1
|
Long Q, Liu C, Zheng H, Wang M, Liu H, Liu Y, Cao Z, Sun Y, Mo Q, Backman LJ, Zhu J, Hu L, Huang J, Zhang W, Chen J. Enhancing Tendon Regeneration: Investigating the Impact of Topography on the Secretome of Adipose-Derived Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417447. [PMID: 40091553 PMCID: PMC12079404 DOI: 10.1002/advs.202417447] [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] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Indexed: 03/19/2025]
Abstract
Tendons are vital for maintaining integrity and movement, but current treatment options are insufficient for their regeneration after injuries. Previous studies have shown that the secretome from mesenchymal stem cells (MSCs) promoted tendon regeneration. However, limited studies have explored the impact of the physical microenvironment on the secretome's efficacy of MSCs. In this study, it is shown that the topographic orientation regulates the secretome of human adipose-derived stem cells (ADSCs) and promotes tendon regeneration. Conditioned medium (CM) is collected from ADSCs cultured on the scaffolds with different topography. The results show that CM generated from aligned structure group has a potent effect in promoting cell migration and proliferation, tenogenic differentiation, macrophage polarization toward M2 phenotype, tendon structure and mechanical function recovery. Proteomic analysis revealed that the aligned structure can up-regulate the secretion of Extracellular matrix (ECM) proteins while down-regulate proinflammatory factors. This modulation activates the MAPK, GPCR and Integrin signaling pathways which may account for the enhanced effect on tendon regeneration. This study offers a promising and safer non-cell-based treatment option for tendon repair.
Collapse
Affiliation(s)
- Qiuzi Long
- Nanjing University of Chinese MedicineNanjing210029China
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- Nanjing Second HospitalNanjing Hospital affiliated to Nanjing University of Chinese MedicineNanjing210003China
| | - Chuanquan Liu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Haotian Zheng
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Mingyue Wang
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Hanmei Liu
- Nanjing University of Chinese MedicineNanjing210029China
| | - Yue Liu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Zhicheng Cao
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Department of Orthopaedic SurgeryInstitute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yuzhi Sun
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Department of Orthopaedic SurgeryInstitute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Qingyun Mo
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Ludvig J. Backman
- Department of Medical and Translational Biology, AnatomyUmeå UniversityUmeå90187Sweden
- Department of Community Medicine and RehabilitationUmeå UniversityUmeå90187Sweden
| | - Jialin Zhu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Lizhi Hu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Jinlong Huang
- Nanjing University of Chinese MedicineNanjing210029China
| | - Wei Zhang
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Jiangsu Key Laboratory for Biomaterials and DevicesSoutheast UniversityNanjing210096China
- China Orthopedic Regenerative Medicine Group (CORMed)Hangzhou310058China
| | - Jialin Chen
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Jiangsu Key Laboratory for Biomaterials and DevicesSoutheast UniversityNanjing210096China
- Department of OphthalmologyZhongda HospitalSoutheast UniversityNanjing210009China
| |
Collapse
|
2
|
Hold LA, Migotsky N, Lamia SN, Steltzer SS, Grossman S, Chen J, Bae SH, Cordts P, Phillips T, O'Meara MJ, Davis C, Brooks SV, Akbar M, Millar NL, Killian ML, Abraham AC. AMP-activated protein kinase (AMPK) is essential for tendon homeostasis and prevents premature senescence and ectopic calcification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635920. [PMID: 39975248 PMCID: PMC11838443 DOI: 10.1101/2025.01.31.635920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Tendinopathy is a debilitating tendon disorder affecting millions of people, characterized by pain, swelling, and diminished biomechanical properties. While the precise mechanisms underlying tendon homeostasis remain unclear, metabolic regulation plays a critical role. In this study, we combine transcriptomic analysis of human tendinopathic samples with a conditional mouse model in which Prkaa1 (encoding AMPKα1) is selectively deleted in tendon progenitors to elucidate the role of AMPK signaling in tendon homeostasis. RNA sequencing of diseased human tendons revealed downregulation of key metabolic genes, including several involved in the mitochondrial electron transport chain and AMPK signaling pathways, alongside an increase in markers associated with senescence and a secretory inflammatory profile. In parallel, mice with loss of Prkaa1 function exhibited normal postnatal development; however, by one month of age, tendons demonstrated widespread transcriptional alterations related to cell cycle regulation and ECM organization. By three months, AMPKα1-deficient tendons showed significant reductions in mechanical strength and increased expression of senescence markers p21 and p16, progressing to prominent ectopic calcification with age. In vitro studies further confirmed that tendon fibroblasts lacking AMPKα1 have altered ECM substrate adhesion profiles. Importantly, voluntary exercise partially rescued these deficits by enhancing ECM organization and reducing senescence marker expression. Collectively, our findings demonstrate that AMPKα1 is critical for maintaining energy balance, regulating ECM remodeling, and preventing premature cellular senescence in tendon. These insights highlight AMPK signaling as a promising therapeutic target and underscore the beneficial role of exercise in mitigating tendinopathic changes.
Collapse
|
3
|
Gao Y, Wang H, Shi L, Lu P, Dai G, Zhang M, Han B, Cao M, Li Y, Rui Y. Erroneous Differentiation of Tendon Stem/Progenitor Cells in the Pathogenesis of Tendinopathy: Current Evidence and Future Perspectives. Stem Cell Rev Rep 2025; 21:423-453. [PMID: 39579294 DOI: 10.1007/s12015-024-10826-z] [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] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Tendinopathy is a condition characterized by persistent tendon pain, structural damage, and compromised functionality. Presently, the treatment for tendinopathy remains a formidable challenge, partly because of its unclear pathogenesis. Tendon stem/progenitor cells (TSPCs) are essential for tendon homeostasis, regeneration, remodeling, and repair. An innovative theory has been previously proposed, with insufficient evidence, that the erroneous differentiation of TSPCs may constitute one of the fundamental mechanisms underpinning tendinopathy. Over the past few years, there has been accumulating evidence for plausibility of this theory. In this review, we delve into alterations in the differentiation potential of TSPCs and the underlying mechanisms in the context of injury-induced tendinopathy, diabetic tendinopathy, and age-related tendinopathy to provide updated evidence on the erroneous differentiation theory. Despite certain limitations inherent in the existing body of evidence, the erroneous differentiation theory emerges as a promising and highly pertinent avenue for understanding tendinopathy. In the future, advanced methodologies will be harnessed to further deepen comprehension of this theory, paving the way for prospective developments in clinical therapies targeting TSPCs for the management of tendinopathy.
Collapse
Affiliation(s)
- Yucheng Gao
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Hao Wang
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Liu Shi
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Panpan Lu
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Guangchun Dai
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Ming Zhang
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Bowen Han
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Mumin Cao
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yingjuan Li
- Department of Geriatrics, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yunfeng Rui
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, China.
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
4
|
Ciardulli MC, Lovecchio J, Parolini O, Giordano E, Maffulli N, Della Porta G. Fibrin Scaffolds Perfused with Transforming Growth Factor-β1 as an In Vitro Model to Study Healthy and Tendinopathic Human Tendon Stem/Progenitor Cells. Int J Mol Sci 2024; 25:9563. [PMID: 39273510 PMCID: PMC11395617 DOI: 10.3390/ijms25179563] [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: 07/17/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
A limited understanding of tendon cell biology in healthy and pathological conditions has impeded the development of effective treatments, necessitating in vitro biomimetic models for studying tendon events. We established a dynamic culture using fibrin scaffolds, bioengineered with tendon stem/progenitor cells (hTSPCs) from healthy or diseased human biopsies and perfused with 20 ng/mL of human transforming growth factor-β1 for 21 days. Both cell types showed long-term viability and upregulated Scleraxis (SCX-A) and Tenomodulin (TNMD) gene expressions, indicating tenogenic activity. However, diseased hTSPCs underexpressed collagen type I and III (COL1A1 and COL3A1) genes and exhibited lower SCX-A and TNMD protein levels, but increased type I collagen production, with a type I/type III collagen ratio > 1.5 by day 14, matching healthy cells. Diseased hTSPCs also showed constant high levels of pro-inflammatory cytokines, such as IL-8 and IL-6. This biomimetic environment is a valuable tool for studying tenogenic and inflammatory events in healthy and diseased tendon cells and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Maria Camilla Ciardulli
- Translational Nanomedicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende 43, 84081 Baronissi, Italy
| | - Joseph Lovecchio
- School of Science and Engineering, Reykjavík University, 102 Reykjavík, Iceland
- Institute of Biomedical and Neural Engineering, Reykjavik University, 102 Reykjavík, Iceland
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00136 Rome, Italy
| | - Emanuele Giordano
- Laboratory of Cellular and Molecular Engineering "Silvio Cavalcanti", Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, 47522 Cesena, Italy
- Advanced Research Center on Electronic Systems (ARCES), University of Bologna, 40126 Bologna, Italy
| | - Nicola Maffulli
- Department of Trauma and Orthopaedics, Faculty of Medicine and Psychology, Sant' Andrea Hospital, Sapienza University, 00189 Rome, Italy
| | - Giovanna Della Porta
- Translational Nanomedicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende 43, 84081 Baronissi, Italy
- Interdepartment Centre BIONAM, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| |
Collapse
|
5
|
He W, Jiang C, Zhou P, Hu X, Gu X, Zhang S. Role of tendon-derived stem cells in tendon and ligament repair: focus on tissue engineer. Front Bioeng Biotechnol 2024; 12:1357696. [PMID: 39175617 PMCID: PMC11338810 DOI: 10.3389/fbioe.2024.1357696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
This review offered a comprehensive analysis of tendon and ligament injuries, emphasizing the crucial role of tendon-derived stem cells (TDSCs) in tissue engineering as a potential solution for these challenging medical conditions. Tendon and ligament injuries, prevalent among athletes, the elderly, and laborers, often result in long-term disability and reduced quality of life due to the poor intrinsic healing capacity of these avascular structures. The formation of biomechanically inferior scar tissue and a high rate of reinjury underscore the need for innovative approaches to enhance and guide the regenerative process. This review delved into the complexities of tendon and ligament structure and function, types of injuries and their impacts, and the limitations of the natural repair process. It particularly focused on the role of TDSCs within the context of tissue engineering. TDSCs, with their ability to differentiate into tenocytes, are explored in various applications, including biocompatible scaffolds for cell tracking, co-culture systems to optimize tendon-bone healing, and graft healing techniques. The review also addressed the challenges of immunoreactivity post-transplantation, the importance of pre-treating TDSCs, and the potential of hydrogels and decellularized matrices in supporting tendon regeneration. It concluded by highlighting the essential roles of mechanical and molecular stimuli in TDSC differentiation and the current challenges in the field, paving the way for future research directions.
Collapse
Affiliation(s)
- Wei He
- Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Chao Jiang
- Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Ping Zhou
- Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Xujun Hu
- Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - XiaoPeng Gu
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Orthopedics, Zhoushan Guhechuan Hospital, Zhoushan, Zhejiang, China
| | - SongOu Zhang
- Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Orthopedics, Zhoushan Guhechuan Hospital, Zhoushan, Zhejiang, China
| |
Collapse
|
6
|
Chen R, Chen F, Chen K, Xu J. Advances in the application of hydrogel-based scaffolds for tendon repair. Genes Dis 2024; 11:101019. [PMID: 38560496 PMCID: PMC10978548 DOI: 10.1016/j.gendis.2023.04.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/23/2023] [Accepted: 04/30/2023] [Indexed: 04/04/2024] Open
Abstract
Tendon injuries often lead to joint dysfunction due to the limited self-regeneration capacity of tendons. Repairing tendons is a major challenge for surgeons and imposes a significant financial burden on society. Therefore, there is an urgent need to develop effective strategies for repairing injured tendons. Tendon tissue engineering using hydrogels has emerged as a promising approach that has attracted considerable interest. Hydrogels possess excellent biocompatibility and biodegradability, enabling them to create an extracellular matrix-like growth environment for cells. They can also serve as a carrier for cells or other substances to accelerate tendon repair. In the past decade, numerous studies have made significant progress in the preparation of hydrogel scaffolds for tendon healing. This review aims to provide an overview of recent research on the materials of hydrogel-based scaffolds used for tendon tissue engineering and discusses the delivery systems based on them.
Collapse
Affiliation(s)
- Renqiang Chen
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, China
| | - Fanglin Chen
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, China
| | - Kenian Chen
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, China
| | - Jian Xu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
7
|
Augustin G, Jeong JH, Kim M, Hur SS, Lee JH, Hwang Y. Stem Cell‐Based Therapies and Tissue Engineering Innovations for Tendinopathy: A Comprehensive Review of Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202300425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 01/06/2025]
Abstract
AbstractTendon diseases commonly lead to physical disability, exerting a profound impact on the routine of affected patients. These conditions respond poorly to existing treatments, presenting a substantial challenge for orthopedic scientists. Research into clinical translational therapy has yet to yield highly versatile interventions capable of effectively addressing tendon diseases, including tendinopathy. Stem cell‐based therapies have emerged as a promising avenue for modifying the biological milieu through the secretion of regenerative and immunomodulatory factors. The current review provides an overview of the intricate tendon microenvironment, encompassing various tendon stem progenitor cells within distinct tendon sublocations, gene regulation, and pathways pertinent to tendon development, and the pathology of tendon diseases. Subsequently, the advantages of stem cell‐based therapies are discussed that utilize distinct types of autologous and allogeneic stem cells for tendon regeneration at the translational level. Moreover, this review outlines the challenges, gaps, and future innovations to propose a consolidated stem cell‐based therapy to treat tendinopathy. Finally, regenerative soluble therapies, insoluble bio‐active therapies, along with insoluble bio‐active therapies, and implantable 3D scaffolds for tendon tissue engineering are discussed, thereby presenting a pathway toward enhanced tissue regeneration and engineering.
Collapse
Affiliation(s)
- George Augustin
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Biochemistry and Biophysics Oregon State University Corvallis OR 92331 USA
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Integrated Biomedical Science Soonchunhyang University Asan‐si, Chungnam‐Do 31538 Republic of Korea
| | - Min‐Kyu Kim
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
| | - Sung Sik Hur
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
| | - Joon Ho Lee
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Integrated Biomedical Science Soonchunhyang University Asan‐si, Chungnam‐Do 31538 Republic of Korea
| |
Collapse
|
8
|
Squier K, Mousavizadeh R, Damji F, Beck C, Hunt M, Scott A. In vitro collagen biomarkers in mechanically stimulated human tendon cells: a systematic review. Connect Tissue Res 2024; 65:89-101. [PMID: 38375562 DOI: 10.1080/03008207.2024.2313582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/25/2024] [Indexed: 02/21/2024]
Abstract
OBJECTIVE The aim of this study was to comprehensively examine and summarize the available in vitro evidence regarding the relationship between mechanical stimulation and biomarkers of collagen synthesis in human-derived tendon cells. METHODS Systematic review with narrative analyses and risk of bias assessment guided by the Health Assessment and Translation tool. The electronic databases MEDLINE (Ovid), EMBASE (Ovid), CENTRAL (Ovid) and COMPENDEX (Engineering Village) were systematically searched from inception to 3 August 2023. Inclusion criteria encompassed English language, original experimental, or quasi-experimental in vitro publications that subjected human tendon cells to mechanical stimulation, with collagen synthesis (total collagen, type I, III, V, XI, XII, and XIV) and related biomarkers (matrix metalloproteinases, transforming growth factor β, scleraxis, basic fibroblast growth factor) as outcomes. RESULTS Twenty-one publications were included. A pervasive definite high risk of bias was evident in all included studies. Owing to incomplete outcome reporting and heterogeneity in mechanical stimulation protocols, planned meta-analyses were unfeasible. Reviewed data suggested that human tendon cells respond to mechanical stimulation with increased synthesis of collagen (e.g., COL1A1, procollagen, total soluble collagen, etc.), scleraxis and several matrix metalloproteinases. Results also indicate that mechanical stimulation dose magnitude may influence synthesis in several biomarkers. CONCLUSIONS A limited number of studies, unfortunately characterized by a definite high risk of bias, suggest that in vitro mechanical stimulation primarily increases type I collagen synthesis by human tendon cells. Findings from this systematic review provide researchers and clinicians with biological evidence concerning the possible beneficial influence of exercise and loading on cellular-level tendon adaptation.
Collapse
Affiliation(s)
- Kipling Squier
- Department of Physical Therapy, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at VCH, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Rouhollah Mousavizadeh
- Centre for Aging SMART at VCH, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Faraz Damji
- Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Charlotte Beck
- Woodward Library, University of British Columbia, Vancouver, Canada
| | - Michael Hunt
- Department of Physical Therapy, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at VCH, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Alexander Scott
- Department of Physical Therapy, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at VCH, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
9
|
Li Z, Sun X, Shen C, Deng Z, Tang K, Xie Y, Chen L, Nie M. Dynamic Tensile Stress Promotes Regeneration of Achilles Tendon in a Panda Rope Bridge Technique Mice Model. Ann Biomed Eng 2023; 51:2735-2748. [PMID: 37482574 DOI: 10.1007/s10439-023-03320-z] [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: 04/01/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
Regeneration of ruptured Achilles tendon remains a clinical challenge owing to its limited regenerative capacity. Dynamic tensile stress plays a positive role in the regeneration of tendon, although the specific underlying mechanisms remain unclear. In this study, the Achilles tendon defect-regeneration model was created in male C57BL/6 mice aged 8 weeks. The animals were randomly assigned to four groups-repair, non-repair, repair with fixation, and non-repair with fixation. The repair group and repair with fixation group adopted the panda rope bridge technique (PRBT) repair method. Our results demonstrated the presence of more densely aligned and mature collagen fibers, as well as more tendon-related makers, in the repair group at both 2- and 4-week post-surgery. Furthermore, the biomechanical strength of the regenerated tendon in the repair group was highly improved. Most importantly, the expressions of integrin αv and its downstream and the phosphorylation levels of FAK and ERK were remarkably higher in the repair group than in the other groups. Furthermore, blocking FAK or ERK with selective inhibitors PF573228 and U0126 resulted in obvious adverse effects on the histological structure of the regenerated Achilles tendon. In summary, this study demonstrated that dynamic tensile stress based on the PRBT could effectively promote the regeneration of the Achilles tendon, suggesting that dynamic tensile stress enhances the cell proliferation and tenogenic differentiation via the activation of the integrin/FAK/ERK signaling pathway.
Collapse
Affiliation(s)
- Zhi Li
- Department of Orthopedic, Center for Joint Surgery, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road No.76, Yuzhong District, Chongqing, 400010, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Repair and Rehabilitation, Daping Hospital, Trauma Center, Research Institute of Surgery, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Orthopedic, Center for Joint Surgery, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road No.76, Yuzhong District, Chongqing, 400010, China
| | - Chen Shen
- Department of Orthopedic, Center for Joint Surgery, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road No.76, Yuzhong District, Chongqing, 400010, China
| | - Zhibo Deng
- Department of Orthopedic, Center for Joint Surgery, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road No.76, Yuzhong District, Chongqing, 400010, China
| | - Kaiying Tang
- Department of Orthopedic, Center for Joint Surgery, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road No.76, Yuzhong District, Chongqing, 400010, China
| | - Yangli Xie
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Repair and Rehabilitation, Daping Hospital, Trauma Center, Research Institute of Surgery, Army Medical University, Chongqing, China.
| | - Lin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Repair and Rehabilitation, Daping Hospital, Trauma Center, Research Institute of Surgery, Army Medical University, Chongqing, China.
| | - Mao Nie
- Department of Orthopedic, Center for Joint Surgery, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road No.76, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
10
|
Lu J, Chen H, Lyu K, Jiang L, Chen Y, Long L, Wang X, Shi H, Li S. The Functions and Mechanisms of Tendon Stem/Progenitor Cells in Tendon Healing. Stem Cells Int 2023; 2023:1258024. [PMID: 37731626 PMCID: PMC10509002 DOI: 10.1155/2023/1258024] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Tendon injury is one of the prevalent disorders of the musculoskeletal system in orthopedics and is characterized by pain and limitation of joint function. Due to the difficulty of spontaneous tendon healing, and the scar tissue and low mechanical properties that usually develops after healing. Therefore, the healing of tendon injury remains a clinical challenge. Although there are a multitude of approaches to treating tendon injury, the therapeutic effects have not been satisfactory to date. Recent studies have shown that stem cell therapy has a facilitative effect on tendon healing. In particular, tendon stem/progenitor cells (TSPCs), a type of stem cell from tendon tissue, play an important role not only in tendon development and tendon homeostasis, but also in tendon healing. Compared to other stem cells, TSPCs have the potential to spontaneously differentiate into tenocytes and express higher levels of tendon-related genes. TSPCs promote tendon healing by three mechanisms: modulating the inflammatory response, promoting tenocyte proliferation, and accelerating collagen production and balancing extracellular matrix remodeling. However, current investigations have shown that TSPCs also have a negative effect on tendon healing. For example, misdifferentiation of TSPCs leads to a "failed healing response," which in turn leads to the development of chronic tendon injury (tendinopathy). The focus of this paper is to describe the characteristics of TSPCs and tenocytes, to demonstrate the roles of TSPCs in tendon healing, while discussing the approaches used to culture and differentiate TSPCs. In addition, the limitations of TSPCs in clinical application and their potential therapeutic strategies are elucidated.
Collapse
Affiliation(s)
- Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Hui Chen
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Longhai Long
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoqiang Wang
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Houyin Shi
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
11
|
Shou Y, Liu L, Liu Q, Le Z, Lee KL, Li H, Li X, Koh DZ, Wang Y, Liu TM, Yang Z, Lim CT, Cheung C, Tay A. Mechano-responsive hydrogel for direct stem cell manufacturing to therapy. Bioact Mater 2023; 24:387-400. [PMID: 36632503 PMCID: PMC9817177 DOI: 10.1016/j.bioactmat.2022.12.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cell (MSC) is one of the most actively studied cell types due to its regenerative potential and immunomodulatory properties. Conventional cell expansion methods using 2D tissue culture plates and 2.5D microcarriers in bioreactors can generate large cell numbers, but they compromise stem cell potency and lack mechanical preconditioning to prepare MSC for physiological loading expected in vivo. To overcome these challenges, in this work, we describe a 3D dynamic hydrogel using magneto-stimulation for direct MSC manufacturing to therapy. With our technology, we found that dynamic mechanical stimulation (DMS) enhanced matrix-integrin β1 interactions which induced MSCs spreading and proliferation. In addition, DMS could modulate MSC biofunctions including directing MSC differentiation into specific lineages and boosting paracrine activities (e.g., growth factor secretion) through YAP nuclear localization and FAK-ERK pathway. With our magnetic hydrogel, complex procedures from MSC manufacturing to final clinical use, can be integrated into one single platform, and we believe this 'all-in-one' technology could offer a paradigm shift to existing standards in MSC therapy.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Ling Liu
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| | - Qimin Liu
- School of Civil Engineering and Architecture, Wuhan University of Technology, 430070, Wuhan, China
| | - Zhicheng Le
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
| | - Hua Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Dion Zhanyun Koh
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Yuwen Wang
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Tong Ming Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Zheng Yang
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
12
|
Fragoulis A, Tohidnezhad M, Kubo Y, Wruck CJ, Craveiro RB, Bock A, Wolf M, Pufe T, Jahr H, Suhr F. The Contribution of the Nrf2/ARE System to Mechanotransduction in Musculoskeletal and Periodontal Tissues. Int J Mol Sci 2023; 24:ijms24097722. [PMID: 37175428 PMCID: PMC10177782 DOI: 10.3390/ijms24097722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Mechanosensing plays an essential role in maintaining tissue functions. Across the human body, several tissues (i.e., striated muscles, bones, tendons, ligaments, as well as cartilage) require mechanical loading to exert their physiological functions. Contrary, mechanical unloading triggers pathological remodeling of these tissues and, consequently, human body dysfunctions. At the cellular level, both mechanical loading and unloading regulate a wide spectrum of cellular pathways. Among those, pathways regulated by oxidants such as reactive oxygen species (ROS) represent an essential node critically controlling tissue organization and function. Hence, a sensitive balance between the generation and elimination of oxidants keeps them within a physiological range. Here, the Nuclear Factor-E2-related factor 2/Antioxidant response element (Nrf2/ARE) system plays an essential role as it constitutes the major cellular regulation against exogenous and endogenous oxidative stresses. Dysregulations of this system advance, i.a., liver, neurodegenerative, and cancer diseases. Herein, we extend our comprehension of the Nrf2 system to the aforementioned mechanically sensitive tissues to explore its role in their physiology and pathology. We demonstrate the relevance of it for the tissues' functionality and highlight the imperative to further explore the Nrf2 system to understand the physiology and pathology of mechanically sensitive tissues in the context of redox biology.
Collapse
Affiliation(s)
- Athanassios Fragoulis
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Yusuke Kubo
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Christoph Jan Wruck
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Rogerio Bastos Craveiro
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna Bock
- Department of Oral and Maxillofacial Surgery, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Michael Wolf
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Holger Jahr
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
- Institute of Structural Mechanics and Lightweight Design, RWTH Aachen University, 52062 Aachen, Germany
| | - Frank Suhr
- Division of Molecular Exercise Physiology, Faculty of Life Sciences: Food, Nutrition and Health, University of Bayreuth, 95326 Kulmbach, Germany
| |
Collapse
|
13
|
Fu S, Lan Y, Wang G, Bao D, Qin B, Zheng Q, Liu H, Wong VKW. External stimulation: A potential therapeutic strategy for tendon-bone healing. Front Bioeng Biotechnol 2023; 11:1150290. [PMID: 37064229 PMCID: PMC10102526 DOI: 10.3389/fbioe.2023.1150290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Injuries at the tendon-bone interface are very common in the field of sports medicine, and healing at the tendon-bone interface is complex. Injuries to the tendon-bone interface can seriously affect a patient’s quality of life, so it is essential to restore stability and promote healing of the tendon-bone interface. In addition to surgical treatment, the healing of tendons and bones can also be properly combined with extracorporeal stimulation therapy during the recovery process. In this review, we discuss the effects of extracorporeal shock waves (ESWs), low-intensity pulsed ultrasound (LIPUS), and mechanical stress on tendon-bone healing, focusing on the possible mechanisms of action of mechanical stress on tendon-bone healing in terms of transcription factors and biomolecules. The aim is to provide possible therapeutic approaches for subsequent clinical treatment.
Collapse
Affiliation(s)
- Shijie Fu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Yujian Lan
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Guoyou Wang
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Dingsu Bao
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Qin
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiu Zheng
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Huan Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
- *Correspondence: Huan Liu, ; Vincent Kam Wai Wong,
| | - Vincent Kam Wai Wong
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
- *Correspondence: Huan Liu, ; Vincent Kam Wai Wong,
| |
Collapse
|
14
|
Vinhas A, Almeida AF, Rodrigues MT, Gomes ME. Prospects of magnetically based approaches addressing inflammation in tendon tissues. Adv Drug Deliv Rev 2023; 196:114815. [PMID: 37001644 DOI: 10.1016/j.addr.2023.114815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
Tendon afflictions constitute a significant share of musculoskeletal diseases and represent a primary cause of incapacity worldwide. Unresolved/chronic inflammatory states have been associated with the onset and progression of tendon disorders, contributing to undesirable immune stimulation and detrimental tissue effects. Thus, targeting persistent inflammatory events could assist important developments to solve pathophysiological processes and innovative therapeutics to address impaired healing and accomplish complete tendon regeneration. This review overviews the impact of inflammation and inflammatory mediators in tendon niches, unveiling the importance of tendon cell populations and their signature features, and the influence of microenvironmental factors on inflamed and injured tendons. The demand for non-invasive instructive strategies to manage persistent inflammatory mediators, guide inflammatory pathways, and modulate cellular responses will also be approached by exploring the role of pulsed electromagnetic field (PEMF). PEMF alone or combined with more sophisticated systems triggered by magnetic fields will be considered in the design of successful therapies to control inflammation in tendinopathic conditions.
Collapse
|
15
|
Wang P, Meng X, Xue J, Fan C, Wang J. Genome-wide analysis for nanofiber induced global gene expression profile: A study in MC3T3-E1 cells by RNA-Seq. Colloids Surf B Biointerfaces 2023; 223:113143. [PMID: 36682297 DOI: 10.1016/j.colsurfb.2023.113143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/01/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Nanofibers are one of the attractive biomaterials that can provide unique environments to direct cell behaviors. However, how nanofiber structure affects the global gene expression of laden cells remains unclear. Herein, high-throughput mRNA sequencing (RNA-seq) is applied to analyze the transcriptome of the MC3T3-E1 cells (a model osteoblast cell line) cultured on electrospun nanofibers. The cell-adhesive poly(L-lactide) nanofibers and membranes are developed by the mussel-inspired coating of gelatin-dopamine conjugate under H2O2-mediated oxidation. The MC3T3-E1 cells cultured on nanofibers exhibit elongated morphology and increased proliferation compared with those on membranes. The differences in global gene expression profiles are determined by RNA-seq, in which 905 differentially expressed genes (DEGs) are identified. Significantly, the DEGs related to cytoskeleton, promotion of cell cycle progression, cell adhesion, and cell proliferation, are higher expressed in the cells on nanofibers, while the DEGs involved in cell-cycle arrest and osteoblast mineralization are up-regulated in the cells on membranes. This study elucidates the roles of nanofiber structure in affecting gene expression of laden cells at the whole transcriptome level, and it will lay the foundation for understanding nanofiber-guided cell behaviors.
Collapse
Affiliation(s)
- Peiyan Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, Shandong, PR China
| | - Xinyue Meng
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China
| | - Junqiang Xue
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, Shandong, PR China
| | - Changjiang Fan
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, Shandong, PR China.
| | - Jianxun Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, Shandong, PR China.
| |
Collapse
|
16
|
Citro V, Clerici M, Boccaccini AR, Della Porta G, Maffulli N, Forsyth NR. Tendon tissue engineering: An overview of biologics to promote tendon healing and repair. J Tissue Eng 2023; 14:20417314231196275. [PMID: 37719308 PMCID: PMC10501083 DOI: 10.1177/20417314231196275] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/06/2023] [Indexed: 09/19/2023] Open
Abstract
Tendons are dense connective tissues with a hierarchical polarized structure that respond to and adapt to the transmission of muscle contraction forces to the skeleton, enabling motion and maintaining posture. Tendon injuries, also known as tendinopathies, are becoming more common as populations age and participation in sports/leisure activities increases. The tendon has a poor ability to self-heal and regenerate given its intrinsic, constrained vascular supply and exposure to frequent, severe loading. There is a lack of understanding of the underlying pathophysiology, and it is not surprising that disorder-targeted medicines have only been partially effective at best. Recent tissue engineering approaches have emerged as a potential tool to drive tendon regeneration and healing. In this review, we investigated the physiochemical factors involved in tendon ontogeny and discussed their potential application in vitro to reproduce functional and self-renewing tendon tissue. We sought to understand whether stem cells are capable of forming tendons, how they can be directed towards the tenogenic lineage, and how their growth is regulated and monitored during the entire differentiation path. Finally, we showed recent developments in tendon tissue engineering, specifically the use of mesenchymal stem cells (MSCs), which can differentiate into tendon cells, as well as the potential role of extracellular vesicles (EVs) in tendon regeneration and their potential for use in accelerating the healing response after injury.
Collapse
Affiliation(s)
- Vera Citro
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire, UK
- Department of Materials Science and Engineering, Institute of Biomaterials University of Erlangen-Nuremberg, Cauerstrasse 6, Erlangen, Germany
| | - Marta Clerici
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire, UK
- Department of Medicine, Surgery and Dentistry, University of Salerno, via S. Allende, Baronissi, Salerno, Italy
| | - Aldo R. Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials University of Erlangen-Nuremberg, Cauerstrasse 6, Erlangen, Germany
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, via S. Allende, Baronissi, Salerno, Italy
- Interdepartmental Centre BIONAM, University of Salerno, via Giovanni Paolo I, Fisciano, Salerno, Italy
| | - Nicola Maffulli
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire, UK
- Department of Medicine, Surgery and Dentistry, University of Salerno, via S. Allende, Baronissi, Salerno, Italy
- Department of Trauma and Orthopaedic Surgery, University Hospital ‘San Giovanni di Dio e Ruggi D’Aragona’, Salerno, Italy
| | - Nicholas R. Forsyth
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire, UK
- Vice Principals’ Office, University of Aberdeen, Kings College, Aberdeen, UK
| |
Collapse
|
17
|
Asymptomatic Hyperuricemia Is Associated with Achilles Tendon Rupture through Disrupting the Normal Functions of Tendon Stem/Progenitor Cells. Stem Cells Int 2022; 2022:6795573. [PMID: 36504525 PMCID: PMC9731760 DOI: 10.1155/2022/6795573] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Hyperuricemia is a metabolic disorder that is essential to the development of inflammatory gout, with increasing prevalence over recent years. Emerging clinical findings has evidenced remarkable tendon damage in individuals with longstanding asymptomatic hyperuricemia, yet the impact of hyperuricemia on tendon homeostasis and associated repercussions is largely unknown. Here, we investigated whether asymptomatic hyperuricemia was associated with spontaneous ruptures in the Achilles tendon and the pathological effect of hyperuricemia on the tendon stem/progenitor cells (TSPCs). Significantly higher serum uric acid (SUA) levels were found in 648 closed Achilles tendon rupture (ATR) patients comparing to those in 12559 healthy volunteers. In vitro study demonstrated that uric acid (UA) dose dependently reduced rat Achilles TSPC viability, decreased the expressions of tendon collagens, and deformed their structural organization while significantly increased the transcript levels of matrix degradative enzymes and proinflammatory factors. Consistently, marked disruptions in Achilles tendon tissue structural and functional integrity were found in a rat model of hyperuricemia, together with enhanced immune cell infiltration. Transcriptome analysis revealed a significant elevation in genes involved in metabolic stress and tissue degeneration in TSPCs challenged by hyperuricemia. Specifically, reduced activity of the AKT-mTOR pathway with enhanced autophagic signaling was confirmed. Our findings indicate that asymptomatic hyperuricemia may be a predisposition of ATR by impeding the normal functions of TSPCs. This information may provide theoretical and experimental basis for exploring the early prevention and care of ATR.
Collapse
|
18
|
Gomez-Florit M, Labrador-Rached CJ, Domingues RM, Gomes ME. The tendon microenvironment: Engineered in vitro models to study cellular crosstalk. Adv Drug Deliv Rev 2022; 185:114299. [PMID: 35436570 DOI: 10.1016/j.addr.2022.114299] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022]
Abstract
Tendinopathy is a multi-faceted pathology characterized by alterations in tendon microstructure, cellularity and collagen composition. Challenged by the possibility of regenerating pathological or ruptured tendons, the healing mechanisms of this tissue have been widely researched over the past decades. However, so far, most of the cellular players and processes influencing tendon repair remain unknown, which emphasizes the need for developing relevant in vitro models enabling to study the complex multicellular crosstalk occurring in tendon microenvironments. In this review, we critically discuss the insights on the interaction between tenocytes and the other tendon resident cells that have been devised through different types of existing in vitro models. Building on the generated knowledge, we stress the need for advanced models able to mimic the hierarchical architecture, cellularity and physiological signaling of tendon niche under dynamic culture conditions, along with the recreation of the integrated gradients of its tissue interfaces. In a forward-looking vision of the field, we discuss how the convergence of multiple bioengineering technologies can be leveraged as potential platforms to develop the next generation of relevant in vitro models that can contribute for a deeper fundamental knowledge to develop more effective treatments.
Collapse
|
19
|
Dede Eren A, Vermeulen S, Schmitz TC, Foolen J, de Boer J. The loop of phenotype: Dynamic reciprocity links tenocyte morphology to tendon tissue homeostasis. Acta Biomater 2022; 163:275-286. [PMID: 35584748 DOI: 10.1016/j.actbio.2022.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/24/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
Cells and their surrounding extracellular matrix (ECM) are engaged in dynamic reciprocity to maintain tissue homeostasis: cells deposit ECM, which in turn presents the signals that define cell identity. This loop of phenotype is obvious for biochemical signals, such as collagens, which are produced by and presented to cells, but the role of biomechanical signals is also increasingly recognised. In addition, cell shape goes hand in hand with cell function and tissue homeostasis. Aberrant cell shape and ECM is seen in pathological conditions, and control of cell shape in micro-fabricated platforms disclose the causal relationship between cell shape and cell function, often mediated by mechanotransduction. In this manuscript, we discuss the loop of phenotype for tendon tissue homeostasis. We describe cell shape and ECM organization in normal and diseased tissue, how ECM composition influences tenocyte shape, and how that leads to the activation of signal transduction pathways and ECM deposition. We further describe the use of technologies to control cell shape to elucidate the link between cell shape and its phenotypical markers and focus on the causal role of cell shape in the loop of phenotype. STATEMENT OF SIGNIFICANCE: The dynamic reciprocity between cells and their surrounding extracellular matrix (ECM) influences biomechanical and biochemical properties of ECM as well as cell function through activation of signal transduction pathways that regulate gene and protein expression. We refer to this reciprocity as Loop of Phenotype and it has been studied and demonstrated extensively by using micro-fabricated platforms to manipulate cell shape and cell fate. In this manuscript, we discuss this concept in tendon tissue homeostasis by giving examples in healthy and pathological tenson tissue. Furthermore, we elaborate this by showing how biomaterials are used to feed this reciprocity to manipulate cell shape and function. Finally, we elucidate the link between cell shape and its phenotypical markers and focus on the activation of signal transduction pathways and ECM deposition.
Collapse
Affiliation(s)
- Aysegul Dede Eren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Steven Vermeulen
- Maastricht University, MERLN Institute for Technology Inspired Regenerative Medicine, Instructive Biomaterial Engineering, Maastricht, the Netherlands
| | - Tara C Schmitz
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jasper Foolen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
20
|
Chatterjee M, Muljadi PM, Andarawis-Puri N. The role of the tendon ECM in mechanotransduction: disruption and repair following overuse. Connect Tissue Res 2022; 63:28-42. [PMID: 34030531 DOI: 10.1080/03008207.2021.1925663] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Tendon overuse injuries are prevalent conditions with limited therapeutic options to halt disease progression. The specialized extracellular matrix (ECM) both enables joint function and mediates mechanical signals to tendon cells, driving biological responses to exercise or injury. With overuse, tendon ECM composition and structure changes at multiple scales, disrupting mechanotransduction and resulting in inadequate repair and disease progression. This review highlights the multiscale ECM changes that occur with tendon overuse and corresponding effects on cell-matrix interactions and cellular response to load.Results: Different functional joint requirements and tendon types experience a wide range of loading profiles, creating varied downstream mechanical stimuli. Distinct ECM structure and mechanical properties within the fascicle matrix, interfascicle matrix, and enthesis and their varied disruption with overuse are considered. The pericellular matrix (PCM) comprising the microscale tendon cell environment has a unique composition that changes with overuse injury and exercise, suggesting an important role in mechanotransduction and promoting repair. Cell-matrix interactions are mediated by structures including cilia, integrins, connexins and cytoskeleton that signal downstream homeostasis, adaptation, or repair. ECM disruption with tendon overuse may cause altered mechanical loading and cell-matrix interactions, resulting in mechanobiological understimulation, apoptosis, and ineffective repair. Current interventions to promote repair of tendon overuse injuries including exercise, targeting cell signaling, and modulating inflammation are considered.Conclusion: Future therapeutics should be assessed with regard of their effects on multiscale mechanotransduction in addition to joint function, with consideration of the central role of ECM.
Collapse
Affiliation(s)
- Monideepa Chatterjee
- Nancy E. And Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Patrick M Muljadi
- Nancy E. And Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 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
|
21
|
Fernandez‐Yague MA, Trotier A, Demir S, Abbah SA, Larrañaga A, Thirumaran A, Stapleton A, Tofail SAM, