Zhu Y, Liu H, Yan L, Fan K, Liu P, Yu B, Li JJ, Wang B. Preclinical studies and clinical trials on mesenchymal stem cell therapy for knee osteoarthritis: A systematic review on models and cell doses. Int J Rheum Dis 2022; 25:532-562. [PMID: 35244339 DOI: 10.1111/1756-185x.14306] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/30/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022]
Abstract
AIM To provide a systematic analysis of the study design in knee osteoarthritis (OA) preclinical studies, focusing on the characteristics of animal models and cell doses, and to compare these to the characteristics of clinical trials using mesenchymal stem cells (MSCs) for the treatment of knee OA. METHOD A systematic and comprehensive search was conducted using the PubMed, Web of Science, Ovid, and Embase electronic databases for research papers published in 2009-2020 on testing MSC treatment in OA animal models. The PubMed database and ClinicalTrials.gov website were used to search for published studies reporting clinical trials of MSC therapy for knee OA. RESULTS In total, 9234 articles and two additional records were retrieved, of which 120 studies comprising preclinical and clinical studies were included for analysis. Among the preclinical studies, rats were the most commonly used species for modeling knee OA, and anterior cruciate ligament transection was the most commonly used method for inducing OA. There was a correlation between the cell dose and body weight of the animal. In clinical trials, there was large variation in the dose of MSCs used to treat knee OA, ranging from 1 × 106 to 200 × 106 cells with an average of 37.91 × 106 cells. CONCLUSION Mesenchymal stem cells have shown great potential in improving pain relief and tissue protection in both preclinical and clinical studies of knee OA. Further high-quality preclinical and clinical studies are needed to explore the dose effectiveness relationship of MSC therapy and to translate the findings from preclinical studies to humans.
Collapse
Affiliation(s)
- Guishan Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China.,Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Beijing, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | - Yuanyuan Zhu
- Department of Pharmacy, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Haifeng Liu
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Lei Yan
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Kenan Fan
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Peidong Liu
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Baofeng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jiao Jiao Li
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Bin Wang
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China.,Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
29
|
Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthritis Cartilage 2022; 30:357-380. [PMID: 34536528 DOI: 10.1016/j.joca.2021.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is increasingly recognised as a disease of diverse phenotypes with variable clinical presentation, progression, and response to therapeutic intervention. This same diversity is readily apparent in the many animal models of OA. However, model selection, study design, and interpretation of resultant findings, are not routinely done in the context of the target human (or veterinary) patient OA sub-population or phenotype. This review discusses the selection and use of animal models of OA in discovery and therapeutic-development research. Beyond evaluation of the different animal models on offer, this review suggests focussing the approach to OA-animal model selection on study objective(s), alignment of available models with OA-patient sub-types, and the resources available to achieve valid and translatable results. How this approach impacts model selection is discussed and an experimental design checklist for selecting the optimal model(s) is proposed. This approach should act as a guide to new researchers and a reminder to those already in the field, as to issues that need to be considered before embarking on in vivo pre-clinical research. The ultimate purpose of using an OA animal model is to provide the best possible evidence if, how, when and where a molecule, pathway, cell or process is important in clinical disease. By definition this requires both model and study outcomes to align with and be predictive of outcomes in patients. Keeping this at the forefront of research using pre-clinical OA models, will go a long way to improving the quality of evidence and its translational value.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Australia.
| | - C L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Australia; Murray Maxwell Biomechanics Laboratory, The Kolling Institute, University of Sydney Faculty of Medicine and Health, At Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Australia.
| |
Collapse
|
30
|
Hodgkinson T, Amado IN, O'Brien FJ, Kennedy OD. The role of mechanobiology in bone and cartilage model systems in characterizing initiation and progression of osteoarthritis. APL Bioeng 2022. [DOI: 10.1063/5.0068277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Tom Hodgkinson
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Isabel N. Amado
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fergal J. O'Brien
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Oran D. Kennedy
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Townsend K, Imbert I, Eaton V, Stevenson GW, King T. Voluntary exercise blocks ongoing pain and diminishes bone remodeling while sparing protective mechanical pain in a rat model of advanced osteoarthritis pain. Pain 2022; 163:e476-e487. [PMID: 34224496 PMCID: PMC8712625 DOI: 10.1097/j.pain.0000000000002392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/18/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Exercise is the most common treatment recommended by healthcare providers for the treatment of musculoskeletal pain. We examined whether voluntary running wheel exercise improves pain and bone remodeling in rats with monosodium iodoacetate-induced unilateral knee joint pain. During acquisition of wheel running before osteoarthritis (OA) treatment, rats separated into 2 groups characterized by either high or low levels of voluntary wheel running as indicated by distance and peak speed. After the induction of knee joint OA, all rats showed diminished voluntary wheel running throughout the study. Voluntary wheel running failed to alter evoked nociceptive responses evaluated as weight asymmetry or hind paw tactile thresholds at any timepoint of the study. By contrast, relief of ongoing pain was demonstrated by conditioned place preference produced by lidocaine injection into the monosodium iodoacetate-treated knee in high but not low-running rats. Both high and low voluntary runners showed diminished trabecular bone loss compared with sedentary controls. These observations indicate that both high-intensity and low-intensity exercise is beneficial in protecting against bone remodeling in advanced OA. The data suggest that similar to clinical observation, bone remodeling does not correlate with pain. In addition, these results suggest that higher intensity exercise may relieve persistent ongoing OA pain while maintaining movement-evoked nociception. The relief of ongoing pain can potentially offer significant improvement in quality of life, whereas preservation of responses to movement-evoked pain may be especially important in protecting the joint from damage because of overuse.
