1
|
Aykora D, Taşçı B, Şahin MZ, Tekeoğlu I, Uzun M, Sarafian V, Docheva D. Tendon regeneration deserves better: focused review on In vivo models, artificial intelligence and 3D bioprinting approaches. Front Bioeng Biotechnol 2025; 13:1580490. [PMID: 40352349 PMCID: PMC12062838 DOI: 10.3389/fbioe.2025.1580490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Tendon regeneration has been one of the most challenging issues in orthopedics. Despite various surgical techniques and rehabilitation methods, tendon tears or ruptures cannot wholly regenerate and gain the load-bearing capacity the tendon tissue had before the injury. The enhancement of tendon regeneration mostly requires grafting or an artificial tendon-like tissue to replace the damaged tendon. Tendon tissue engineering offers promising regenerative effects with numerous techniques in the additive manufacturing context. 3D bioprinting is a widely used additive manufacturing method to produce tendon-like artificial tissues based on biocompatible substitutes. There are multiple techniques and bio-inks for fabricating innovative scaffolds for tendon applications. Nevertheless, there are still many drawbacks to overcome for the successful regeneration of injured tendon tissue. The most important target is to catch the highest similarity to the tissue requirements such as anisotropy, porosity, viscoelasticity, mechanical strength, and cell-compatible constructs. To achieve the best-designed artificial tendon-like structure, novel AI-based systems in the field of 3D bioprinting may unveil excellent final products to re-establish tendon integrity and functionality. AI-driven optimization can enhance bio-ink selection, scaffold architecture, and printing parameters, ensuring better alignment with the biomechanical properties of native tendons. Furthermore, AI algorithms facilitate real-time process monitoring and adaptive adjustments, improving reproducibility and precision in scaffold fabrication. Thus, in vitro biocompatibility and in vivo application-based experimental processes will make it possible to accelerate tendon healing and reach the required mechanical strength. Integrating AI-based predictive modeling can further refine these experimental processes to evaluate scaffold performance, cell viability, and mechanical durability, ultimately improving translation into clinical applications. Here in this review, 3D bioprinting approaches and AI-based technology incorporation were given in addition to in vivo models.
Collapse
Affiliation(s)
- Damla Aykora
- Health Services Vocational School, Department of Medical Services and Techniques, First and Emergency Aid, Bitlis Eren University, Bitlis, Türkiye
| | - Burak Taşçı
- Vocational School of Technical Sciences, Fırat University, Elazığ, Türkiye
| | - Muhammed Zahid Şahin
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Sakarya University Training and Research Hospital, Sakarya University, Sakarya, Türkiye
| | - Ibrahim Tekeoğlu
- Faculty of Medicine, Department of Internal Medicine, Department of Physical Medicine and Rehabilitation, Kütahya Health Sciences University, Kütahya, Türkiye
| | - Metehan Uzun
- Health Services Vocational School, Department of Medical Services and Techniques, First and Emergency Aid, Bitlis Eren University, Bitlis, Türkiye
| | - Victoria Sarafian
- Department of Medical Biology, Medical University-Plovdiv, Plovdiv, Bulgaria
- Department of Molecular and Regenerative Medicine, Research Institute at Medical University-Plovdiv, Plovdiv, Bulgaria
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, Julius-Maximilians-University Würzburg, Wuerzburg, Germany
| |
Collapse
|
2
|
Gao H, Wang L, Lyu Y, Jin H, Lin Z, Kang Y, Li Z, Zhang X, Jiang Y, Zhang G, Tao Z, Zhang X, Yang B, Bai X, Ma X, Liu S, Jiang J. The P2X7R/NLRP3 inflammasome axis suppresses enthesis regeneration through inflammatory and metabolic macrophage-stem cell cross-talk. SCIENCE ADVANCES 2025; 11:eadr4894. [PMID: 40279432 PMCID: PMC12024643 DOI: 10.1126/sciadv.adr4894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/21/2025] [Indexed: 04/27/2025]
Abstract
The regeneration of the enthesis remains a formidable challenge in regenerative medicine. However, key regulators underlying unsatisfactory regeneration remain poorly understood. This study reveals that the purinergic receptor P2X7 (P2X7R)/Nod-like receptor family protein 3 (NLRP3) inflammasome axis suppresses enthesis regeneration by amplifying IL-1β-mediated inflammatory cross-talk and suppressing docosatrienoic acid (DTA) metabolic cross-talk. NLRP3 inflammasomes were activated in macrophages following enthesis injury, thereby impairing the histological and functional recovery of the injured enthesis. Single-cell RNA sequencing (scRNA-seq) indicated that Nlrp3 knockout attenuated pathological inflammation and ameliorated the detrimental effects of IL-1β signaling cross-talk. Furthermore, NLRP3 inflammasomes suppressed the secretion of anti-inflammatory cytokines (IL-10 and IL-13) and DTA. The NLRP3 inflammasome-mediated secretome reduced differentiation and migration of stem cells. Neutralizing IL-1β or replenishing docosatrienoic acid accelerated enthesis regeneration. Moreover, conditional knockout of P2rx7 in myeloid cells attenuated NLRP3 inflammasome activation and facilitated enthesis regeneration. This study demonstrates that the P2X7R/NLRP3 inflammasome axis represents a promising therapeutic target for enthesis repair.
Collapse
Affiliation(s)
- Haihan Gao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Liren Wang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Yangbao Lyu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haocheng Jin
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhiqi Lin
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yuhao Kang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ziyun Li
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xueying Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yuhan Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Guoyang Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zaijin Tao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaofeng Zhang
- Department of Orthopedic Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201500, China
| | - Bin Yang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xingyu Bai
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xin Ma
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shen Liu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jia Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
3
|
Aykora D, Taşçı B, Şahin MZ, Tekeoğlu I, Uzun M, Sarafian V, Docheva D. Tendon regeneration deserves better: focused review on In vivo models, artificial intelligence and 3D bioprinting approaches. Front Bioeng Biotechnol 2025; 13. [DOI: doi.org/10.3389/fbioe.2025.1580490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Tendon regeneration has been one of the most challenging issues in orthopedics. Despite various surgical techniques and rehabilitation methods, tendon tears or ruptures cannot wholly regenerate and gain the load-bearing capacity the tendon tissue had before the injury. The enhancement of tendon regeneration mostly requires grafting or an artificial tendon-like tissue to replace the damaged tendon. Tendon tissue engineering offers promising regenerative effects with numerous techniques in the additive manufacturing context. 3D bioprinting is a widely used additive manufacturing method to produce tendon-like artificial tissues based on biocompatible substitutes. There are multiple techniques and bio-inks for fabricating innovative scaffolds for tendon applications. Nevertheless, there are still many drawbacks to overcome for the successful regeneration of injured tendon tissue. The most important target is to catch the highest similarity to the tissue requirements such as anisotropy, porosity, viscoelasticity, mechanical strength, and cell-compatible constructs. To achieve the best-designed artificial tendon-like structure, novel AI-based systems in the field of 3D bioprinting may unveil excellent final products to re-establish tendon integrity and functionality. AI-driven optimization can enhance bio-ink selection, scaffold architecture, and printing parameters, ensuring better alignment with the biomechanical properties of native tendons. Furthermore, AI algorithms facilitate real-time process monitoring and adaptive adjustments, improving reproducibility and precision in scaffold fabrication. Thus, in vitro biocompatibility and in vivo application-based experimental processes will make it possible to accelerate tendon healing and reach the required mechanical strength. Integrating AI-based predictive modeling can further refine these experimental processes to evaluate scaffold performance, cell viability, and mechanical durability, ultimately improving translation into clinical applications. Here in this review, 3D bioprinting approaches and AI-based technology incorporation were given in addition to in vivo models.
Collapse
|
4
|
Shang X, Chen C, Zhou J, Yang Y, Qu J. Muscle loading and endochondral ossification are involved in the regeneration of a fibrocartilaginous enthesis during tendon to bone healing in rabbits. BMC Musculoskelet Disord 2025; 26:277. [PMID: 40108557 PMCID: PMC11921697 DOI: 10.1186/s12891-025-08508-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE The purposes of the present study are to investigate the effects of reduced muscle loading by prolonged immobilization on the regeneration of fibrocartilaginous enthesis through endochondral ossification in rabbits. METHODS Forty-eight rabbits underwent standard partial patellectomy were randomly divided into the control group and the prolonged immobilization (PIM) group. The immobilized cast was only maintained for the first 4 weeks in the control group, while for the first 12 weeks or until euthanization in the PIM group. The Patella-patella tendon complexes were harvested for Micro-CT and histology at week 6, 12 and 18. RESULTS There was significantly lower bone volume in the PIM group than the control group at week 12, but not at week 6 and 18. At week 6, new bone was formed at the osteotomy site of the residual patella through endochondral ossification. At week 12, the chondrocytes in the tendon to bone interface were ordered and arranged in longitudinal rows separated by collagen fibres in the control group. While there were no visible fibers running continuously from tendon into bone in the PIM group. At week 18, a nearly normal fibrocartilaginous enthesis were regenerated in the control group. A similar fibrocartilaginous enthesis were also formed at the tendon to bone interface in the PIM group, but the four zones were not as distinct as that in the control group. CONCLUSION Muscle loading and endochondral ossification are involved in the regeneration of a fibrocartilaginous enthesis during tendon to bone healing in this partial patellectomy model.
Collapse
Affiliation(s)
- Xiaoke Shang
- Department of Orthopedic Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Can Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Jiefu Zhou
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Yang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Jin Qu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
5
|
Fang J, Wang X, Lai H, Li W, Yao X, Pan Z, Mao R, Yan Y, Xie C, Lin J, Sun W, Li R, Wang J, Dai J, Xu K, Yu X, Xu T, Duan W, Qian J, Ouyang H, Dai X. Decoding the mechanical characteristics of the human anterior cruciate ligament entheses through graduated mineralization interfaces. Nat Commun 2024; 15:9253. [PMID: 39462005 PMCID: PMC11513108 DOI: 10.1038/s41467-024-53542-5] [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: 11/30/2023] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
The anterior cruciate ligament is anchored to the femur and tibia via specialized interfaces known as entheses. These play a critical role in ligament homeostasis and joint stability by transferring forces, varying in magnitude and direction between structurally and functionally dissimilar tissues. However, the precise structural and mechanical characteristics underlying the femoral and tibial entheses and their intricate interplay remain elusive. In this study, two thin-graduated mineralization regions in the femoral enthesis (~21 μm) and tibial enthesis (~14 μm) are identified, both exhibiting distinct biomolecular compositions and mineral assembly patterns. Notably, the femoral enthesis interface exhibits progressively maturing hydroxyapatites, whereas the mineral at the tibial enthesis interface region transitions from amorphous calcium phosphate to hydroxyapatites with increasing crystallinity. Proteomics results reveal that Matrix Gla protein uniquely enriched at the tibial enthesis interface, may stabilize amorphous calcium phosphate, while C-type lectin domain containing 11 A, enriched at the femoral enthesis interface, could facilitate the interface mineralization. Moreover, the finite element analysis indicates that the femoral enthesis model exhibited higher resistance to shearing, whereas the tibial enthesis model contributes to tensile resistance, suggesting that the discrepancy in biomolecular expression and the corresponding mineral assembly heterogeneities collectively contribute to the superior mechanical properties of both the femoral enthesis and tibial enthesis models. These findings provide novel perspectives on the structure-function relationships of anterior cruciate ligament entheses, paving the way for improved management of anterior cruciate ligament injury and regeneration.
Collapse
Affiliation(s)
- Jinghua Fang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaozhao Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Huinan Lai
- Department of Engineering Mechanics, Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Wenyue Li
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xudong Yao
- Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Zongyou Pan
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Renwei Mao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yiyang Yan
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Chang Xie
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group, Hangzhou (CorMed), Hangzhou, China
| | - Junxin Lin
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Sun
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Rui Li
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group, Hangzhou (CorMed), Hangzhou, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jiacheng Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Kaiwang Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinning Yu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Wangping Duan
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jin Qian
- Department of Engineering Mechanics, Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang University, Hangzhou, China.
| | - Hongwei Ouyang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, China.
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.
- China Orthopedic Regenerative Medicine Group, Hangzhou (CorMed), Hangzhou, China.
| | - Xuesong Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China.
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
Muscat S, Nichols AEC. Leveraging in vivo animal models of tendon loading to inform tissue engineering approaches. Front Bioeng Biotechnol 2024; 12:1449372. [PMID: 39434716 PMCID: PMC11491380 DOI: 10.3389/fbioe.2024.1449372] [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: 06/14/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Tendon injuries disrupt successful transmission of force between muscle and bone, resulting in reduced mobility, increased pain, and significantly reduced quality of life for affected patients. There are currently no targeted treatments to improve tendon healing beyond conservative methods such as rest and physical therapy. Tissue engineering approaches hold great promise for designing instructive biomaterials that could improve tendon healing or for generating replacement graft tissue. More recently, engineered microphysiological systems to model tendon injuries have been used to identify therapeutic targets. Despite these advances, current tissue engineering efforts that aim to regenerate, replace, or model injured tendons have largely failed due in large part to a lack of understanding of how the mechanical environment of the tendon influences tissue homeostasis and how altered mechanical loading can promote or prevent disease progression. This review article draws inspiration from what is known about tendon loading from in vivo animal models and identifies key metrics that can be used to benchmark success in tissue engineering applications. Finally, we highlight important challenges and opportunities for the field of tendon tissue engineering that should be taken into consideration in designing engineered platforms to understand or improve tendon healing.
Collapse
Affiliation(s)
- Samantha Muscat
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
- Department of Orthopedics and Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Anne E. C. Nichols
- Department of Orthopedics and Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
7
|
Fagni F, Tascilar K, Noversa de Sousa R, Bayat S, Sollfrank L, Kleyer A, Sticherling M, Regensburger AP, Knieling F, Neurath MF, Schett G, Waldner M, Simon D. Unveiling Metabolic Similarities of Entheses in Patients with Psoriasis and Psoriatic Arthritis Using Noninvasive In Vivo Molecular Imaging: Results From a Cross-sectional Exploratory Study. Arthritis Rheumatol 2024; 76:1387-1396. [PMID: 38751108 DOI: 10.1002/art.42917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/02/2024] [Accepted: 05/06/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE We assessed and compared molecular tissue changes at the entheses in patients with psoriasis (PsO) and psoriatic arthritis (PsA) and in healthy controls (HCs) in vivo using multispectral optoacoustic tomography (MSOT) and described their relationship with clinical and ultrasound findings of enthesitis. METHODS A cross-sectional study (MSOT and Arthrosonography in PsA) in biologic disease-modifying antirheumatic drug-naïve patients with PsA and PsO and HCs was performed. Participants underwent clinical, ultrasonographic, and MSOT examination of six entheses (lateral humeral epicondyle, distal patellar tendon attachment, and Achilles tendon attachment). MSOT-measured hemoglobin (Hb), oxygen saturation (SO2), collagen, and lipid levels were quantified, and mean differences between groups were calculated using linear mixed effects models. MSOT-measured analytes were compared between entheses with and without clinical and ultrasound anomalies. RESULTS Ninety participants were included (30 PsO, 30 PsA, and 30 HCs), 540 entheses were clinically assessed, and 540 ultrasound and 830 MSOT scans were obtained. Patients with PsA and PsO showed increased oxygenated Hb (PsA: P = 0.003; PsO: P = 0.054) and SO2 (PsA: P < 0.001; PsO: P = 0.001) levels and decreased collagen signals (PsA: P < 0.001; PsO: P < 0.001) compared with HCs, with more pronounced changes in PsA. Significantly lower collagen levels (P = 0.01) and increased lipids (P = 0.03) were recorded in tender entheses compared with nontender ones. Erosions and enthesophytes on ultrasound were associated with significant differences in SO2 (P = 0.014) and lipid signals (P = 0.020), respectively. CONCLUSION Patients with PsA and PsO exhibit an analogous metabolic pattern at the entheses that is exacerbated in the presence of inflammation. These findings support the notion of a psoriatic disease spectrum characterized by common immunometabolic tissue changes.
Collapse
Affiliation(s)
- Filippo Fagni
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Koray Tascilar
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rita Noversa de Sousa
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sara Bayat
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lukas Sollfrank
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Sticherling
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Adrian P Regensburger
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ferdinand Knieling
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markus F Neurath
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maximilian Waldner
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Simon
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
8
|
Liang C, Fan Z, Zhang Z, Wang P, Deng H, Tao J. Electrospinning technology: a promising approach for tendon-bone interface tissue engineering. RSC Adv 2024; 14:26077-26090. [PMID: 39161449 PMCID: PMC11332360 DOI: 10.1039/d4ra04043k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/29/2024] [Indexed: 08/21/2024] Open
Abstract
The regeneration of tendon-bone interface tissue has become a topic of great interest in recent years. However, the complex nature of this interface has posed challenges in finding suitable solutions. Tissue engineering, with its potential to improve clinical outcomes and play a crucial role in musculoskeletal function, has been increasingly explored for tendon-bone interface regeneration. This review focuses on the research advancements of electrospinning technology in interface tissue engineering. By utilizing electrospinning, researchers have been able to fabricate scaffolds with tailored properties to promote the regeneration and integration of tendon and bone tissues. The review discusses the unique structure and function of the tendon-bone interface, the mechanisms involved in its healing, and the limitations currently faced in achieving successful regeneration. Additionally, it highlights the potential of electrospinning technology in scaffold fabrication and its role in facilitating the development of functional and integrated tendon-bone interface tissues. Overall, this review provides valuable insights into the application of electrospinning technology for tendon-bone interface tissue engineering, emphasizing its significance in addressing the challenges associated with regeneration in this complex interface.
Collapse
Affiliation(s)
- Chengzhi Liang
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Zaiwei Fan
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Zirui Zhang
- Department of Rehabilitation Medicine, The 960th Hospital of the Chinese People's Liberation Army Jinan 250000 China
| | - Pinkai Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Hui Deng
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Jun Tao
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| |
Collapse
|
9
|
Naredo E, D'Agostino MA, Terslev L, Pineda C, Miguel MI, Blasi J, Bruyn GA, Kortekaas MC, Mandl P, Nestorova R, Szkudlarek M, Todorov P, Vlad V, Wong P, Bakewell C, Filippucci E, Zabotti A, Micu M, Vreju F, Mortada M, Mendonça JA, Guillen-Astete CA, Olivas-Vergara O, Iagnocco A, Hanova P, Tinazzi I, Balint PV, Aydin SZ, Kane D, Keen H, Kaeley GS, Möller I. Validation and incorporation of digital entheses into a preliminary GLobal OMERACT Ultrasound DActylitis Score (GLOUDAS) in psoriatic arthritis. Ann Rheum Dis 2024; 83:1060-1071. [PMID: 38531611 DOI: 10.1136/ard-2023-225278] [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: 11/13/2023] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
OBJECTIVES The main objective was to generate a GLobal OMERACT Ultrasound DActylitis Score (GLOUDAS) in psoriatic arthritis and to test its reliability. To this end, we assessed the validity, feasibility and applicability of ultrasound assessment of finger entheses to incorporate them into the scoring system. METHODS The study consisted of a stepwise process. First, in cadaveric specimens, we identified enthesis sites of the fingers by ultrasound and gross anatomy, and then verified presence of entheseal tissue in histological samples. We then selected the entheses to be incorporated into a dactylitis scoring system through a Delphi consensus process among international experts. Next, we established and defined the ultrasound components of dactylitis and their scoring systems using Delphi methodology. Finally, we tested the interobserver and intraobserver reliability of the consensus- based scoring systemin patients with psoriatic dactylitis. RESULTS 32 entheses were identified in cadaveric fingers. The presence of entheseal tissues was confirmed in all cadaveric samples. Of these, following the consensus process, 12 entheses were selected for inclusion in GLOUDAS. Ultrasound components of GLOUDAS agreed on through the Delphi process were synovitis, tenosynovitis, enthesitis, subcutaneous tissue inflammation and periextensor tendon inflammation. The scoring system for each component was also agreed on. Interobserver reliability was fair to good (κ 0.39-0.71) and intraobserver reliability good to excellent (κ 0.80-0.88) for dactylitis components. Interobserver and intraobserver agreement for the total B-mode and Doppler mode scores (sum of the scores of the individual abnormalities) were excellent (interobserver intraclass correlation coefficient (ICC) 0.98 for B-mode and 0.99 for Doppler mode; intraobserver ICC 0.98 for both modes). CONCLUSIONS We have produced a consensus-driven ultrasound dactylitis scoring system that has shown acceptable interobserver reliability and excellent intraobserver reliability. Through anatomical knowledge, small entheses of the fingers were identified and histologically validated.
Collapse
Affiliation(s)
- Esperanza Naredo
- Department of Rheumatology and Joint and Bone Research Unit, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Madrid, Spain
- Autonomous University of Madrid, Madrid, Spain
| | - Maria Antonietta D'Agostino
- Department of Rheumatology, Università Cattolica del Sacro Cuore, Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Lene Terslev
- Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Pineda
- Division of Musculoskeletal and Rheumatic Disorders, Instituto Nacional de Rehabilitacion Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - M Isabel Miguel
- Human Anatomy and Embryology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences (Campus de Bellvitge), University of Barcelona, Barcelona, Spain
| | - Joan Blasi
- Histology Unit, Faculty of Medicine and Health Sciences (Campus de Bellvitge), University of Barcelona, Barcelona, Spain
| | - George A Bruyn
- Tergooi MC Hospital, Hilversum and Reumakliniek Lelystad, Lelystad, Netherlands
- Reumakliniek Flevoland, Lelystad, Netherlands
| | - Marion C Kortekaas
- Department of Rheumatology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Rheumatology, Flevoziekenhuis, Almere, The Netherlands
| | - Peter Mandl
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Wien, Austria
| | | | - Marcin Szkudlarek
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Rheumatology, Zealand's University Hospital, Køge, Denmark
| | - Plamen Todorov
- Department of Internal Disease Propaedeutics and Rheumatology, Medical University of Plovdiv, Clinic of Rheumatology, University Hospital "Kaspela", Plovdiv, Bulgaria
| | - Violeta Vlad
- Rheumatology, Clinical Hospital Sf Maria, Bucharest, Romania
| | - Priscilla Wong
- Virtus Medical Group, Hong Kong SAR, Hong Kong, Hong Kong
| | | | - Emilio Filippucci
- Rheumatology Unit, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Jesi, Italy
| | - Alen Zabotti
- Rheumatology Clinic, Department of Medicine, University of Udine, c/o Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Mihaela Micu
- Rheumatology Division, 2nd Rehabilitation Department, Spitalul Clinic de Recuperare Cluj-Napoca, Cluj-Napăoca, Romania
| | - Florentin Vreju
- Rheumatology Department, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Mohamed Mortada
- Rheumatology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - José Alexandre Mendonça
- Postgraduate Program in Health Sciences/Rheumatology/Ultrasonography Service, Pontifical Catholic University of Campinas, Sao Paulo, Brazil
| | | | - Otto Olivas-Vergara
- Department of Rheumatology and Joint and Bone Research Unit, Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Autonomous University, Madrid, Spain
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Department of Clinical and Biological Sciences, Università degli Studi di Torino, Turin, Italy
| | - Petra Hanova
- Department of Rheumatology, Institute of Rheumatology, Prague, Czech Republic
| | - Ilaria Tinazzi
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria, Negrar, Italy
| | - Peter V Balint
- 3rd Rheumatology Department, National Institute of Musculoskeletal Diseases, Budapest, Hungary
- Musculoskeletal Radiology Group, Medical Imaging Clinic, Semmelweis University, Budapest, Hungary
| | - Sibel Zehra Aydin
- Division of Rheumatology, University of Ottawa, the Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - David Kane
- Department of Rheumatology, Tallaght University Hospital, Dublin, Ireland
| | - Helen Keen
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Gurjit S Kaeley
- University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Ingrid Möller
- Human Anatomy and Embryology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences (Campus de Bellvitge), University of Barcelona, Barcelona, Spain
- Instituto Poal de Reumatología, Barcelona, Spain
| |
Collapse
|
10
|
Liu N, Jiang J, Liu T, Chen H, Jiang N. Compositional, Structural, and Biomechanical Properties of Three Different Soft Tissue-Hard Tissue Insertions: A Comparative Review. ACS Biomater Sci Eng 2024; 10:2659-2679. [PMID: 38697939 DOI: 10.1021/acsbiomaterials.3c01796] [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] [Indexed: 05/05/2024]
Abstract
Connective tissue attaches to bone across an insertion with spatial gradients in components, microstructure, and biomechanics. Due to regional stress concentrations between two mechanically dissimilar materials, the insertion is vulnerable to mechanical damage during joint movements and difficult to repair completely, which remains a significant clinical challenge. Despite interface stress concentrations, the native insertion physiologically functions as the effective load-transfer device between soft tissue and bone. This review summarizes tendon, ligament, and meniscus insertions cross-sectionally, which is novel in this field. Herein, the similarities and differences between the three kinds of insertions in terms of components, microstructure, and biomechanics are compared in great detail. This review begins with describing the basic components existing in the four zones (original soft tissue, uncalcified fibrocartilage, calcified fibrocartilage, and bone) of each kind of insertion, respectively. It then discusses the microstructure constructed from collagen, glycosaminoglycans (GAGs), minerals and others, which provides key support for the biomechanical properties and affects its physiological functions. Finally, the review continues by describing variations in mechanical properties at the millimeter, micrometer, and nanometer scale, which minimize stress concentrations and control stretch at the insertion. In summary, investigating the contrasts between the three has enlightening significance for future directions of repair strategies of insertion diseases and for bioinspired approaches to effective soft-hard interfaces and other tough and robust materials in medicine and engineering.
Collapse
Affiliation(s)
- Nian Liu
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610207, China
| | - Jialing Jiang
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610207, China
| | - Tiancheng Liu
- West China Hospital, Sichuan University, Chengdu, Sichuan 610207, China
| | - Haozhe Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Nan Jiang
- State Key Laboratory of Oral Diseases, & National Clinical Research Center for Oral Disease, & West China Hospital of Stomatology and the Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
11
|
Silverman AA, Olszewski JD, Siadat SM, Ruberti JW. Tension in the ranks: Cooperative cell contractions drive force-dependent collagen assembly in human fibroblast culture. MATTER 2024; 7:1533-1557. [PMID: 40213036 PMCID: PMC11984634 DOI: 10.1016/j.matt.2024.01.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Currently, there is no mechanistic model that fully explains the initial synthesis and organization of durable animal structure. As a result, our understanding of extracellular matrix (ECM) development and pathologies (e.g., persistent fibrosis) remains limited. Here, we identify and characterize cell-generated mechanical strains that direct the assembly of the ECM. Cell kinematics comprise cooperative retrograde "pulls" that organize and precipitate biopolymer structure along lines of tension. High-resolution optical microscopy revealed five unique classes of retrograde "pulls" that result in the production of filaments. Live-cell confocal imaging confirmed that retrograde pulls can directly cause the formation of fibronectin filaments that then colocalize with collagen aggregates exported from the cell, producing persistent elongated structures aligned with the direction of the tension. The findings suggest a new model for initial durable structure formation in animals. The results have important implications for ECM development and growth and life-threatening pathologies of the ECM, such as fibrosis.
Collapse
Affiliation(s)
| | | | | | - Jeffrey W. Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Lead contact
| |
Collapse
|
12
|
Steltzer SS, Abraham AC, Killian ML. Interfacial Tissue Regeneration with Bone. Curr Osteoporos Rep 2024; 22:290-298. [PMID: 38358401 PMCID: PMC11060924 DOI: 10.1007/s11914-024-00859-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE OF REVIEW Interfacial tissue exists throughout the body at cartilage-to-bone (osteochondral interface) and tendon-to-bone (enthesis) interfaces. Healing of interfacial tissues is a current challenge in regenerative approaches because the interface plays a critical role in stabilizing and distributing the mechanical stress between soft tissues (e.g., cartilage and tendon) and bone. The purpose of this review is to identify new directions in the field of interfacial tissue development and physiology that can guide future regenerative strategies for improving post-injury healing. RECENT FINDINGS Cues from interfacial tissue development may guide regeneration including biological cues such as cell phenotype and growth factor signaling; structural cues such as extracellular matrix (ECM) deposition, ECM, and cell alignment; and mechanical cues such as compression, tension, shear, and the stiffness of the cellular microenvironment. In this review, we explore new discoveries in the field of interfacial biology related to ECM remodeling, cellular metabolism, and fate. Based on emergent findings across multiple disciplines, we lay out a framework for future innovations in the design of engineered strategies for interface regeneration. Many of the key mechanisms essential for interfacial tissue development and adaptation have high potential for improving outcomes in the clinic.
Collapse
Affiliation(s)
- Stephanie S Steltzer
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Adam C Abraham
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Megan L Killian
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Song L, Golman M, Abraham AC, Zelzer E, Thomopoulos S. A role for TGFβ signaling in Gli1+ tendon and enthesis cells. FASEB J 2024; 38:e23568. [PMID: 38522021 PMCID: PMC10962263 DOI: 10.1096/fj.202301452r] [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: 07/16/2023] [Revised: 02/16/2024] [Accepted: 03/07/2024] [Indexed: 03/25/2024]
Abstract
The development of musculoskeletal tissues such as tendon, enthesis, and bone relies on proliferation and differentiation of mesenchymal progenitor cells. Gli1+ cells have been described as putative stem cells in several tissues and are presumed to play critical roles in tissue formation and maintenance. For example, the enthesis, a fibrocartilage tissue that connects tendon to bone, is mineralized postnatally by a pool of Gli1+ progenitor cells. These cells are regulated by hedgehog signaling, but it is unclear if TGFβ signaling, necessary for tenogenesis, also plays a role in their behavior. To examine the role of TGFβ signaling in Gli1+ cell function, the receptor for TGFβ, TbR2, was deleted in Gli1-lineage cells in mice at P5. Decreased TGFβ signaling in these cells led to defects in tendon enthesis formation by P56, including defective bone morphometry underlying the enthesis and decreased mechanical properties. Immunohistochemical staining of these Gli1+ cells showed that loss of TGFβ signaling reduced proliferation and increased apoptosis. In vitro experiments using Gli1+ cells isolated from mouse tail tendons demonstrated that TGFβ controls cell proliferation and differentiation through canonical and non-canonical pathways and that TGFβ directly controls the tendon transcription factor scleraxis by binding to its distant enhancer. These results have implications in the development of treatments for tendon and enthesis pathologies.
Collapse
Affiliation(s)
- Lee Song
- Department of Orthopedic Surgery, Columbia University, New York, NY10032, USA
| | - Mikhail Golman
- Department of Orthopedic Surgery, Columbia University, New York, NY10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY10027, USA
| | - Adam C. Abraham
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Israel
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY10027, USA
| |
Collapse
|
14
|
Camy C, Grünewald T, Lamy E, Roseren F, Caumes M, Fovet T, Brioche T, Genovesio C, Chopard A, Pithioux M, Roffino S. Characterization of the mechanical properties of the mouse Achilles tendon enthesis by microindentation. Effects of unloading and subsequent reloading. Bone Rep 2024; 20:101734. [PMID: 38292933 PMCID: PMC10825371 DOI: 10.1016/j.bonr.2024.101734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 12/14/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
The fibrocartilaginous tendon enthesis, i.e. the site where a tendon is attached to bone through a fibrocartilaginous tissue, is considered as a functionally graded interface. However, at local scale, a very limited number of studies have characterized micromechanical properties of this transitional tissue. The first goal of this work was to characterize the micromechanical properties of the mineralized part of the healthy Achilles tendon enthesis (ATE) through microindentation testing and to assess the degree of mineralization and of carbonation of mineral crystals by Raman spectroscopy. Since little is known about enthesis biological plasticity, our second objective was to examine the effects of unloading and reloading, using a mouse hindlimb-unloading model, on both the micromechanical properties and the mineral phase of the ATE. Elastic modulus, hardness, degree of mineralization, and degree of carbonation were assessed after 14 days of hindlimb suspension and again after a subsequent 6 days of reloading. The elastic modulus gradually increased along the mineralized part of the ATE from the tidemark to the subchondral bone, with the same trend being found for hardness. Whereas the degree of carbonation did not differ according to zone of measurement, the degree of mineralization increased by >70 % from tidemark to subchondral bone. Thus, the gradient in micromechanical properties is in part explained by a mineralization gradient. A 14-day unloading period did not appear to affect the gradient of micromechanical properties of the ATE, nor the degree of mineralization or carbonation. However, contrary to a short period of unloading, early return to normal mechanical load reduced the micromechanical properties gradient, regardless of carbonate-to-phosphate ratios, likely due to the more homogeneous degree of mineralization. These findings provide valuable data not only for tissue bioengineering, but also for musculoskeletal clinical studies and microgravity studies focusing on long-term space travel by astronauts.
Collapse
Affiliation(s)
- Claire Camy
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
| | - Tilman Grünewald
- Aix Marseille Univ, CNRS, Centrale Marseille, Institut Fresnel, Marseille, France
| | - Edouard Lamy
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
| | - Flavy Roseren
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
- Aix Marseille Univ, APHM, CNRS, ISM, Mecabio Platform, Department of Orthopaedics and Traumatology, 13009 Marseille, France
| | | | - Théo Fovet
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Thomas Brioche
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | | | - Angèle Chopard
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Martine Pithioux
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
- Aix Marseille Univ, APHM, CNRS, ISM, Mecabio Platform, Department of Orthopaedics and Traumatology, 13009 Marseille, France
- Aix Marseille Univ, APHM, CNRS, ISM, Sainte-Marguerite Hospital, Institute for Locomotion, Department of Orthopaedics and Traumatology, 13009 Marseille, France
| | | |
Collapse
|
15
|
Suito H, Fujikawa K, Ohsako M. ENPP1 downregulation and FGF23 upregulation in growth-related calcification of the tibial tuberosity in rats. J Anat 2024; 244:333-342. [PMID: 37814911 PMCID: PMC10780148 DOI: 10.1111/joa.13958] [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: 03/24/2023] [Revised: 08/15/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023] Open
Abstract
During tibial tuberosity growth, superficial and deep portions can be observed; however, the deep portion is not observed after the growth period, as it develops into bone tissues. Calcification in vivo is known to be constitutively suppressed by ectonucleotide pyrophosphatase/phosphodiesterase-1 (ENPP1) but promoted by tissue-nonspecific alkaline phosphatase (TNAP). FGF23 promotes calcification of enthesis. Gene expression of FGF23 increased rapidly at 13W in this study. Therefore, the tibial tuberosity is speculated to develop via Enpp1 downregulation and Tnap upregulation; however, the understanding of these processes remains unclear. Hence, in the present study, we aimed to explore the age-related structural changes and underlying gene expression changes in the tibial tuberosity of rats. Male Wistar rats were divided into three groups (3-, 7-, and 13-week-old; eight each). The tibial tuberosity superficial and deep portions were clearly observed in 3- and 7-week-old rats, but the presence of the deep portion was not confirmed in 13-week-old rats. The extracellular matrix of hypertrophic chondrocytes was calcified. Furthermore, the Enpp1 expression was the highest in 3-week-old rats and decreased with growth. The TNAP expression did not differ significantly among the groups. The deep portion area was significantly lower in 3-week-old rats than in 7-week-old rats. Generally, the extracellular matrix of the immature chondrocytes is not calcified. Therefore, we speculated that the cartilaginous tibial tuberosity calcifies and ossifies with growth. The Enpp1 expression decreased with growth, whereas the Tnap expression remained unchanged. Thus, we surmise that the tibial tuberosity calcifies with growth and that this process involves Enpp1 downregulation and FGF23 upregulation. As Osgood-Schlatter disease is closely related to the calcification of the tibial tuberosity, these findings may help clarify the pathogenesis of this disease.
Collapse
Affiliation(s)
- Hirai Suito
- Graduate School of Human Life Design, Toyo University, Tokyo, Japan
- Japan Society for the Promotion of Science Research Fellowships DC, Tokyo, Japan
| | - Kaoru Fujikawa
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, Tokyo, Japan
| | - Masafumi Ohsako
- Graduate School of Health and Sports Science, Toyo University, Tokyo, Japan
| |
Collapse
|
16
|
Nag S, De Bruyker I, Nelson A, Moody M, Fais M, Deymier AC. Acidosis induces significant changes to the murine supraspinatus enthesis organic matrix. Connect Tissue Res 2024; 65:41-52. [PMID: 37962089 DOI: 10.1080/03008207.2023.2275044] [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: 05/03/2023] [Accepted: 10/13/2023] [Indexed: 11/15/2023]
Abstract
Rotator cuff pathology is a common musculoskeletal condition that disproportionately affects older adults, as well as patients with diabetes mellitus and chronic kidney disease. It is known that increased age and kidney dysfunction have been correlated to acidotic states, which may be related to the increased incidence of rotator cuff injury. In order to investigate the potential relationship between acidosis and rotator cuff composition and mechanics, this study utilizes a 14-day murine model of metabolic acidosis and examines the effects on the supraspinatus tendon-humeral head attachment complex. The elastic matrix in the enthesis exhibited significant changes beginning at day 3 of acidosis exposure. At day 3 and day 7 timepoints, there was a decrease in collagen content seen in both mineralized and unmineralized tissue as well as a decrease in mineral:matrix ratio. There is also evidence of both mineral dissolution and reprecipitation as buffering ions continually promote pH homeostasis. Mechanical properties of the tendon-to-bone attachment were studied; however, no significant changes were elicited in this 14-day model of acidosis. These findings suggest that acidosis can result in significant changes in enthesis composition over the course of 14 days; however, enthesis mechanics may be more structurally mediated rather than affected by compositional changes.
Collapse
Affiliation(s)
- Saparja Nag
- School of Medicine, University of Connecticut, Farmington, CT, USA
| | | | - Ashley Nelson
- Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Mikayla Moody
- Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, USA
| | - Marla Fais
- Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Alix C Deymier
- Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
17
|
Brown ME, Puetzer JL. Enthesis maturation in engineered ligaments is differentially driven by loads that mimic slow growth elongation and rapid cyclic muscle movement. Acta Biomater 2023; 172:106-122. [PMID: 37839633 DOI: 10.1016/j.actbio.2023.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/17/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
Entheses are complex attachments that translate load between elastic-ligaments and stiff-bone via organizational and compositional gradients. Neither natural healing, repair, nor engineered replacements restore these gradients, contributing to high re-tear rates. Previously, we developed a culture system which guides ligament fibroblasts in high-density collagen gels to develop early postnatal-like entheses, however further maturation is needed. Mechanical cues, including slow growth elongation and cyclic muscle activity, are critical to enthesis development in vivo but these cues have not been widely explored in engineered entheses and their individual contribution to maturation is largely unknown. Our objective here was to investigate how slow stretch, mimicking ACL growth rates, and intermittent cyclic loading, mimicking muscle activity, individually drive enthesis maturation in our system so to shed light on the cues governing enthesis development, while further developing our tissue engineered replacements. Interestingly, we found these loads differentially drive organizational maturation, with slow stretch driving improvements in the interface/enthesis region, and cyclic load improving the ligament region. However, despite differentially affecting organization, both loads produced improvements to interface mechanics and zonal composition. This study provides insight into how mechanical cues differentially affect enthesis development, while producing some of the most organized engineered enthesis to date. STATEMENT OF SIGNIFICANCE: Entheses attach ligaments to bone and are critical to load transfer; however, entheses do not regenerate with repair or replacement, contributing to high re-tear rates. Mechanical cues are critical to enthesis development in vivo but their individual contribution to maturation is largely unknown and they have not been widely explored in engineered replacements. Here, using a novel culture system, we provide new insight into how slow stretch, mimicking ACL growth rates, and intermittent cyclic loading, mimicking muscle activity, differentially affect enthesis maturation in engineered ligament-to-bone tissues, ultimately producing some of the most organized entheses to date. This system is a promising platform to explore cues regulating enthesis formation so to produce functional engineered replacements and better drive regeneration following repair.
Collapse
Affiliation(s)
- M Ethan Brown
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, 23284, United States.
| |
Collapse
|
18
|
Chatterjee M, Evans MK, Bell R, Nguyen PK, Kamalitdinov TB, Korntner S, Kuo CK, Dyment NA, Andarawis-Puri N. Histological and immunohistochemical guide to tendon tissue. J Orthop Res 2023; 41:2114-2132. [PMID: 37321983 DOI: 10.1002/jor.25645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 06/02/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Tendons are unique dense connective tissues with discrete zones having specific structure and function. They are juxtaposed with other tissues (e.g., bone, muscle, and fat) with different compositional, structural, and mechanical properties. Additionally, tendon properties change drastically with growth and development, disease, aging, and injury. Consequently, there are unique challenges to performing high quality histological assessment of this tissue. To address this need, histological assessment was one of the breakout session topics at the 2022 Orthopaedic Research Society (ORS) Tendon Conference hosted at the University of Pennsylvania. The purpose of the breakout session was to discuss needs from members of the ORS Tendon Section related to histological procedures, data presentation, knowledge dissemination, and guidelines for future work. Therefore, this review provides a brief overview of the outcomes of this discussion and provides a set of guidelines, based on the perspectives from our laboratories, for histological assessment to assist researchers in their quest to utilize these techniques to enhance the outcomes and interpretations of their studies.
Collapse
Affiliation(s)
- Monideepa Chatterjee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Mary K Evans
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rebecca Bell
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| | - Phong K Nguyen
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| | - Timur B Kamalitdinov
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stefanie Korntner
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Catherine K Kuo
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Nathaniel A Dyment
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nelly Andarawis-Puri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
- Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
19
|
Subramanian A, Kanzaki LF, Schilling TF. Mechanical force regulates Sox9 expression at the developing enthesis. Development 2023; 150:dev201141. [PMID: 37497608 PMCID: PMC10445799 DOI: 10.1242/dev.201141] [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: 07/15/2022] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
Entheses transmit force from tendons and ligaments to the skeleton. Regional organization of enthesis extracellular matrix (ECM) generates differences in stiffness required for force transmission. Two key transcription factors co-expressed in entheseal tenocytes, scleraxis (Scx) and Sox9, directly control production of enthesis ECM components. Formation of embryonic craniofacial entheses in zebrafish coincides with onset of jaw movements, possibly in response to the force of muscle contraction. We show dynamic changes in scxa and sox9a mRNA levels in subsets of entheseal tenocytes that correlate with their roles in force transmission. We also show that transcription of a direct target of Scxa, Col1a, in enthesis ECM is regulated by the ratio of scxa to sox9a expression. Eliminating muscle contraction by paralyzing embryos during early stages of musculoskeletal differentiation alters relative levels of scxa and sox9a in entheses, primarily owing to increased sox9a expression. Force-dependent TGF-β (TGFβ) signaling is required to maintain this balance of scxa and sox9a expression. Thus, force from muscle contraction helps establish a balance of transcription factor expression that controls specialized ECM organization at the tendon enthesis and its ability to transmit force.
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Lauren F. Kanzaki
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
20
|
Abdalla AA, Pendegrass CJ. Biological approaches to the repair and regeneration of the rotator cuff tendon-bone enthesis: a literature review. BIOMATERIALS TRANSLATIONAL 2023; 4:85-103. [PMID: 38283917 PMCID: PMC10817785 DOI: 10.12336/biomatertransl.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/15/2023] [Accepted: 05/05/2023] [Indexed: 01/30/2024]
Abstract
Entheses are highly specialised organs connecting ligaments and tendons to bones, facilitating force transmission, and providing mechanical strengths to absorb forces encountered. Two types of entheses, fibrocartilaginous and fibrous, exist in interfaces. The gradual fibrocartilaginous type is in rotator cuff tendons and is more frequently injured due to the poor healing capacity that leads to loss of the original structural and biomechanical properties and is attributed to the high prevalence of retears. Fluctuating methodologies and outcomes of biological approaches are challenges to overcome for them to be routinely used in clinics. Therefore, stratifying the existing literature according to different categories (chronicity, extent of tear, and studied population) would effectively guide repair approaches. This literature review supports tissue engineering approaches to promote rotator cuff enthesis healing employing cells, growth factors, and scaffolds period. Outcomes suggest its promising role in animal studies as well as some clinical trials and that combination therapies are more beneficial than individualized ones. It then highlights the importance of tailoring interventions according to the tear extent, chronicity, and the population being treated. Contributing factors such as loading, deficiencies, and lifestyle habits should also be taken into consideration. Optimum results can be achieved if biological, mechanical, and environmental factors are approached. It is challenging to determine whether variations are due to the interventions themselves, the animal models, loading regimen, materials, or tear mechanisms. Future research should focus on tailoring interventions for different categories to formulate protocols, which would best guide regenerative medicine decision making.
Collapse
Affiliation(s)
- Ahlam A. Abdalla
- Institute of Sport, Exercise and Health (ISEH), Division of Surgery & Interventional Sciences, University College London, London, UK
| | - Catherine J. Pendegrass
- Department of Orthopaedics & Musculoskeletal Science, Division of Surgery & Interventional Sciences, University College London, Brockley Hill, Stanmore, UK
| |
Collapse
|
21
|
Ganji E, Lamia SN, Stepanovich M, Whyte N, Goulet RW, Abraham AC, Killian ML. Optogenetic-induced muscle loading leads to mechanical adaptation of the Achilles tendon enthesis in mice. SCIENCE ADVANCES 2023; 9:eadf4683. [PMID: 37352350 PMCID: PMC10289645 DOI: 10.1126/sciadv.adf4683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/17/2023] [Indexed: 06/25/2023]
Abstract
Skeletal shape depends on the transmission of contractile muscle forces from tendon to bone across the enthesis. Loss of muscle loading impairs enthesis development, yet little is known if and how the postnatal enthesis adapts to increased loading. Here, we studied adaptations in enthesis structure and function in response to increased loading, using optogenetically induced muscle contraction in young (i.e., growth) and adult (i.e., mature) mice. Daily bouts of unilateral optogenetic loading in young mice led to radial calcaneal expansion and warping. This also led to a weaker enthesis with increased collagen damage in young tendon and enthisis, with little change in adult mice. We then used RNA sequencing to identify the pathways associated with increased mechanical loading during growth. In tendon, we found enrichment of glycolysis, focal adhesion, and cell-matrix interactions. In bone, we found enrichment of inflammation and cell cycle. Together, we demonstrate the utility of optogenetic-induced muscle contraction to elicit in vivo adaptation of the enthesis.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Mathews Ave., Urbana, IL 61801, USA
- Department of Biomedical Engineering, University of Delaware, 540 S. College Ave., Newark, DE 19713, USA
| | - Syeda N. Lamia
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
- Department of Mechanical Engineering, University of Michigan, 2350 Hayward St., Ann Arbor, MI 48109, USA
| | - Matthew Stepanovich
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Noelle Whyte
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Robert W. Goulet
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Adam C. Abraham
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
| | - Megan L. Killian
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Delaware, 540 S. College Ave., Newark, DE 19713, USA
| |
Collapse
|
22
|
Tan J, Liu X, Zhou M, Wang F, Ma L, Tang H, He G, Kang X, Bian X, Tang K. Effect of treadmill training on fibrocartilage complex repair in tendon-bone insertion healing in the postinflammatory stage. Bone Joint Res 2023; 12:339-351. [PMID: 37219405 PMCID: PMC10204653 DOI: 10.1302/2046-3758.125.bjr-2022-0340.r2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Aims Mechanical stimulation is a key factor in the development and healing of tendon-bone insertion. Treadmill training is an important rehabilitation treatment. This study aims to investigate the benefits of treadmill training initiated on postoperative day 7 for tendon-bone insertion healing. Methods A tendon-bone insertion injury healing model was established in 92 C57BL/6 male mice. All mice were divided into control and training groups by random digital table method. The control group mice had full free activity in the cage, and the training group mice started the treadmill training on postoperative day 7. The quality of tendon-bone insertion healing was evaluated by histology, immunohistochemistry, reverse transcription quantitative polymerase chain reaction, Western blotting, micro-CT, micro-MRI, open field tests, and CatWalk gait and biomechanical assessments. Results Our results showed a significantly higher tendon-bone insertion histomorphological score in the training group, and the messenger RNA and protein expression levels of type II collagen (COL2A1), SOX9, and type X collagen (COL10A1) were significantly elevated. Additionally, tendon-bone insertion resulted in less scar hyperplasia after treadmill training, the bone mineral density (BMD) and bone volume/tissue volume (BV/TV) were significantly improved, and the force required to induce failure became stronger in the training group. Functionally, the motor ability, limb stride length, and stride frequency of mice with tendon-bone insertion injuries were significantly improved in the training group compared with the control group. Conclusion Treadmill training initiated on postoperative day 7 is beneficial to tendon-bone insertion healing, promoting biomechanical strength and motor function. Our findings are expected to guide clinical rehabilitation training programmes.
Collapse
Affiliation(s)
- Jindong Tan
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiao Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Mei Zhou
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Feng Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Lin Ma
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Hong Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Gang He
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xia Kang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xuting Bian
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Kanglai Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopaedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Ganji E, Lamia SN, Stepanovich M, Whyte N, Abraham AC, Killian ML. Optogenetic-Induced Muscle Loading Leads to Mechanical Adaptation of the Achilles Tendon Enthesis in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536376. [PMID: 37090593 PMCID: PMC10120626 DOI: 10.1101/2023.04.11.536376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The growth of the skeleton depends on the transmission of contractile muscle forces from tendon to bone across the extracellular matrix-rich enthesis. Loss of muscle loading leads to significant impairments in enthesis development. However, little is known about how the enthesis responds to increased loading during postnatal growth. To study the cellular and matrix adaptations of the enthesis in response to increased muscle loading, we used optogenetics to induce skeletal muscle contraction and unilaterally load the Achilles tendon and enthesis in young (i.e., during growth) and adult (i.e., mature) mice. In young mice, daily bouts of unilateral optogenetic loading led to expansion of the calcaneal apophysis and growth plate, as well as increased vascularization of the normally avascular enthesis. Daily loading bouts, delivered for 3 weeks, also led to a mechanically weaker enthesis with increased molecular-level accumulation of collagen damage in young mice. However, adult mice did not exhibit impaired mechanical properties or noticeable structural adaptations to the enthesis. We then focused on the transcriptional response of the young tendon and bone following optogenetic-induced loading. After 1 or 2 weeks of loading, we identified, in tendon, transcriptional activation of canonical pathways related to glucose metabolism (glycolysis) and inhibited pathways associated with cytoskeletal remodeling (e.g., RHOA and CREB signaling). In bone, we identified activation of inflammatory signaling (e.g., NFkB and STAT3 signaling) and inhibition of ERK/MAPK and PTEN signaling. Thus, we have demonstrated the utility of optogenetic-induced skeletal muscle contraction to elicit structural, functional, and molecular adaptation of the enthesis in vivo especially during growth.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Mathews Ave., Urbana, Illinois, 61801
| | - Syeda N Lamia
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
- Department of Mechanical Engineering, University of Michigan, 2350 Hayward St., Ann Arbor, Michigan, 48109
| | - Matthew Stepanovich
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
| | - Noelle Whyte
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
| | - Adam C Abraham
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
| | - Megan L Killian
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, 109 Zina Pitcher Pl., Ann Arbor, Michigan, 48109
- Department of Biomedical Engineering, University of Delaware, 540 S. College Ave., Newark, Delaware, 19713
| |
Collapse
|
24
|
Wang L, Li S, Xiao H, Zhang T, Liu Y, Hu J, Xu D, Lu H. TGF-β1 derived from macrophages contributes to load-induced tendon-bone healing in the murine rotator cuff repair model by promoting chondrogenesis. Bone Joint Res 2023; 12:219-230. [PMID: 37051812 PMCID: PMC10032229 DOI: 10.1302/2046-3758.123.bjr-2022-0368.r1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
It has been established that mechanical stimulation benefits tendon-bone (T-B) healing, and macrophage phenotype can be regulated by mechanical cues; moreover, the interaction between macrophages and mesenchymal stem cells (MSCs) plays a fundamental role in tissue repair. This study aimed to investigate the role of macrophage-mediated MSC chondrogenesis in load-induced T-B healing in depth. C57BL/6 mice rotator cuff (RC) repair model was established to explore the effects of mechanical stimulation on macrophage polarization, transforming growth factor (TGF)-β1 generation, and MSC chondrogenesis within T-B enthesis by immunofluorescence and enzyme-linked immunosorbent assay (ELISA). Macrophage depletion was performed by clodronate liposomes, and T-B healing quality was evaluated by histology and biomechanics. In vitro, bone marrow-derived macrophages (BMDMs) were stretched with CELLOAD-300 load system and macrophage polarization was identified by flow cytometry and quantitative real-time polymerase chain reaction (qRT-PCR). MSC chondrogenic differentiation was measured by histochemical analysis and qRT-PCR. ELISA and qRT-PCR were performed to screen the candidate molecules that mediated the pro-chondrogenic function of mechanical stimulated BMDMs. Mechanical stimulation promoted macrophage M2 polarization in vivo and in vitro. The conditioned media from mechanically stimulated BMDMs (MS-CM) enhanced MSC chondrogenic differentiation, and mechanically stimulated BMDMs generated more TGF-β1. Further, neutralizing TGF-β1 in MS-CM can attenuate its pro-chondrogenic effect. In vivo, mechanical stimulation promoted TGF-β1 generation, MSC chondrogenesis, and T-B healing, which were abolished following macrophage depletion. Macrophages subjected to appropriate mechanical stimulation could polarize toward the M2 phenotype and secrete TGF-β1 to promote MSC chondrogenesis, which subsequently augments T-B healing.
Collapse
Affiliation(s)
- Linfeng Wang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shengcan Li
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Han Xiao
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuqian Liu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jianzhong Hu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Daqi Xu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
Ganji E, Leek C, Duncan W, Patra D, Ornitz DM, Killian ML. Targeted deletion of Fgf9 in tendon disrupts mineralization of the developing enthesis. FASEB J 2023; 37:e22777. [PMID: 36734881 PMCID: PMC10108073 DOI: 10.1096/fj.202201614r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023]
Abstract
The enthesis is a transitional tissue between tendon and bone that matures postnatally. The development and maturation of the enthesis involve cellular processes likened to an arrested growth plate. In this study, we explored the role of fibroblast growth factor 9 (Fgf9), a known regulator of chondrogenesis and vascularization during bone development, on the structure and function of the postnatal enthesis. First, we confirmed spatial expression of Fgf9 in the tendon and enthesis using in situ hybridization. We then used Cre-lox recombinase to conditionally knockout Fgf9 in mouse tendon and enthesis (Scx-Cre) and characterized enthesis morphology as well as mechanical properties in Fgf9ScxCre and wild-type (WT) entheses. Fgf9ScxCre mice had smaller calcaneal and humeral apophyses, thinner cortical bone at the attachment, increased cellularity, and reduced failure load in mature entheses compared to WT littermates. During postnatal development, we found reduced chondrocyte hypertrophy and disrupted type X collagen (Col X) in Fgf9ScxCre entheses. These findings support that tendon-derived Fgf9 is important for functional development of the enthesis, including its postnatal mineralization. Our findings suggest the potential role of FGF signaling during enthesis development.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Orthopaedic Surgery, Michigan Medicine, Michigan, Ann Arbor, USA.,Department of Mechanical Engineering, University of Delaware, Delaware, Newark, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 61801, IL, Urbana, United States.,Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| | - Connor Leek
- Department of Orthopaedic Surgery, Michigan Medicine, Michigan, Ann Arbor, USA.,Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| | - William Duncan
- Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| | - Debabrata Patra
- Department of Developmental Biology, Washington University School of Medicine, Missouri, St Louis, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Missouri, St Louis, USA
| | - Megan L Killian
- Department of Orthopaedic Surgery, Michigan Medicine, Michigan, Ann Arbor, USA.,Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| |
Collapse
|
26
|
Lin AH, Slater CA, Martinez CJ, Eppell SJ, Yu SM, Weiss JA. Collagen fibrils from both positional and energy-storing tendons exhibit increased amounts of denatured collagen when stretched beyond the yield point. Acta Biomater 2023; 155:461-470. [PMID: 36400348 PMCID: PMC9805521 DOI: 10.1016/j.actbio.2022.11.018] [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: 07/14/2022] [Revised: 11/04/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
Collagen molecules are the base structural unit of tendons, which become denatured during mechanical overload. We recently demonstrated that during tendon stretch, collagen denaturation occurs at the yield point of the stress-strain curve in both positional and energy-storing tendons. We were interested in investigating how this load is transferred throughout the collagen hierarchy, and sought to determine the onset of collagen denaturation when collagen fibrils are stretched. Fibrils are one level above the collagen molecule in the collagen hierarchy, allowing more direct probing of the effect of strain on collagen molecules. We isolated collagen fibrils from both positional and energy-storing tendon types and stretched them using a microelectromechanical system device to various levels of strain. We stained the fibrils with fluorescently labeled collagen hybridizing peptides that specifically bind to denatured collagen, and examined whether samples stretched beyond the yield point of the stress-strain curve exhibited increased amounts of denatured collagen. We found that collagen denaturation in collagen fibrils from both tendon types occurs at the yield point. Greater amounts of denatured collagen were found in post-yield positional fibrils than in energy-storing fibrils. This is despite a greater yield strain and yield stress in fibrils from energy-storing tendons compared to positional tendons. Interestingly, the peak modulus of collagen fibrils from both tendon types was the same. These results are likely explained by the greater crosslink density found in energy-storing tendons compared to positional tendons. The insights gained from this study could help management of tendon and other musculoskeletal injuries by targeting collagen molecular damage at the fibril level. STATEMENT OF SIGNIFICANCE: When tendons are stretched or torn, this can lead to collagen denaturation (damage). Depending on their biomechanical function, tendons are considered positional or energy-storing with different crosslink profiles. By stretching collagen fibrils instead of fascicles from both tendon types, we can more directly examine the effect of tensile stretch on the collagen molecule in tendons. We found that regardless of tendon type, collagen denaturation in fibrils occurs when they are stretched beyond the yield point of the stress-strain curve. This provides insight into how load affects different tendon sub-structures during tendon injuries and failure, which will help clinicians and researchers understand mechanisms of injuries and potentially target collagen molecular damage as a treatment strategy, leading to improved clinical outcomes following injury.
Collapse
Affiliation(s)
- Allen H Lin
- Department of Biomedical Engineering, University of Utah, United States; Scientific Computing and Imaging Institute, University of Utah, United States
| | - Christopher A Slater
- Department of Biomedical Engineering, Case Western Reserve University, United States
| | - Callie-Jo Martinez
- Department of Biomedical Engineering, University of Utah, United States; Scientific Computing and Imaging Institute, University of Utah, United States
| | - Steven J Eppell
- Department of Biomedical Engineering, Case Western Reserve University, United States
| | - S Michael Yu
- Department of Biomedical Engineering, University of Utah, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, United States
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, United States; Scientific Computing and Imaging Institute, University of Utah, United States; Department of Orthopaedics, University of Utah, United States.
| |
Collapse
|
27
|
Zhang H, Ma Y, Wang Y, Niu L, Zou R, Zhang M, Liu H, Genin GM, Li A, Xu F. Rational Design of Soft-Hard Interfaces through Bioinspired Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204498. [PMID: 36228093 DOI: 10.1002/smll.202204498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Soft-hard tissue interfaces in nature present a diversity of hierarchical transitions in composition and structure to address the challenge of stress concentrations that would otherwise arise at their interface. The translation of these into engineered materials holds promise for improved function of biomedical interfaces. Here, soft-hard tissue interfaces found in the body in health and disease, and the application of the diverse, functionally graded, and hierarchical structures that they present to bioinspired engineering materials are reviewed. A range of such bioinspired engineering materials and associated manufacturing technologies that are on the horizon in interfacial tissue engineering, hydrogel bioadhesion at the interfaces, and healthcare and medical devices are described.
Collapse
Affiliation(s)
- Hui Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Min Zhang
- State Key Laboratory of Military Stomatology, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Hao Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
- NSF Science and Technology Center for Engineering MechanoBiology, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
28
|
Novel Use of Botulinum Toxin in Long-Standing Adductor-Related Groin Pain: A Case Series. Clin J Sport Med 2022; 32:567-573. [PMID: 36070357 DOI: 10.1097/jsm.0000000000001066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 07/28/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Adductor-related groin pain (ARGP) is the most common groin injury in athletes. If conservative treatment fails, then adductor tenotomy to relieve tension can be considered. The use of botulinum toxin A (BoNT-A) has shown good results in other musculoskeletal pathologies. OBJECTIVE Assess the effectiveness of BoNT-A injections in ARGP in cases where usual treatment has failed. DESIGN Retrospective cohort study. SETTING Orthopedic Medicine and Rehabilitation Unit, University of Bordeaux. PARTICIPANTS Fifty patients treated by BoNT-A injection in ARGP after failure of medical and/or surgical treatment were included in this study. INTERVENTIONS One or several adductor muscles were injected with BoNT-A, according to clinical evaluation using ultrasound and electrical stimulation guidance. Patients were followed up at 1, 3, 6, and 12 months. MAIN OUTCOME MEASURES The primary assessment criterion was the improvement of Hip and Groin Outcome Score subscales at day 30. Secondary outcomes included pain intensity and impact on sport, work, and quality of life (QoL), the Blazina scale, and side effects. RESULTS All the first 50 injected patients (45 male and 5 female patients) were included. A significant improvement was noted regarding the majority of Hip and Groin Outcome Score subscales at day 30 ( P < 0.05). Pain intensity and its impacts were both significantly reduced ( P < 0.001): less sport and professional disability and lower impact on QoL. Severity of symptoms assessed by the Blazina scale was significantly reduced ( P < 0.001). The improvements remained significant until 1-year postinjection. CONCLUSIONS BoNT-A is promising as a new treatment for ARGP but should be fully assessed in a randomized controlled trial.
Collapse
|
29
|
Liu Y, Wang L, Li S, Zhang T, Chen C, Hu J, Sun D, Lu H. Mechanical stimulation improves rotator cuff tendon-bone healing via activating IL-4/JAK/STAT signaling pathway mediated macrophage M2 polarization. J Orthop Translat 2022; 37:78-88. [PMID: 36262964 PMCID: PMC9550856 DOI: 10.1016/j.jot.2022.08.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/08/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Background It is well known that appropriate mechanical stimulation benefits tendon-bone (T-B) healing, however, the mechanisms behind this are still uncovered completely. Here, we aimed to explore whether the IL-4/JAK/STAT signaling pathway mediated macrophage polarization was involved in mechanical stimulation induced T-B healing. Method C57BL/6 mice rotator cuff (RC) repair model was established, and the mice were randomly allocated to the following group. 1. Mice were allowed for free cage activities after surgery (FC group); 2. Mice received treadmill running initiated on postoperative day 7 (TR group); 3. Mice only received a local injection of hydrogel containing IL-4 neutralizing antibody without postoperative intervention (FC + AF-404-SP group); 4. Mice received a local injection of hydrogel containing IL-4 neutralizing antibody and postoperative treadmill running (TR + AF-404-SP group). The expression of IL-4 within supraspinatus tendon (SST) enthesis was measured by Enzyme-linked immunosorbent assay (ELISA). In addition, the activation of JAK/STAT signaling pathway in macrophages and identification of macrophage phenotype at the RC insertion site was detected by Flow cytometry and qRT-PCR. T-B healing quality in this RC repair model was evaluated by histological staining, Micro-computed tomography (Micro-CT) scanning, and biomechanical testing. Result In this study, using the RC repair model, we confirmed that generation of IL-4, activation of the JAK/STAT signaling pathway in macrophages, the ability of macrophages to polarize towards M2 subtype, and T-B healing quality were significantly enhanced in TR group compared to FC group. When comparing FC + AF-404-SP group with TR + AF-404-SP group, it was found that the mechanical stimulation induced this effect was depleted following the blockade of the IL-4/JAK/STAT signaling pathway. Conclusion Our finding suggested that mechanical stimulation could accelerate T-B healing via activating the IL-4/JAK/STAT signaling pathway that modulates macrophages to polarize towards M2 subtype. The translational potential of this article This is the first study to reveal a significant role of mechanical stimulation in the IL-4/JAK/STAT signaling pathway activation and macrophage polarization during RC T-B healing, which highlights the IL-4/JAK/STAT signaling pathway as a potential target to mediate macrophage M2 polarization and improves T-B healing for RC repair.
Collapse
Affiliation(s)
- Yuqian Liu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Linfeng Wang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shengcan Li
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Can Chen
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jianzhong Hu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Spine Surgery and Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Deyi Sun
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
30
|
Camy C, Brioche T, Senni K, Bertaud A, Genovesio C, Lamy E, Fovet T, Chopard A, Pithioux M, Roffino S. Effects of hindlimb unloading and subsequent reloading on the structure and mechanical properties of Achilles tendon-to-bone attachment. FASEB J 2022; 36:e22548. [PMID: 36121701 DOI: 10.1096/fj.202200713r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/10/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
While muscle and bone adaptations to deconditioning have been widely described, few studies have focused on the tendon enthesis. Our study examined the effects of mechanical loading on the structure and mechanical properties of the Achilles tendon enthesis. We assessed the fibrocartilage surface area, the organization of collagen, the expression of collagen II, the presence of osteoclasts, and the tensile properties of the mouse enthesis both after 14 days of hindlimb suspension (HU) and after a subsequent 6 days of reloading. Although soleus atrophy was severe after HU, calcified fibrocartilage (CFc) was a little affected. In contrast, we observed a decrease in non-calcified fibrocartilage (UFc) surface area, collagen fiber disorganization, modification of morphological characteristics of the fibrocartilage cells, and altered collagen II distribution. Compared to the control group, restoring normal loads increased both UFc surface area and expression of collagen II, and led to a crimp pattern in collagen. Reloading induced an increase in CFc surface area, probably due to the mineralization front advancing toward the tendon. Functionally, unloading resulted in decreased enthesis stiffness and a shift in site of failure from the osteochondral interface to the bone, whereas 6 days of reloading restored the original elastic properties and site of failure. In the context of spaceflight, our results suggest that care must be taken when performing countermeasure exercises both during missions and during the return to Earth.
Collapse
Affiliation(s)
- Claire Camy
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France
| | - Thomas Brioche
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Karim Senni
- Laboratoire EBInnov, Ecole de Biologie Industrielle-EBI, Cergy, France
| | - Alexandrine Bertaud
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France.,Laboratoire de Biochimie, Faculté de Pharmacie, Marseille, France
| | - Cécile Genovesio
- Laboratoire de Biochimie, Faculté de Pharmacie, Marseille, France
| | - Edouard Lamy
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France.,Laboratoire de Biochimie, Faculté de Pharmacie, Marseille, France
| | - Théo Fovet
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Angèle Chopard
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Martine Pithioux
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France.,Department of Orthopaedics and Traumatology, Aix Marseille Univ, APHM, CNRS, ISM, Sainte-Marguerite Hospital, Institute for Locomotion, Marseille, France.,Aix Marseille Univ, APHM, CNRS, Centrale Marseille, ISM, Mecabio Platform, Anatomy Laboratory, Timone, Marseille, France
| | - Sandrine Roffino
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France.,Aix Marseille Univ, APHM, CNRS, Centrale Marseille, ISM, Mecabio Platform, Anatomy Laboratory, Timone, Marseille, France
| |
Collapse
|
31
|
Muscular hydraulics drive larva-polyp morphogenesis. Curr Biol 2022; 32:4707-4718.e8. [PMID: 36115340 DOI: 10.1016/j.cub.2022.08.065] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 07/14/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022]
Abstract
Development is a highly dynamic process in which organisms often experience changes in both form and behavior, which are typically coupled to each other. However, little is known about how organismal-scale behaviors such as body contractility and motility impact morphogenesis. Here, we use the cnidarian Nematostella vectensis as a developmental model to uncover a mechanistic link between organismal size, shape, and behavior. Using quantitative live imaging in a large population of developing animals, combined with molecular and biophysical experiments, we demonstrate that the muscular-hydraulic machinery that controls body movement also drives larva-polyp morphogenesis. We show that organismal size largely depends on cavity inflation through fluid uptake, whereas body shape is constrained by the organization of the muscular system. The generation of ethograms identifies different trajectories of size and shape development in sessile and motile animals, which display distinct patterns of body contractions. With a simple theoretical model, we conceptualize how pressures generated by muscular hydraulics can act as a global mechanical regulator that coordinates tissue remodeling. Altogether, our findings illustrate how organismal contractility and motility behaviors can influence morphogenesis.
Collapse
|
32
|
Vinestock RC, Felsenthal N, Assaraf E, Katz E, Rubin S, Heinemann-Yerushalmi L, Krief S, Dezorella N, Levin-Zaidman S, Tsoory M, Thomopoulos S, Zelzer E. Neonatal Enthesis Healing Involves Noninflammatory Acellular Scar Formation through Extracellular Matrix Secretion by Resident Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1122-1135. [PMID: 35659946 PMCID: PMC9379688 DOI: 10.1016/j.ajpath.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Wound healing typically recruits the immune and vascular systems to restore tissue structure and function. However, injuries to the enthesis, a hypocellular and avascular tissue, often result in fibrotic scar formation and loss of mechanical properties, severely affecting musculoskeletal function and life quality. This raises questions about the healing capabilities of the enthesis. Herein, this study established an injury model to the Achilles entheses of neonatal mice to study the effectiveness of early-age enthesis healing. Histology and immunohistochemistry analyses revealed an atypical process that did not involve inflammation or angiogenesis. Instead, healing was mediated by secretion of collagen types I and II by resident cells, which formed a permanent hypocellular and avascular scar. Transmission electron microscopy showed that the cellular response to injury, including endoplasmic reticulum stress, autophagy, and cell death, varied between the tendon and cartilage ends of the enthesis. Single-molecule in situ hybridization, immunostaining, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays verified these differences. Finally, gait analysis showed that these processes effectively restored function of the injured leg. These findings reveal a novel healing mechanism in neonatal entheses, whereby local extracellular matrix secretion by resident cells forms an acellular extracellular matrix deposit without inflammation, allowing gait restoration. These insights into the healing mechanism of a complex transitional tissue may lead to new therapeutic strategies for adult enthesis injuries.
Collapse
Affiliation(s)
- Ron C Vinestock
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Assaraf
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Katz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nili Dezorella
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Smadar Levin-Zaidman
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, New York; Department of Biomedical Engineering, Columbia University, New York, New York
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
33
|
Donderwinkel I, Tuan RS, Cameron NR, Frith JE. Tendon tissue engineering: Current progress towards an optimized tenogenic differentiation protocol for human stem cells. Acta Biomater 2022; 145:25-42. [PMID: 35470075 DOI: 10.1016/j.actbio.2022.04.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 12/19/2022]
Abstract
Tendons are integral to our daily lives by allowing movement and locomotion but are frequently injured, leading to patient discomfort and impaired mobility. Current clinical procedures are unable to fully restore the native structure of the tendon, resulting in loss of full functionality, and the weakened tissue following repair often re-ruptures. Tendon tissue engineering, involving the combination of cells with biomaterial scaffolds to form new tendon tissue, holds promise to improve patient outcomes. A key requirement for efficacy in promoting tendon tissue formation is the optimal differentiation of the starting cell populations, most commonly adult tissue-derived mesenchymal stem/stromal cells (MSCs), into tenocytes, the predominant cellular component of tendon tissue. Currently, a lack of consensus on the protocols for effective tenogenic differentiation is hampering progress in tendon tissue engineering. In this review, we discuss the current state of knowledge regarding human stem cell differentiation towards tenocytes and tendon tissue formation. Tendon development and healing mechanisms are described, followed by a comprehensive overview of the current protocols for tenogenic differentiation, including the effects of biochemical and biophysical cues, and their combination, on tenogenesis. Lastly, a synthesis of the key features of these protocols is used to design future approaches. The holistic evaluation of current knowledge should facilitate and expedite the development of efficacious stem cell tenogenic differentiation protocols with future impact in tendon tissue engineering. STATEMENT OF SIGNIFICANCE: The lack of a widely-adopted tenogenic differentiation protocol has been a major hurdle in the tendon tissue engineering field. Building on current knowledge on tendon development and tendon healing, this review surveys peer-reviewed protocols to present a holistic evaluation and propose a pathway to facilitate and expedite the development of a consensus protocol for stem cell tenogenic differentiation and tendon tissue engineering.
Collapse
|
34
|
Peng Y, Li X, Wu W, Ma H, Wang G, Jia S, Zheng C. Effect of Mechanical Stimulation Combined With Platelet-Rich Plasma on Healing of the Rotator Cuff in a Murine Model. Am J Sports Med 2022; 50:1358-1368. [PMID: 35188809 DOI: 10.1177/03635465211073339] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Mechanical stimulation and platelet-rich plasma (PRP) have been shown to be beneficial for healing of the bone-tendon interface (BTI), but few studies have explored the efficacy of a combination of these applications. We investigated the effect of mechanical stimulation combined with PRP on rotator cuff repair in mice. HYPOTHESIS Mechanical stimulation combined with PRP can enhance BTI healing in a murine model of rotator cuff repair. STUDY DESIGN Controlled laboratory study. METHODS A total of 160 C57BL/6 mice were used. Overall, 40 mice were used to prepare PRP, while 120 mice underwent acute supraspinatus tendon (SST) repair. The animals were randomly assigned to 4 groups: control group, mechanical stimulation group, PRP group, and mechanical stimulation combined with PRP group (combination group). At 4 and 8 weeks postoperatively, animals were sacrificed, the eyeballs were removed to collect blood, and the SST-humeral complexes were collected. Histological, biomechanical, immunological, and bone morphometric tests were performed. RESULTS Histologically, at 4 and 8 weeks after surgery, the area of the fibrocartilage layer at the BTI in the combination group was larger than in the other groups. The content and distribution of proteoglycans in this layer in the combination group were significantly greater than in the other groups. At 8 weeks postoperatively, trabecular number, and trabecular bone thickness of the subchondral bone area of interest at the BTI of the combination group were greater than those of the other groups, bone volume fraction of the combination group was greater than the control group. On biomechanical testing at 4 and 8 weeks after surgery, the failure load and ultimate strength of the SST-humeral complex in the combination group were higher than in the other groups. Enzyme-linked immunosorbent assay results showed that, at 4 weeks postoperatively, the serum concentrations of transforming growth factor beta 1 and platelet-derived growth factor (PDGF) in the combination group were significantly higher than in the other groups; at 8 weeks, the PDGF-AB concentration in the combination group was higher than in the control and mechanical stimulation groups. CONCLUSION Mechanical stimulation combined with PRP can effectively promote the early stage of healing after a rotator cuff injury. CLINICAL RELEVANCE These findings imply that mechanical stimulation combined with PRP can serve as a potential therapeutic strategy for rotator cuff healing.
Collapse
Affiliation(s)
- Yundong Peng
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Xiaomei Li
- College of Health Science, Wuhan Sports University, Wuhan, China.,Medical College, Huainan Union University, Huainan, China
| | - Wenxia Wu
- College of Health Science, Wuhan Sports University, Wuhan, China.,Department of Rehabilitation Therapy, Jinci College of Shanxi Medical University, Taiyuan, China
| | - Haozhe Ma
- College of International Education, Wuhan Sports University, Wuhan, China
| | - Guanglan Wang
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Shaohui Jia
- Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Cheng Zheng
- Department of Sports Medicine, Affiliated Hospital, Wuhan Sports University, Wuhan, China
| |
Collapse
|
35
|
Abraham AC, Fang F, Golman M, Oikonomou P, Thomopoulos S. The role of loading in murine models of rotator cuff disease. J Orthop Res 2022; 40:977-986. [PMID: 34081350 PMCID: PMC8639823 DOI: 10.1002/jor.25113] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/07/2021] [Accepted: 05/31/2021] [Indexed: 02/04/2023]
Abstract
Rotator cuff disease pathogenesis is associated with intrinsic (e.g., age, joint laxity, muscle weakness) and extrinsic (e.g., mechanical load, fatigue) factors that lead to chronic degeneration of the cuff tissues. However, etiological studies are difficult to perform in patients due to the long duration of disease onset and progression. Therefore, the purpose of this study was to determine the effects of altered joint loading on the rotator cuff. Mice were subjected to one of three load-dependent rotator cuff tendinopathy models: underuse loading, achieved by injecting botulinum toxin-A into the supraspinatus muscle; overuse loading, achieved using downhill treadmill running; destabilization loading, achieved by surgical excision of the infraspinatus tendon. All models were compared to cage activity animals. Whole joint function was assessed longitudinally using gait analysis. Tissue-scale structure and function were determined using microCT, tensile testing, and histology. The molecular response of the supraspinatus tendon and enthesis was determined by measuring the expression of 84 wound healing-associated genes. Underuse and destabilization altered forepaw weight-bearing, decreased tendon-to-bone attachment strength, decreased mineral density of the humeral epiphysis, and reduced tendon strength. Transcriptional activity of the underuse group returned to baseline levels by 4 weeks, while destabilization had significant upregulation of inflammation, growth factors, and extracellular matrix remodeling genes. Surprisingly, overuse activity caused changes in walking patterns, increased tendon stiffness, and primarily suppressed expression of wound healing-related genes. In summary, the tendinopathy models demonstrated how divergent muscle loading can result in clinically relevant alterations in rotator cuff structure, function, and gene expression.
Collapse
Affiliation(s)
- Adam C. Abraham
- University of Michigan, Department of Orthopaedic Surgery, Biomedical Science Research Building, 109 Zina Pitcher Pl, Ann Arbor, MI 48109, USA,Corresponding author Adam C. Abraham, Ph.D., Research Investigator, University of Michigan, Department of Orthopaedic Surgery, Ann Arbor, MI 48109, USA,
| | - Fei Fang
- Columbia University Irving Medical Center, Department of Orthopaedic Surgery, New York, NY 10032, USA
| | - Mikhail Golman
- Columbia University, Department of Biomedical Engineering, New York, NY 10027
| | | | - Stavros Thomopoulos
- Columbia University Irving Medical Center, Department of Orthopaedic Surgery, New York, NY 10032, USA,Columbia University, Department of Biomedical Engineering, New York, NY 10027
| |
Collapse
|
36
|
Hong JP, Huang SW, Lee CH, Chen HC, Charoenpong P, Lin HW. Osteoporosis increases the risk of rotator cuff tears: a population-based cohort study. J Bone Miner Metab 2022; 40:348-356. [PMID: 35059890 DOI: 10.1007/s00774-021-01293-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/14/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Osteoporosis has been demonstrated to be a risk factor for rotator cuff retears after surgery; however, no studies have directly investigated the association between osteoporosis and the development of rotator cuff tears. To investigate whether osteoporosis is associated with an increased risk of rotator cuff tears. MATERIALS AND METHODS We conducted a population-based, matched-cohort study with a 7-year follow-uTwo matched cohorts (n = 3511 with osteoporosis and 17,555 without osteoporosis) were recruited from Taiwan's Longitudinal Health Insurance Dataset. Person-year data and incidence rates were evaluated. A multivariable Cox model was used to derive an adjusted hazard ratio (aHR) after controlling for age, sex, and various prespecified comorbidities. Age and sex were added in the model to test for interaction with osteoporosis. RESULTS Women constituted 88.5% of the cohorts. During follow-up of 17,067 and 100,501 person-years for the osteoporosis and nonosteoporosis cohorts, 166 and 89 rotator cuff tears occurred, respectively. The cumulative incidence of rotator cuff tears was significantly higher in the osteoporosis cohort than in the nonosteoporosis cohort (p < 0.001, log-rank). The Cox model revealed a 1.79-fold increase in rotator cuff tears in the osteoporosis cohort, with an aHR of 1.79 (95% confidence interval, 1.55-2.05). Effect modification of sex and age on rotator cuff tears was not found in patients with osteoporosis. CONCLUSION This population-based study supports the hypothesis that compared with individuals without osteoporosis, those with osteoporosis have a higher risk of developing rotator cuff tears.
Collapse
Affiliation(s)
- Jia-Pei Hong
- Department of Physical Medicine and Rehabilitation, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shih-Wei Huang
- Department of Physical Medicine and Rehabilitation, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hong Lee
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and Department of Neurology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chou Chen
- Department of Physical Medicine and Rehabilitation, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Center for Evidence-Based Health Care, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Prangthip Charoenpong
- Department of Internal Medicine, Division of Pulmonary and Critical Care, Louisiana State University, Shreveport, LA, USA
| | - Hui-Wen Lin
- Department of Mathematics, Soochow University, 70 Linhsi Road, Shihlin, Taipei, 111, Taiwan.
| |
Collapse
|
37
|
Growth and mechanobiology of the tendon-bone enthesis. Semin Cell Dev Biol 2022; 123:64-73. [PMID: 34362655 PMCID: PMC8810906 DOI: 10.1016/j.semcdb.2021.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022]
Abstract
Tendons are cable-like connective tissues that transfer both active and passive forces generated by skeletal muscle to bone. In the mature skeleton, the tendon-bone enthesis is an interfacial zone of transitional tissue located between two mechanically dissimilar tissues: compliant, fibrous tendon to rigid, dense mineralized bone. In this review, we focus on emerging areas in enthesis development related to its structure, function, and mechanobiology, as well as highlight established and emerging signaling pathways and physiological processes that influence the formation and adaptation of this important transitional tissue.
Collapse
|
38
|
Driving native-like zonal enthesis formation in engineered ligaments using mechanical boundary conditions and β-tricalcium phosphate. Acta Biomater 2022; 140:700-716. [PMID: 34954418 DOI: 10.1016/j.actbio.2021.12.020] [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: 06/28/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 11/21/2022]
Abstract
Fibrocartilaginous entheses are structurally complex tissues that translate load from elastic ligaments to stiff bone via complex zonal gradients in the organization, mineralization, and cell phenotype. Currently, these complex gradients necessary for long-term mechanical function are not recreated in soft tissue-to-bone healing or engineered replacements, contributing to high failure rates. Previously, we developed a culture system that guides ligament fibroblasts to develop aligned native-sized collagen fibers using high-density collagen gels and mechanical boundary conditions. These constructs are promising ligament replacements, however functional ligament-to-bone attachments, or entheses, are required for long-term function in vivo. The objective of this study was to investigate the effect of compressive mechanical boundary conditions and the addition of beta-tricalcium phosphate (βTCP), a known osteoconductive agent, on the development of zonal ligament-to-bone entheses. We found that compressive boundary clamps, that restrict cellular contraction and produce a zonal tensile-compressive environment, guide ligament fibroblasts to produce 3 unique zones of collagen organization and zonal accumulation of glycosaminoglycans (GAGs), type II, and type X collagen. Ultimately, by 6 weeks of culture these constructs had similar organization and composition as immature bovine entheses. Further, βTCP applied under the clamp enhanced maturation of these entheses, leading to significantly increased tensile moduli, and zonal GAG accumulation, ALP activity, and calcium-phosphate accumulation, suggesting the initiation of endochondral ossification. This culture system produced some of the most organized entheses to date, closely mirroring early postnatal enthesis development, and provides an in vitro platform to better understand the cues that drive enthesis maturation in vivo. STATEMENT OF SIGNIFICANCE: Ligaments are attached to bone via entheses. Entheses are complex tissues with gradients in organization, composition, and cell phenotype. Entheses are necessary for proper transfer of load from ligament-to-bone, but currently are not restored with healing or replacements. Here, we provide new insight into how tensile-compressive boundary conditions and βTCP drive zonal gradients in collagen organization, mineralization, and matrix composition, producing tissues similar to immature ligament-to-bone attachments. Collectively, this culture system uses a bottom-up approach with mechanical and biochemical cues to produce engineered replacements which closely mirror postnatal enthesis development. This culture system is a promising platform to better understanding the cues that regulate enthesis formation so to better drive enthesis regeneration following graft repair and in engineered replacements.
Collapse
|
39
|
Navarro J, Korcari A, Nguyen P, Bah I, AlKhalifa A, Fink S, Buckley M, Kuo CK. Method development and characterization of chick embryo tendon mechanical properties. J Biomech 2022; 133:110970. [PMID: 35123205 PMCID: PMC11416868 DOI: 10.1016/j.jbiomech.2022.110970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/10/2022] [Accepted: 01/21/2022] [Indexed: 12/16/2022]
Abstract
Tendons are involved in multiple disorders and injuries, ranging from birth deformities to tendinopathies to acute ruptures. The ability to characterize embryonic tendon mechanical properties will enable elucidation of mechanisms responsible for functional tendon formation. In turn, an understanding of tendon development could inform approaches for adult and embryonic tendon tissue engineering and regenerative medicine. The chick embryo is a scientifically relevant model that we have been using to study Achilles (calcaneal) tendon development. Chick embryo calcaneal tendons are challenging to mechanically test due to small size and delicate nature, and difficulty distinguishing embryonic tendons from muscle and fibrocartilage using the naked eye. Here, we developed and implemented a "marking protocol" to identify and isolate calcaneal tendons at different stages of chick embryonic development. Mechanical testing of tendons isolated using the marking protocol revealed trends in mechanical property development that were not observed with tendons isolated by naked eye (eyeballing). Marked tendons exhibited non-linear increases in tensile modulus and ultimate tensile strength, whereas eyeballed tendons exhibited linear increases in the same properties, reflecting a need for the marking protocol. Furthermore, the tensile mechanical properties characterized for marked tendons are consistent with previously reported trends in cell length-scale mechanical properties measured using atomic force microscopy. This report establishes new methodology to enable tensile testing of chick embryo tendons and provides new information about embryonic tendon mechanical property development.
Collapse
Affiliation(s)
- Javier Navarro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States; Department of Biomedical Engineering, University of Rochester, NY, United States; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Antonion Korcari
- Department of Biomedical Engineering, University of Rochester, NY, United States; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Phong Nguyen
- Department of Biomedical Engineering, University of Rochester, NY, United States; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Ibrahima Bah
- Department of Biomedical Engineering, University of Rochester, NY, United States; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | | | - Spencer Fink
- Department of Biomedical Engineering, University of Rochester, NY, United States
| | - Mark Buckley
- Department of Biomedical Engineering, University of Rochester, NY, United States; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Catherine K Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States; Department of Biomedical Engineering, University of Rochester, NY, United States; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States; Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
40
|
Jiang X, Wojtkiewicz M, Patwardhan C, Greer S, Kong Y, Kuss M, Huang X, Liao J, Lu Y, Dudley A, Gundry RL, Fuchs M, Streubel P, Duan B. The effects of maturation and aging on the rotator cuff tendon-to-bone interface. FASEB J 2021; 35:e22066. [PMID: 34822203 DOI: 10.1096/fj.202101484r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/29/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
Rotator cuff tendon injuries often occur at the tendon-to-bone interface (i.e., enthesis) area, with a high prevalence for the elderly population, but the underlying reason for this phenomenon is still unknown. The objective of this study is to identify the histological, molecular, and biomechanical alterations of the rotator cuff enthesis with maturation and aging in a mouse model. Four different age groups of mice (newborn, young, adult, and old) were studied. Striking variations of the entheses were observed between the newborn and other matured groups, with collagen content, proteoglycan deposition, collagen fiber dispersion was significantly higher in the newborn group. The compositional and histological features of young, adult, and old groups did not show significant differences, except having increased proteoglycan deposition and thinner collagen fibers at the insertion sites in the old group. Nanoindentation testing showed that the old group had a smaller compressive modulus at the insertion site when compared with other groups. However, tensile mechanical testing reported that the old group demonstrated a significantly higher failure stress when compared with the young and adult groups. The proteomics analysis detected dramatic differences in protein content between newborn and young groups but minor changes among young, adult, and old groups. These results demonstrated: (1) the significant alterations of the enthesis composition and structure occur from the newborn to the young time period; (2) the increased risk of rotator cuff tendon injuries in the elderly population is not solely because of old age alone in the rodent model.
Collapse
Affiliation(s)
- Xiping Jiang
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Molecular Genetics and Cell Biology Program, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Melinda Wojtkiewicz
- CardiOmics Program, Center for Heart and Vascular Research, Division of Cardiovascular Medicine, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chinmay Patwardhan
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Sydney Greer
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Molecular Genetics and Cell Biology Program, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Xi Huang
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Yongfeng Lu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Andrew Dudley
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Molecular Genetics and Cell Biology Program, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Rebekah L Gundry
- CardiOmics Program, Center for Heart and Vascular Research, Division of Cardiovascular Medicine, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthias Fuchs
- Department of Physics and Astronomy, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Philipp Streubel
- Department of Orthopedic Surgery and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
41
|
Fosca M, Basoli V, Della Bella E, Russo F, Vadala G, Alini M, Rau JV, Verrier S. Raman spectroscopy in skeletal tissue disorders and tissue engineering: present and prospective. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:949-965. [PMID: 34579558 DOI: 10.1089/ten.teb.2021.0139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Musculoskeletal disorders are the most common reason of chronic pain and disability representing worldwide an enormous socio-economic burden. In this review, new biomedical application fields for Raman spectroscopy (RS) technique related to skeletal tissues are discussed showing that it can provide a comprehensive profile of tissue composition in situ, in a rapid, label-free, and non-destructive manner. RS can be used as a tool to study tissue alterations associated to aging, pathologies, and disease treatments. The main advantage with respect to currently applied methods in clinics is its ability to provide specific information on molecular composition, which goes beyond other diagnostic tools. Being compatible with water, RS can be performed without pre-treatment on unfixed, hydrated tissue samples, without any labelling and chemical fixation used in histochemical methods. This review provides first the description of basic principles of RS as a biotechnology tool and introduces into the field of currently available RS based techniques, developed to enhance Raman signal. The main spectral processing statistical tools, fingerprint identification and available databases are mentioned. The recent literature has been analysed for such applications of RS as tendon and ligaments, cartilage, bone, and tissue engineered constructs for regenerative medicine. Several cases of proof-of-concept preclinical studies have been described. Finally, advantages, limitations, future perspectives, and challenges for translation of RS into clinical practice have been also discussed.
Collapse
Affiliation(s)
- Marco Fosca
- Istituto di Struttura della Materia Consiglio Nazionale delle Ricerche, 204549, Roma, Lazio, Italy;
| | - Valentina Basoli
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| | - Elena Della Bella
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| | - Fabrizio Russo
- Campus Bio-Medico University Hospital, 220431, Roma, Lazio, Italy;
| | - Gianluca Vadala
- Campus Bio-Medico University Hospital, 220431, Roma, Lazio, Italy;
| | - Mauro Alini
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| | - Julietta V Rau
- Istituto di Struttura della Materia Consiglio Nazionale delle Ricerche, 204549, Roma, Lazio, Italy.,I M Sechenov First Moscow State Medical University, 68477, Moskva, Moskva, Russian Federation;
| | - Sophie Verrier
- AO Research Institute Davos, 161930, Regenerative Orthopaedics, Davos, Graubünden, Switzerland;
| |
Collapse
|
42
|
Zhang X, Wang D, Mak KLK, Tuan RS, Ker DFE. Engineering Musculoskeletal Grafts for Multi-Tissue Unit Repair: Lessons From Developmental Biology and Wound Healing. Front Physiol 2021; 12:691954. [PMID: 34504435 PMCID: PMC8421786 DOI: 10.3389/fphys.2021.691954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022] Open
Abstract
In the musculoskeletal system, bone, tendon, and skeletal muscle integrate and act coordinately as a single multi-tissue unit to facilitate body movement. The development, integration, and maturation of these essential components and their response to injury are vital for conferring efficient locomotion. The highly integrated nature of these components is evident under disease conditions, where rotator cuff tears at the bone-tendon interface have been reported to be associated with distal pathological alterations such as skeletal muscle degeneration and bone loss. To successfully treat musculoskeletal injuries and diseases, it is important to gain deep understanding of the development, integration and maturation of these musculoskeletal tissues along with their interfaces as well as the impact of inflammation on musculoskeletal healing and graft integration. This review highlights the current knowledge of developmental biology and wound healing in the bone-tendon-muscle multi-tissue unit and perspectives of what can be learnt from these biological and pathological processes within the context of musculoskeletal tissue engineering and regenerative medicine. Integrating these knowledge and perspectives can serve as guiding principles to inform the development and engineering of musculoskeletal grafts and other tissue engineering strategies to address challenging musculoskeletal injuries and diseases.
Collapse
Affiliation(s)
- Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - King-Lun Kingston Mak
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
43
|
Chen H, Li S, Xiao H, Wu B, Zhou L, Hu J, Lu H. Effect of Exercise Intensity on the Healing of the Bone-Tendon Interface: A Mouse Rotator Cuff Injury Model Study. Am J Sports Med 2021; 49:2064-2073. [PMID: 33989078 DOI: 10.1177/03635465211011751] [Citation(s) in RCA: 12] [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] [Indexed: 01/31/2023]
Abstract
BACKGROUND Injuries at the bone-tendon interface (BTI) are common findings in clinical practice. Rehabilitation procedures after BTI surgery are important but are controversial. PURPOSE To investigate the effects of different exercise intensities on BTI healing by means of an established mouse rotator cuff injury model. STUDY DESIGN Controlled laboratory study. METHODS A total of 150 specific pathogen free male C57BL/6 mice, with supraspinatus insertion injury, were randomly assigned to 1 of 5 groups according to postoperative rehabilitation of different exercise intensities: (1) control group, (2) low-intensity exercise group, (3) moderate-intensity exercise group, (4) high-intensity exercise group, and (5) increasing-intensity exercise group (IG). The specimens were harvested 4 or 8 weeks postoperatively for microarchitectural, histological, molecular biological, and mechanical evaluations. RESULTS Histological test results showed that the degrees of tissue fusion and polysaccharide protein distribution at the healing interface at 4 and 8 weeks after surgery were significantly better in the IG than in the other 4 groups. Synchrotron radiation micro-computed tomography showed that the quantity of subchondral bone at the enthesis (bone volume/total volume fraction, trabecular thickness, trabecular number) was higher and trabecular separation was lower in the IG than in the other 4 groups. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis revealed that the healing interface in the IG expressed more transcription factors, such as sox 9, runx 2, and scleraxis, than the interfaces in the other groups. Although no significant difference was seen in the cross-sectional area between the groups at postoperative weeks 4 and 8 (P > .05), the tensile load, ultimate strength, and stiffness of the specimens in the IG were significantly better than those in the other 4 groups (P < .05). CONCLUSION The rehabilitation program with increasing-intensity exercise was beneficial for BTI healing. CLINICAL RELEVANCE The results of this study provide evidence supporting the use of a simple and progressive exercise rehabilitation program after rotator cuff surgery.
Collapse
Affiliation(s)
- Huabin Chen
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
| | - Shengcan Li
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
| | - Han Xiao
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
| | - Bing Wu
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
| | - Li Zhou
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- Department of Orthopedic Center, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Jianzhong Hu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Lu
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
| |
Collapse
|
44
|
Theodossiou SK, Pancheri NM, Martes AC, Bozeman AL, Brumley MR, Raveling AR, Courtright JM, Schiele NR. Neonatal Spinal Cord Transection Decreases Hindlimb Weight-Bearing and Affects Formation of Achilles and Tail Tendons. J Biomech Eng 2021; 143:061012. [PMID: 33537729 PMCID: PMC8114905 DOI: 10.1115/1.4050031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/06/2021] [Indexed: 01/08/2023]
Abstract
Mechanical loading may be required for proper tendon formation. However, it is not well understood how tendon formation is impacted by the development of weight-bearing locomotor activity in the neonate. This study assessed tendon mechanical properties, and concomitant changes in weight-bearing locomotion, in neonatal rats subjected to a low thoracic spinal cord transection or a sham surgery at postnatal day (P)1. On P10, spontaneous locomotion was evaluated in spinal cord transected and sham controls to determine impacts on weight-bearing hindlimb movement. The mechanical properties of P10 Achilles tendons (ATs), as representative energy-storing, weight-bearing tendons, and tail tendons (TTs), as representative positional, non-weight-bearing tendons were evaluated. Non- and partial weight-bearing hindlimb activity decreased in spinal cord transected rats compared to sham controls. No spinal cord transected rats showed full weight-bearing locomotion. ATs from spinal cord transected rats had increased elastic modulus, while cross-sectional area trended lower compared to sham rats. TTs from spinal cord transected rats had higher stiffness and cross-sectional area. Collagen structure of ATs and TTs did not appear impacted by surgery condition, and no significant differences were detected in the collagen crimp pattern. Our findings suggest that mechanical loading from weight-bearing locomotor activity during development regulates neonatal AT lateral expansion and maintains tendon compliance, and that TTs may be differentially regulated. The onset and gradual increase of weight-bearing movement in the neonate may provide the mechanical loading needed to direct functional postnatal tendon formation.
Collapse
Affiliation(s)
- Sophia K. Theodossiou
- Biological Engineering, University of Idaho, 875 Perimeter Drive, MS 0904, Moscow, ID 83844
| | - Nicholas M. Pancheri
- Biological Engineering, University of Idaho, 875 Perimeter Drive, MS 0904, Moscow, ID 83844
| | - Alleyna C. Martes
- Psychology, Idaho State University, 921 South 8th Avenue Stop 8112, Pocatello, ID 83209
| | - Aimee L. Bozeman
- Psychology, Idaho State University, 921 South 8th Avenue Stop 8112, Pocatello, ID 83209
| | - Michele R. Brumley
- Psychology, Idaho State University, 921 South 8th Avenue Stop 8087, Pocatello, ID 83209
| | - Abigail R. Raveling
- Biological Engineering, University of Idaho, 875 Perimeter Drive, MS 0904, Moscow, ID 83844
| | - Jeffrey M. Courtright
- Biological Engineering, University of Idaho, 875 Perimeter Drive, MS 0904, Moscow, ID 83844
| | - Nathan R. Schiele
- Biological Engineering, University of Idaho, 875 Perimeter Drive, MS 0904, Moscow, ID 83844
| |
Collapse
|
45
|
Roffino S, Camy C, Foucault-Bertaud A, Lamy E, Pithioux M, Chopard A. Negative impact of disuse and unloading on tendon enthesis structure and function. LIFE SCIENCES IN SPACE RESEARCH 2021; 29:46-52. [PMID: 33888287 DOI: 10.1016/j.lssr.2021.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/19/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
Exposure to chronic skeletal muscle disuse and unloading that astronauts experience results in muscle deconditioning and bone remodeling. Tendons involved in the transmission of force from muscles to skeleton are also affected. Understanding the changes that occur in muscle, tendon, and bone is an essential step toward limiting or preventing the deleterious effects of chronic reduction in mechanical load. Numerous reviews have reported the effects of this reduction on both muscle and bone, and to a lesser extent on the tendon. However, none focused on the tendon enthesis, the tendon-to-bone attachment site. While the enthesis structure appears to be determined by mechanical stress, little is known about enthesis plasticity. Our review first looks at the relationship between entheses and mechanical stress, exploring how tensile and compressive loads determine and influence enthesis structure and composition. The second part of this review addresses the deleterious effects of skeletal muscle disuse and unloading on enthesis structure, composition, and function. We discuss the possibility that spaceflight-induced enthesis remodeling could impact both the capacity of the enthesis to withstand compressive stress and its potential weakness. Finally, we point out how altered compressive strength at entheses could expose astronauts to the risk of developing enthesopathies.
Collapse
Affiliation(s)
- S Roffino
- ISM Inst Movement Sci, Aix-Marseille University, CNRS, Marseille, France.
| | - C Camy
- ISM Inst Movement Sci, Aix-Marseille University, CNRS, Marseille, France
| | - A Foucault-Bertaud
- INSERM 1263, INRA 1260, C2VN, Aix-Marseille University, Marseille, France
| | - E Lamy
- ISM Inst Movement Sci, Aix-Marseille University, CNRS, Marseille, France
| | - M Pithioux
- ISM Inst Movement Sci, Aix-Marseille University, CNRS, Marseille, France
| | - A Chopard
- DMEM, Montpellier University, INRAE, Montpellier, France
| |
Collapse
|
46
|
Mechano-chemical enforcement of tendon apical ECM into nano-filaments during Drosophila flight muscle development. Curr Biol 2021; 31:1366-1378.e7. [PMID: 33545042 DOI: 10.1016/j.cub.2021.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/16/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023]
Abstract
Contractile tension is critical for musculoskeletal system development and maintenance. In insects, the muscular force is transmitted to the exoskeleton through the tendon cells and tendon apical extracellular matrix (ECM). In Drosophila, we found tendon cells secrete Dumpy (Dpy), a zona pellucida domain (ZPD) protein, to form the force-resistant filaments in the exuvial space, anchoring the tendon cells to the pupal cuticle. We showed that Dpy undergoes filamentous conversion in response to the tension increment during indirect flight muscle development. We also found another ZPD protein Quasimodo (Qsm) protects the notum epidermis from collapsing under the muscle tension by enhancing the tensile strength of Dpy filaments. Qsm is co-transported with Dpy in the intracellular vesicles and diffuses into the exuvial space after secretion. Tissue-specific qsm expression rescued the qsm mutant phenotypes in distant tissues, suggesting Qsm can function in a long-range, non-cell-autonomous manner. In the cell culture assay, Qsm interacts with Dpy-ZPD and promotes secretion and polymerization of Dpy-ZPD. The roles of Qsm underlies the positive feedback mechanism of force-dependent organization of Dpy filaments, providing new insights into apical ECM remodeling through the unconventional interaction of ZPD proteins.
Collapse
|
47
|
Abstract
Nature faces the challenge of stably attaching soft muscles to a stiff skeleton. A new study combines live imaging and fly genetics to reveal that mechanical tension and a putative intracellular chaperone assist in assembling the gigantic extracellular matrix protein Dumpy at fly tendon-skeleton interfaces.
Collapse
Affiliation(s)
- Clara Sidor
- Turing Center for Living Systems, Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Frank Schnorrer
- Turing Center for Living Systems, Aix Marseille University, CNRS, IBDM, 13288 Marseille, France.
| |
Collapse
|
48
|
Ozone K, Oka Y, Minegishi Y, Kano T, Kokubun T, Murata K, Kanemura N. Effect of Various Types of Muscle Contraction with Different Running Conditions on Mouse Humerus Morphology. Life (Basel) 2021; 11:life11040284. [PMID: 33801768 PMCID: PMC8065967 DOI: 10.3390/life11040284] [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: 02/20/2021] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 12/01/2022] Open
Abstract
How various types of muscle contraction during exercises affect bone formation remains unclear. This study aimed to determine how exercises with different muscle contraction types affect bone morphology. In total, 20 mice were used and divided into four groups: Control, Level, Down Slow, and Down. Different types of muscle contraction were induced by changing the running angle of the treadmill. After the intervention, micro-computed tomography (Micro-CT), tartrate-resistant acid phosphatase/alkaline phosphatase (ALP) staining, and immunohistochemical staining were used to analyze the humerus head, tendon-to-bone attachment, and humerus diaphyseal region. Micro-CT found that the volume ratio of the humeral head, the volume of the tendon-to-bone attachment region, and the area of the humeral diaphyseal region increased in the Down group. However, no difference was detected in bone morphology between the Level and Down Slow groups. In addition, histology showed activation of ALP in the subarticular subchondral region in the Down Slow and Down groups and the fibrocartilage region in the tendon-to-bone attachment. Moreover, Osterix increased predominantly in the Down Slow and Down groups.Overall bone morphological changes in the humerus occur only when overuse is added to EC-dominant activity. Furthermore, different type of muscle contractile activities might promote bone formation in a site-specific manner.
Collapse
Affiliation(s)
- Kaichi Ozone
- Graduate Course of Health and Social Services, Graduate School of Saitama Prefectural University, Saitama 343-8540, Japan; (K.O.); (Y.O.); (Y.M.); (T.K.)
- Research Fellowship for Young Scientists, Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - Yuichiro Oka
- Graduate Course of Health and Social Services, Graduate School of Saitama Prefectural University, Saitama 343-8540, Japan; (K.O.); (Y.O.); (Y.M.); (T.K.)
| | - Yuki Minegishi
- Graduate Course of Health and Social Services, Graduate School of Saitama Prefectural University, Saitama 343-8540, Japan; (K.O.); (Y.O.); (Y.M.); (T.K.)
- Research Fellowship for Young Scientists, Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - Takuma Kano
- Graduate Course of Health and Social Services, Graduate School of Saitama Prefectural University, Saitama 343-8540, Japan; (K.O.); (Y.O.); (Y.M.); (T.K.)
| | - Takanori Kokubun
- Department of Health and Social Services, Saitama Prefectural University, Saitama 343-8540, Japan; (T.K.); (K.M.)
| | - Kenji Murata
- Department of Health and Social Services, Saitama Prefectural University, Saitama 343-8540, Japan; (T.K.); (K.M.)
| | - Naohiko Kanemura
- Department of Health and Social Services, Saitama Prefectural University, Saitama 343-8540, Japan; (T.K.); (K.M.)
- Correspondence: ; Tel.: +81-48-971-0500
| |
Collapse
|
49
|
Puetzer JL, Ma T, Sallent I, Gelmi A, Stevens MM. Driving Hierarchical Collagen Fiber Formation for Functional Tendon, Ligament, and Meniscus Replacement. Biomaterials 2021; 269:120527. [PMID: 33246739 PMCID: PMC7883218 DOI: 10.1016/j.biomaterials.2020.120527] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/09/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022]
Abstract
Hierarchical collagen fibers are the primary source of strength in musculoskeletal tendons, ligaments, and menisci. It has remained a challenge to develop these large fibers in engineered replacements or in vivo after injury. The objective of this study was to investigate the ability of restrained cell-seeded high density collagen gels to drive hierarchical fiber formation for multiple musculoskeletal tissues. We found boundary conditions applied to high density collagen gels were capable of driving tenocytes, ligament fibroblasts, and meniscal fibrochondrocytes to develop native-sized hierarchical collagen fibers 20-40 μm in diameter. The fibers organize similar to bovine juvenile collagen with native fibril banding patterns and hierarchical fiber bundles 50-350 μm in diameter by 6 weeks. Mirroring fiber organization, tensile properties of restrained samples improved significantly with time, reaching ~1 MPa. Additionally, tendon, ligament, and meniscal cells produced significantly different sized fibers, different degrees of crimp, and different GAG concentrations, which corresponded with respective juvenile tissue. To our knowledge, these are some of the largest, most organized fibers produced to date in vitro. Further, cells produced tissue specific hierarchical fibers, suggesting this system is a promising tool to better understand cellular regulation of fiber formation to better stimulate it in vivo after injury.
Collapse
Affiliation(s)
- Jennifer L Puetzer
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ; Department of Biomedical Engineering and Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States, 23284.
| | - Tianchi Ma
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Ignacio Sallent
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Amy Gelmi
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ.
| |
Collapse
|
50
|
Ganji E, Chan CS, Ward CW, Killian ML. Optogenetic activation of muscle contraction in vivo. Connect Tissue Res 2021; 62:15-23. [PMID: 32777957 PMCID: PMC7718400 DOI: 10.1080/03008207.2020.1798943] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/15/2020] [Indexed: 02/03/2023]
Abstract
Purpose: Optogenetics is an emerging alternative to traditional electrical stimulation to initiate action potentials in activatable cells both ex vivo and in vivo. Optogenetics has been commonly used in mammalian neurons and more recently, it has been adapted for activation of cardiomyocytes and skeletal muscle. Therefore, the aim of this study was to evaluate the stimulation feasibility and sustain isometric muscle contraction and limit decay for an extended period of time (1s), using non-invasive transdermal light activation of skeletal muscle (triceps surae) in vivo. MATERIALS AND METHODS We used inducible Cre recombination to target expression of Channelrhodopsin-2 (ChR2(H134R)-EYFP) in skeletal muscle (Acta1-Cre) in mice. Fluorescent imaging confirmed that ChR2 expression is localized in skeletal muscle and does not have specific expression in sciatic nerve branch, therefore, allowing for non-nerve mediated optical stimulation of skeletal muscle. We induced muscle contraction using transdermal exposure to blue light and selected 10 Hz stimulation after controlled optimization experiments to sustain prolonged muscle contraction. RESULTS Increasing the stimulation frequency from 10 Hz to 40 Hz increased the muscle contraction decay during prolonged 1s stimulation, highlighting frequency dependency and importance of membrane repolarization for effective light activation. Finally, we showed that optimized pulsed optogenetic stimulation of 10 Hz resulted in comparable ankle torque and contractile functionality to that of electrical stimulation. CONCLUSIONS Our results demonstrate the feasibility and repeatability of non-invasive optogenetic stimulation of muscle in vivo and highlight optogenetic stimulation as a powerful tool for non-invasive in vivo direct activation of skeletal muscle.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
- Department of Orthopaedic Surgery, Michigan Medicine, Ann Arbor, Michigan, 48109
| | - C. Savio Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christopher W. Ward
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Megan L. Killian
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
- Department of Orthopaedic Surgery, Michigan Medicine, Ann Arbor, Michigan, 48109
| |
Collapse
|