Collapse
Affiliation(s)
- Kaylee Townsend
- Department of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
- Department of Psychology, College of Arts and Sciences, University of New England, Biddeford, ME, United States
| | - Ian Imbert
- Department of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
- Department of Psychology, College of Arts and Sciences, University of New England, Biddeford, ME, United States
| | - Victoria Eaton
- Department of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
- Department of Psychology, College of Arts and Sciences, University of New England, Biddeford, ME, United States
| | - Glenn W Stevenson
- Department of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
- Department of Psychology, College of Arts and Sciences, University of New England, Biddeford, ME, United States
| | - Tamara King
- Department of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
- Department of Psychology, College of Arts and Sciences, University of New England, Biddeford, ME, United States
| |
Collapse
|
32
|
Joint-on-chip platforms: entering a new era of in vitro models for arthritis. Nat Rev Rheumatol 2022; 18:217-231. [DOI: 10.1038/s41584-021-00736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
|
33
|
van Helvoort E, de Visser H, Lafeber F, Coeleveld K, Versteeg S, Weinans H, Popov-Celeketic J, Eijkelkamp N, Mastbergen S. IL4-10 Fusion Protein Shows DMOAD Activity in a Rat Osteoarthritis Model. Cartilage 2021; 13:1155S-1164S. [PMID: 34159843 PMCID: PMC8721682 DOI: 10.1177/19476035211026736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Ideally, disease-modifying osteoarthritis (OA) drugs (DMOAD) should combine chondroprotective, anti-inflammatory, and analgesic effects in a single molecule. A fusion protein of interleukin-4 (IL-4) and IL-10 (IL4-10 FP) possesses these combined effects. In this study, the DMOAD activity of rat IL4-10 FP (rIL4-10 FP) was tested in a rat model of surgically induced OA under metabolic dysregulation. DESIGN rIL4-10 FP was produced with HEK293F cells. Bioactivity of purified rIL4-10 FP was determined in a whole blood assay. Male Wistar rats (n = 20) were fed a high-fat diet (HFD) to induce metabolic dysregulation. After 12 weeks, OA was induced according to the Groove model. Two weeks after OA induction, rats were randomly divided into 2 groups and treated with 10 weekly, intra-articular injections of either rIL4-10 FP (n = 10) or phosphate buffered saline (PBS; n = 10). Possible antibody formation was evaluated using ELISA, cartilage degeneration and synovial inflammation were evaluated by histology and mechanical allodynia was evaluated using the von Frey test. RESULTS Intra-articular injections with rIL4-10 FP significantly reduced cartilage degeneration (P = 0.042) and decreased mechanical allodynia (P < 0.001) compared with PBS. Only mild synovial inflammation was found (nonsignificant), limiting detection of putative anti-inflammatory effects. Multiple injections of rIL4-10 FP did not induce antibodies against rIL4-10 FP. CONCLUSION rIL4-10 FP showed chondroprotective and analgesic activity in a rat OA model with moderate cartilage damage, mild synovial inflammation, and pain. Future studies will need to address whether less frequent intra-articular injections, for example, with formulations with increased residence time, would also lead to DMOAD activity.
Collapse
Affiliation(s)
- E.M. van Helvoort
- Department of Rheumatology &
Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht,
The Netherlands,E.M. van Helvoort, UMC Utrecht,
Rheumatology and Clinical Immunology, Postbus 85500, Internal Mail No. G02.232,
Utrecht, 3508 GA, The Netherlands.
| | - H.M. de Visser
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - F.P.J.G. Lafeber
- Department of Rheumatology &
Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht,
The Netherlands
| | - K. Coeleveld
- Department of Rheumatology &
Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht,
The Netherlands
| | - S. Versteeg
- Center for Translational Immunology,
University Medical Center Utrecht, Utrecht University, Utrecht, The
Netherlands
| | - H.H Weinans
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - J. Popov-Celeketic
- Department of Rheumatology &
Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht,
The Netherlands
| | - N. Eijkelkamp
- Center for Translational Immunology,
University Medical Center Utrecht, Utrecht University, Utrecht, The
Netherlands
| | - S.C. Mastbergen
- Department of Rheumatology &
Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht,
The Netherlands
| |
Collapse
|
34
|
Arzi B, Webb TL, Koch TG, Volk SW, Betts DH, Watts A, Goodrich L, Kallos MS, Kol A. Cell Therapy in Veterinary Medicine as a Proof-of-Concept for Human Therapies: Perspectives From the North American Veterinary Regenerative Medicine Association. Front Vet Sci 2021; 8:779109. [PMID: 34917671 PMCID: PMC8669438 DOI: 10.3389/fvets.2021.779109] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/13/2021] [Indexed: 01/27/2023] Open
Abstract
In the past decade, the potential to translate scientific discoveries in the area of regenerative therapeutics in veterinary species to novel, effective human therapies has gained interest from the scientific and public domains. Translational research using a One Health approach provides a fundamental link between basic biomedical research and medical clinical practice, with the goal of developing strategies for curing or preventing disease and ameliorating pain and suffering in companion animals and humans alike. Veterinary clinical trials in client-owned companion animals affected with naturally occurring, spontaneous disease can inform human clinical trials and significantly improve their outcomes. Innovative cell therapies are an area of rapid development that can benefit from non-traditional and clinically relevant animal models of disease. This manuscript outlines cell types and therapeutic applications that are currently being investigated in companion animals that are affected by naturally occurring diseases. We further discuss how such investigations impact translational efforts into the human medical field, including a critical evaluation of their benefits and shortcomings. Here, leaders in the field of veterinary regenerative medicine argue that experience gained through the use of cell therapies in companion animals with naturally occurring diseases represent a unique and under-utilized resource that could serve as a critical bridge between laboratory/preclinical models and successful human clinical trials through a One-Health approach.
Collapse
Affiliation(s)
- Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Tracy L Webb
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Thomas G Koch
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Susan W Volk
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - Dean H Betts
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Ashlee Watts
- Department of Large Animal Clinical Sciences, Veterinary Medicine and Biological Sciences, Texas A&M University, Killeen, TX, United States
| | - Laurie Goodrich
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Michael S Kallos
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, and Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Amir Kol
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
35
|
Zaki S, Smith MM, Little CB. Pathology-pain relationships in different osteoarthritis animal model phenotypes: it matters what you measure, when you measure, and how you got there. Osteoarthritis Cartilage 2021; 29:1448-1461. [PMID: 34332049 DOI: 10.1016/j.joca.2021.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine whether osteoarthritis (OA) pain characteristics and mechanistic pathways in pre-clinical models are phenotype-specific. DESIGN Male 11-week-old C57BL6 mice had unilateral medial-meniscal-destabilization (DMM) or antigen-induced-arthritis (AIA), vs sham-surgery/immunised-controls (Sham/Im-CT). Pain behaviour (allodynia, mechanical- and thermal-hyperalgesia, hindlimb static weight-bearing, stride-length) and lumbar dorsal root ganglia (DRG) gene-expression were measured at baseline, day-3, week-1/-2/-4/-8/-16, and pain-behaviour:gene-expression:joint-pathology associations investigated. RESULTS DMM and AIA induced structural OA defined by progressively increasing cartilage erosion, subchondral bone sclerosis and osteophyte size and maturation. All pain-behaviours were modified, with model-specific differences in severity and temporal pattern. Tactile allodynia developed acutely in both models and persisted to week-16. During early-OA (wk4-8) there was; reduced right hindlimb weight-bearing in AIA; thermal-hyperalgesia and reduced stride-length in DMM. During chronic-OA (wk12-16); mechanical-hyperalgesia and reduced right hindlimb weight-bearing were observed in DMM only. There were no associations in either model between different pain-behaviour outcomes. A coordinated DRG-expression profile was observed in sham and Im-CT for all 11 genes tested, but not in AIA and DMM. At wk-16 despite equivalent joint pathology, changes in DRG-expression (Calca, Trpa1, Trpv1, Trpv4) were observed only in DMM. In AIA mechanical-hyperalgesia was associated with Trpv1 (r = -0.79) and Il1b (r = 0.53). In DMM stride-length was associated with Calca, Tac1, Trpv1, Trpv2, Trpv4 and Adamts5 (r = 0.4-0.57). DRG gene-expression change was correlated with subchondral-bone sclerosis in DMM, and cartilage damage in AIA. Positive pain-behaviour:joint-pathology associations were only present in AIA - for synovitis, subchondral-bone resorption, chondrocyte-hypertrophy and cartilage damage. CONCLUSION Pain and peripheral sensory neuronal responses are OA-phenotype-specific with distinct pathology:pain-outcome:molecular-mechanism relationships.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| | - M M Smith
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| |
Collapse
|
36
|
Acevedo Rua L, Mumme M, Manferdini C, Darwiche S, Khalil A, Hilpert M, Buchner DA, Lisignoli G, Occhetta P, von Rechenberg B, Haug M, Schaefer DJ, Jakob M, Caplan A, Martin I, Barbero A, Pelttari K. Engineered nasal cartilage for the repair of osteoarthritic knee cartilage defects. Sci Transl Med 2021; 13:eaaz4499. [PMID: 34516821 DOI: 10.1126/scitranslmed.aaz4499] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lina Acevedo Rua
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcus Mumme
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Cristina Manferdini
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Salim Darwiche
- Musculoskeletal Research Unit MSRU, Equine Department, University of Zurich, 8057 Zürich, Switzerland
| | - Ahmad Khalil
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Morgane Hilpert
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Paola Occhetta
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Brigitte von Rechenberg
- Competence Center for Applied Biotechnology and Molecular Medicine CABMM, University of Zurich, 8057 Zürich, Switzerland
| | - Martin Haug
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Dirk J Schaefer
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcel Jakob
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Arnold Caplan
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14-16, 4123 Allschwil, Switzerland Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| |
Collapse
|
37
|
Aarntzen EHJG, Noriega-Álvarez E, Artiko V, Dias AH, Gheysens O, Glaudemans AWJM, Lauri C, Treglia G, van den Wyngaert T, van Leeuwen FWB, Terry SYA. EANM recommendations based on systematic analysis of small animal radionuclide imaging in inflammatory musculoskeletal diseases. EJNMMI Res 2021; 11:85. [PMID: 34487263 PMCID: PMC8421483 DOI: 10.1186/s13550-021-00820-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022] Open
Abstract
Inflammatory musculoskeletal diseases represent a group of chronic and disabling conditions that evolve from a complex interplay between genetic and environmental factors that cause perturbations in innate and adaptive immune responses. Understanding the pathogenesis of inflammatory musculoskeletal diseases is, to a large extent, derived from preclinical and basic research experiments. In vivo molecular imaging enables us to study molecular targets and to measure biochemical processes non-invasively and longitudinally, providing information on disease processes and potential therapeutic strategies, e.g. efficacy of novel therapeutic interventions, which is of complementary value next to ex vivo (post mortem) histopathological analysis and molecular assays. Remarkably, the large body of preclinical imaging studies in inflammatory musculoskeletal disease is in contrast with the limited reports on molecular imaging in clinical practice and clinical guidelines. Therefore, in this EANM-endorsed position paper, we performed a systematic review of the preclinical studies in inflammatory musculoskeletal diseases that involve radionuclide imaging, with a detailed description of the animal models used. From these reflections, we provide recommendations on what future studies in this field should encompass to facilitate a greater impact of radionuclide imaging techniques on the translation to clinical settings.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Medical Imaging, Radboud University Nijmegen Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Edel Noriega-Álvarez
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine, General University Hospital of Ciudad Real, Ciudad Real, Spain
| | - Vera Artiko
- Inflammation and Infection Committee EANM, Vienna, Austria
- Center for Nuclear Medicine Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000, Belgrade, Serbia
| | - André H Dias
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Olivier Gheysens
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc and Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Andor W J M Glaudemans
- Inflammation and Infection Committee EANM, Vienna, Austria.
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen Medical Imaging Center, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Chiara Lauri
- Inflammation and Infection Committee EANM, Vienna, Austria
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Giorgio Treglia
- Inflammation and Infection Committee EANM, Vienna, Austria
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Faculty of Biology and Medicine, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Tim van den Wyngaert
- Bone and Joint Committee EANM, Vienna, Austria
- Antwerp University Hospital Belgium, Edegem, Belgium
- Molecular Imaging Center Antwerp (MICA) - IPPON, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Fijs W B van Leeuwen
- Translational Molecular Imaging and Therapy Committee EANM, Vienna, Austria
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - Samantha Y A Terry
- Inflammation and Infection Committee EANM, Vienna, Austria.
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
38
|
Pirosa A, Tankus EB, Mainardi A, Occhetta P, Dönges L, Baum C, Rasponi M, Martin I, Barbero A. Modeling In Vitro Osteoarthritis Phenotypes in a Vascularized Bone Model Based on a Bone-Marrow Derived Mesenchymal Cell Line and Endothelial Cells. Int J Mol Sci 2021; 22:ijms22179581. [PMID: 34502489 PMCID: PMC8430538 DOI: 10.3390/ijms22179581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
Abstract
The subchondral bone and its associated vasculature play an important role in the onset of osteoarthritis (OA). Integration of different aspects of the OA environment into multi-cellular and complex human, in vitro models is therefore needed to properly represent the pathology. In this study, we exploited a mesenchymal stromal cell line/endothelial cell co-culture to produce an in vitro human model of vascularized osteogenic tissue. A cocktail of inflammatory cytokines, or conditioned medium from mechanically-induced OA engineered microcartilage, was administered to this vascularized bone model to mimic the inflamed OA environment, hypothesizing that these treatments could induce the onset of specific pathological traits. Exposure to the inflammatory factors led to increased network formation by endothelial cells, reminiscent of the abnormal angiogenesis found in OA subchondral bone, demineralization of the constructs, and increased collagen production, signs of OA related bone sclerosis. Furthermore, inflammation led to augmented expression of osteogenic (alkaline phosphatase (ALP) and osteocalcin (OCN)) and angiogenic (vascular endothelial growth factor (VEGF)) genes. The treatment, with a conditioned medium from the mechanically-induced OA engineered microcartilage, also caused increased demineralization and expression of ALP, OCN, ADAMTS5, and VEGF; however, changes in network formation by endothelial cells were not observed in this second case, suggesting a possible different mechanism of action in inducing OA-like phenotypes. We propose that this vascularized bone model could represent a first step for the in vitro study of bone changes under OA mimicking conditions and possibly serve as a tool in testing anti-OA drugs.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Esma Bahar Tankus
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Andrea Mainardi
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
- Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
| | - Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
| | - Laura Dönges
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Cornelia Baum
- Department of Orthopaedic Surgery and Traumatology, University Hospital Basel, 4031 Basel, Switzerland;
- Department of Research and Development, Schulthess Klinik Zurich, 8008 Zurich, Switzerland
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (P.O.); (M.R.)
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, 4056 Basel, Switzerland; (A.P.); (E.B.T.); (A.M.); (L.D.); (I.M.)
- Correspondence:
| |
Collapse
|
39
|
Samvelyan HJ, Hughes D, Stevens C, Staines KA. Models of Osteoarthritis: Relevance and New Insights. Calcif Tissue Int 2021; 109:243-256. [PMID: 32062692 PMCID: PMC8403120 DOI: 10.1007/s00223-020-00670-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/05/2020] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA) is a progressive and disabling musculoskeletal disease affecting millions of people and resulting in major healthcare costs worldwide. It is the most common form of arthritis, characterised by degradation of the articular cartilage, formation of osteophytes, subchondral sclerosis, synovial inflammation and ultimate loss of joint function. Understanding the pathogenesis of OA and its multifactorial aetiology will lead to the development of effective treatments, which are currently lacking. Two-dimensional (2D) in vitro tissue models of OA allow affordable, high-throughput analysis and stringent control over specific variables. However, they are linear in fashion and are not representative of physiological conditions. Recent in vitro studies have adopted three-dimensional (3D) tissue models of OA, which retain the advantages of 2D models and are able to mimic physiological conditions, thereby allowing investigation of additional variables including interactions between the cells and their surrounding extracellular matrix. Numerous spontaneous and induced animal models are used to reproduce the onset and monitor the progression of OA based on the aetiology under investigation. This therefore allows elucidation of the pathogenesis of OA and will ultimately enable the development of novel and specific therapeutic interventions. This review summarises the current understanding of in vitro and in vivo OA models in the context of disease pathophysiology, classification and relevance, thus providing new insights and directions for OA research.
Collapse
Affiliation(s)
| | - David Hughes
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh, UK
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh, UK
| | - Katherine Ann Staines
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh, UK.
| |
Collapse
|
40
|
Westhof A, Kleinschmidt-Doerr K, Michaelis M, Brenneis C. Dynamic weight-bearing test during jumping: A sensitive outcome measure of chronic osteoarthritis pain in rats. Heliyon 2021; 7:e07906. [PMID: 34522804 PMCID: PMC8427200 DOI: 10.1016/j.heliyon.2021.e07906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/22/2021] [Accepted: 08/30/2021] [Indexed: 11/06/2022] Open
Abstract
Pain due to osteoarthritis (OA) often occurs during locomotion in the vertical direction when joints are subjected to high mechanical load, e.g. during standing up from a chair or using stairs. To investigate joint pain in OA rat models, dynamic weight-bearing or gait analysis is traditionally conducted during horizontal walking on a flat surface. However, in chronic models of OA, which are of particular translational relevance for the disease, differences in the readouts between OA and control rats are often weak and of high variability leading to an insufficient assay window for drug profiling. To measure pain-related symptoms more sensitively, we conducted a dynamic weight-bearing test in the moment of a strong voluntary mechanical load. For that, we permanently housed rats in a four-story rat colony cage (RCC) and determined hind paw forces during voluntary jumping from one level to the next. This outcome measure was named jump incapacitance. After inducing OA by destabilizing the medial meniscus (DMM), we found that during jumps the average ipsilateral over contralateral hind paw forces were significantly reduced compared with healthy controls (jump incapacitance) from early- (day 7) to late-stage disease (day 90). An intra-articular injection of Zilretta (triamcinolone acetonide extended-release injectable suspension) attenuated OA-induced jump incapacitance after 8 days compared with DMM rats receiving vehicle (p = 0.069). In contrast, a CatWalk test for gait disturbance failed to detect any significant alterations in the chronic course of the DMM model. In conclusion, the dynamic weight-bearing test during jumping represents a novel method to characterize joint pain symptoms even in a slowly progressive OA rat model. It is sensitive, observer independent, relates to clinically relevant endpoints and demonstrates backtranslation of a drug that is approved for the treatment of OA knee pain.
Collapse
Affiliation(s)
- Andreas Westhof
- TIP Immunology, Innovation Cluster Osteoarthritis, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Martin Michaelis
- TIP Immunology, Innovation Cluster Osteoarthritis, Merck Healthcare KGaA, Darmstadt, Germany
| | - Christian Brenneis
- TIP Immunology, Innovation Cluster Osteoarthritis, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
41
|
Oh HK, Park M, Choi SW, Jeong DU, Kim BJ, Kim JA, Choi HJ, Lee J, Cho Y, Kim JH, Seong JK, Choi BH, Min BH, Kim DW. Suppression of Osteoarthritis progression by post-natal Induction of Nkx3.2. Biochem Biophys Res Commun 2021; 571:188-194. [PMID: 34330063 DOI: 10.1016/j.bbrc.2021.07.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 12/28/2022]
Abstract
Osteoarthritis (OA) is an incurable joint disease affecting 240 million elderly population, and major unmet medical needs exist for better therapeutic options for OA. During skeletal development, Nkx3.2 has been shown to promote chondrocyte differentiation and survival, but to suppress cartilage hypertrophy and blood vessel invasion. Here we show that Nkx3.2 plays a key role in osteoarthritis (OA) pathogenesis. Marked reduction of Nkx3.2 expression was observed in three different murine OA models. Consistent with these findings, analyses of surgery-induced and age-driven OA models revealed that cartilage-specific post-natal induction of Nkx3.2 can suppress OA progression in mice. These results suggest that Nkx3.2 may serve as a promising target for OA drug development.
Collapse
Affiliation(s)
- Hye-Kyoung Oh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Minsun Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Seung-Won Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Da-Un Jeong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Byoung Ju Kim
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul, Republic of Korea
| | - Jeong-Ah Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hye-Jeong Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jimin Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yongsik Cho
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jin-Hong Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea
| | - Byung Hyune Choi
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Republic of Korea
| | - Byoung-Hyun Min
- Department of Orthopedic Surgery, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Dae-Won Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Chang YM, Menges S, Westhof A, Kleinschmidt-Doerr K, Brenneis C, Pitsillides AA. Systematic analysis reveals that colony housing aligns gait profiles and strengthens link between histological and micro-CT bone markers in rat models of osteoarthritis. FASEB J 2021; 35:e21451. [PMID: 33683776 DOI: 10.1096/fj.202002009r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/01/2021] [Accepted: 02/01/2021] [Indexed: 11/11/2022]
Abstract
Osteoarthritis (OA) etiopathogenesis is complex with strong environmental/lifestyle determinants that, in laboratory animals, extend to social context and stress levels. This study seeks to identify whether colony housing of rats exerts a social impact on locomotion behaviors to influence alignment between symptomatic (gait) and structural (bone micro-CT measures, cartilage morphometry, and histology) OA outcome measures. Rats were randomly allocated to conventional (type IV; n = 48) or rat colony cage (RCC; n = 30) housing, further randomized to OA surgical models (ACLT + tMx, MMT or DMM) or no surgery (control), and maintained for 19 weeks during which multiple gait recordings were made. Standard histological grading and bone micro-CT data were collected at necropsy. Principal component analysis was used to summarize the variation in gait, micro-CT or histology. Linear mixed effects model or two-way ANOVA was employed to evaluate the impact of the housing system, surgery and time on gait, or micro-CT and histology components Analyses reveal that RCC exaggerates trends in gait change via a combined effect of the housing system and surgery. Intriguingly, RCC-housed nonoperated control rats showed similar gait changes to rats subjected to surgery; the latter exhibited significant structural joint changes in both systems. Stronger correlation between histological and micro-CT bone changes were found in medial and lateral tibia joint compartments of rats housed in RCC system. This study has established that rat social housing exaggerates outcomes in traditional histological measures of OA, generates stronger links between histology and micro-CT bone changes and removes gait differences as a variable in their etiology.
Collapse
Affiliation(s)
- Yu-Mei Chang
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | | | | | | | | | - Andrew A Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
43
|
Hart DA, Martin CR, Scott M, Shrive NG. The instrumented sheep knee to elucidate insights into osteoarthritis development and progression: A sensitive and reproducible platform for integrated research efforts. Clin Biomech (Bristol, Avon) 2021; 87:105404. [PMID: 34171651 DOI: 10.1016/j.clinbiomech.2021.105404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/12/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Osteoarthritis of the knee is a very common condition that has been difficult to treat. The majority of cases are considered idiopathic. Much research effort remains focused on biology rather than the biomechanics of such joints. Some new methods were developed and validated to better appreciate the subtleties of the biomechanical integrity of joints, and how changes in biomechanics can contribute to osteoarthritis. METHODS Over the past 15 years our lab has enhanced the sensitivity of the assessment of knee biomechanics of an instrumented, trained large animal model (sheep) of osteoarthritis and integrated the findings with biological and histological assessments. These new methods include gait analysis before and after injury followed by robotic validation post-sacrifice, and more recently using Fibre Bragg Grating sensors to detect alterations in cartilage stresses. RESULTS A review of the findings obtained with this model are presented. The findings indicate that sheep, like humans, exhibit individual characteristics. They also indicate that joint kinetics, rather than kinematics may better define the alterations induced by injury. With the addition of Fibre Bragg Grating sensors, it has been possible to measure with good accuracy, alterations to cartilage stresses following a controlled knee injury. INTERPRETATION Using this model as Proof of Concept, this sheep system can now be viewed as a sensitive platform to address many questions related to risk for development of idiopathic osteoarthritis of the human knee, the efficacy of potential interventions to correct biomechanical disruptions, and how joint biomechanics and biology are integrated during aging.
Collapse
Affiliation(s)
- David A Hart
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada; Department of Surgery, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada; Bone & Joint Health Strategic Clinical Network, Alberta Health Services, Edmonton, AB, Canada.
| | - C Ryan Martin
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada; Section of Orthopedics, Department of Surgery, University of Calgary, Calgary, AB, Canada
| | - Michael Scott
- Department of Veterinary Clinical & Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nigel G Shrive
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada; Department of Surgery, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada; Department of Civil Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
44
|
Tran T, Mach J, Gemikonakli G, Wu H, Allore H, Howlett SE, Little CB, Hilmer SN. Male-Female Differences In The Effects Of Age On Performance Measures Recorded For 23 Hours In Mice. J Gerontol A Biol Sci Med Sci 2021; 76:2141-2146. [PMID: 34171083 DOI: 10.1093/gerona/glab182] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Indexed: 11/13/2022] Open
Abstract
Functional independence is an important aspect of successful aging and differs with age and by sex in humans. Physical performance often declines earlier than other age-associated functional impairments. Rodent models are used to study pharmacological/toxicological effects of human therapies. However, physical outcomes in mice are usually assessed for short periods, with limited information on the influence of age and sex. Here, we investigated how age and sex affected murine physical performance over 23 hours of continuous observation. Young (3 months) and old (22 months) C57BL/6JArc male and female mice were assessed using the Laboratory Animal Behavior Observation, Registration, and Analysis System. Mice were individually housed for recording of distance travelled, mean gait speed, and durations of different physical activities. Compared to young mice of the same sex, old mice travelled significantly shorter distances with slower gait speeds, shorter durations of locomotion, rearing, climbing and immobility. Older mice groomed significantly more than young mice. Old females reared more during the light cycle than old males. Young females climbed substantially more than young males. Significant age*sex interactions were detected for rearing and climbing, whereby an age-related decline was greater in males than females. Our results suggest that old age reduces exploratory activities and increases grooming in mice. Age-related declines vary between sexes and tend to be greater in males. This non-invasive assessment can be applied to investigate how different interventions affect rodents of different ages and sexes, through the day-night cycle.
Collapse
Affiliation(s)
- Trang Tran
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St Leonards, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.,Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - John Mach
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St Leonards, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.,Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Gizem Gemikonakli
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St Leonards, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.,Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Harry Wu
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St Leonards, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.,Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Heather Allore
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States.,Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States
| | - Susan E Howlett
- Department of Pharmacology and Medicine (Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Christopher B Little
- Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.,Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Institute of Bone and Joint Research, Royal North Shore Hospital, University of Sydney, St Leonards, New South Wales, Australia
| | - Sarah N Hilmer
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St Leonards, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.,Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
45
|
Long wait times for knee and hip total joint replacement in Canada: An isolated health system problem, or a symptom of a larger problem? OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100141. [DOI: 10.1016/j.ocarto.2021.100141] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
|
46
|
Armstrong AR, Carlson CS, Rendahl AK, Loeser RF. Optimization of histologic grading schemes in spontaneous and surgically-induced murine models of osteoarthritis. Osteoarthritis Cartilage 2021; 29:536-546. [PMID: 33561541 PMCID: PMC8038967 DOI: 10.1016/j.joca.2021.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/28/2020] [Accepted: 01/12/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To compare the Osteoarthritis Research Society International (OARSI) and Articular Cartilage Structure (ACS) grading schemes applied to multiple and single sections, along with additional histologic measures, in two mouse models of Osteoarthritis (OA). METHODS Six coronal histologic stifle joint sections were collected from 40 C57BL/6J mice, including aged mice with spontaneous OA (approximately 18 months of age; n = 15) and young (12-week-old) mice that either underwent destabilization of the medial meniscus (DMM) surgery (n = 15) or sham surgery (n = 10). Sections were evaluated with the standard OARSI (0-6) scheme, a modified OARSI scheme, the ACS (0-12) scheme, histomorphometry of cartilage and bone, and scoring of osteophytes (0-3) and synovial hyperplasia (0-3). Principal components analysis (PCA) was used to determine the features explaining the greatest variability among the sections. RESULTS The grading schemes performed similarly when applied to a single mid-coronal section or six total coronal sections per joint. OARSI grading produced similar results when applied to hematoxylin and eosin or toluidine blue-stained sections. Aged mice had higher severity scores in the LTP than DMM mice (mid-coronal OARSI grade aged = 2.3 and DMM = 1.1, p = 0.0006; ACS grade aged = 4.1 and DMM = 1.6, p = 0.0024). PCA resulted in retention of four factors that accounted for 78.4% of the total variance. Factor 1 (36.4%) included the OARSI grade, ACS grade, Toluidine blue grade, articular cartilage area and thickness and the osteophyte grade. CONCLUSIONS Grading of a single mid-coronal section using either the OARSI or ACS schemes combined with osteophyte and histomorphometric measures can consistently define OA severity in mice.
Collapse
Affiliation(s)
- A R Armstrong
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, USA.
| | - C S Carlson
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, USA.
| | - A K Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, USA.
| | - R F Loeser
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
47
|
Neuroimmune interactions and osteoarthritis pain: focus on macrophages. Pain Rep 2021; 6:e892. [PMID: 33981927 PMCID: PMC8108586 DOI: 10.1097/pr9.0000000000000892] [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] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/01/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022] Open
Abstract
Bidirectional interactions between the immune system and the nervous system are increasingly appreciated as playing a pathogenic role in chronic pain. Unraveling the mechanisms by which inflammatory pain is mediated through communication between nerves and immune cells may lead to exciting new strategies for therapeutic intervention. In this narrative review, we focus on the role of macrophages in the pathogenesis of osteoarthritis (OA) pain. From regulating homeostasis to conducting phagocytosis, and from inducing inflammation to resolving it, macrophages are plastic cells that are highly adaptable to their environment. They rely on communicating with the environment through cytokines, growth factors, neuropeptides, and other signals to respond to inflammation or injury. The contribution of macrophages to OA joint damage has garnered much attention in recent years. Here, we discuss how macrophages may participate in the initiation and maintenance of pain in OA. We aim to summarize what is currently known about macrophages in OA pain and identify important gaps in the field to fuel future investigations.
Collapse
|
48
|
Effect of Glucosamine Sulfate Alone and Combined with Moderate Intensity Exercise on Serum Levels of CS 846 Epitope and Cartilage Oligomeric Matrix Protein in a Rat Model of Osteoarthritis. MEDICAL LABORATORY JOURNAL 2021. [DOI: 10.52547/mlj.15.2.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
Makarczyk MJ, Gao Q, He Y, Li Z, Gold MS, Hochberg MC, Bunnell BA, Tuan RS, Goodman SB, Lin H. Current Models for Development of Disease-Modifying Osteoarthritis Drugs. Tissue Eng Part C Methods 2021; 27:124-138. [PMID: 33403944 PMCID: PMC8098772 DOI: 10.1089/ten.tec.2020.0309] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling disease that affects millions of people worldwide. Symptom-alleviating treatments exist, although none with long-term efficacy. Furthermore, there are currently no disease-modifying OA drugs (DMOADs) with demonstrated efficacy in OA patients, which is, in part, attributed to a lack of full understanding of the pathogenesis of OA. The inability to translate findings from basic research to clinical applications also highlights the deficiencies in the available OA models at simulating the clinically relevant pathologies and responses to treatments in humans. In this review, the current status in the development of DMOADs will be first presented, with special attention to those in Phase II-IV clinical trials. Next, current in vitro, ex vivo, and in vivo OA models are summarized and the respective advantages and disadvantages of each are highlighted. Of note, the development and application of microphysiological or tissue-on-a-chip systems for modeling OA in humans are presented and the issues that need to be addressed in the future are discussed. Microphysiological systems should be given serious consideration for their inclusion in the DMOAD development pipeline, both for their ability to predict drug safety and efficacy in human clinical trials at present, as well as for their potential to serve as a test platform for personalized medicine. Impact statement At present, no disease-modifying osteoarthritis (OA) drugs (DMOADs) have been approved for widespread clinical use by regulatory bodies. The failure of developing effective DMOADs is likely owing to multiple factors, not the least of which are the intrinsic differences between the intact human knee joint and the preclinical models. This work summarizes the current OA models for the development of DMOADs, discusses the advantages/disadvantages of each, and then proposes future model development to aid in the discovery of effective and personalized DMOADs. The review also highlights the microphysiological systems, which are emerging as a new platform for drug development.
Collapse
Affiliation(s)
- Meagan J. Makarczyk
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, California, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhong Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael S. Gold
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark C. Hochberg
- Department of Medicine and Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, California, USA
- Department of Bioengineering, Stanford University, California, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
50
|
Ai X, Duan Y, Zhang Q, Sun D, Fang RH, Liu‐Bryan R, Gao W, Zhang L. Cartilage-targeting ultrasmall lipid-polymer hybrid nanoparticles for the prevention of cartilage degradation. Bioeng Transl Med 2021; 6:e10187. [PMID: 33532587 PMCID: PMC7823131 DOI: 10.1002/btm2.10187] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 08/30/2020] [Accepted: 08/30/2020] [Indexed: 12/21/2022] Open
Abstract
Current drug delivery approaches for the treatment of cartilage disorders such as osteoarthritis (OA) remain inadequate to achieve sufficient drug penetration and retention in the dense cartilage matrix. Herein, we synthesize sub-30 nm lipid-polymer hybrid nanoparticles functionalized with collagen-targeting peptides for targeted drug delivery to the cartilage. The nanoparticles consist of a polymeric core for drug encapsulation and a lipid shell modified with a collagen-binding peptide. By combining these design features, the nanoparticles can penetrate deep and accumulate preferentially in the cartilage. Using MK-8722, an activator of 5'-adenosine monophosphate-activated protein kinase (AMPK), as a model drug, the nanoparticles can encapsulate the drug molecules in high capacity and release them in a sustained and controllable manner. When injected into the knee joints of the mice with collagenase-induced OA, the drug-loaded nanoparticles can effectively reduce cartilage damage and alleviate the disease severity. Overall, the ultrasmall targeted nanoparticles represent a promising delivery platform to overcome barriers of dense tissues for the treatment of various indications, including cartilage disorders.
Collapse
Affiliation(s)
- Xiangzhao Ai
- Department of NanoEngineeringChemical Engineering Program, and Moores Cancer Center, University of California San DiegoLa JollaCaliforniaUSA
| | - Yaou Duan
- Department of NanoEngineeringChemical Engineering Program, and Moores Cancer Center, University of California San DiegoLa JollaCaliforniaUSA
| | - Qiangzhe Zhang
- Department of NanoEngineeringChemical Engineering Program, and Moores Cancer Center, University of California San DiegoLa JollaCaliforniaUSA
| | - Derrick Sun
- Department of NanoEngineeringChemical Engineering Program, and Moores Cancer Center, University of California San DiegoLa JollaCaliforniaUSA
| | - Ronnie H. Fang
- Department of NanoEngineeringChemical Engineering Program, and Moores Cancer Center, University of California San DiegoLa JollaCaliforniaUSA
| | - Ru Liu‐Bryan
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Weiwei Gao
- Department of NanoEngineeringChemical Engineering Program, and Moores Cancer Center, University of California San DiegoLa JollaCaliforniaUSA
| | - Liangfang Zhang
- Department of NanoEngineeringChemical Engineering Program, and Moores Cancer Center, University of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|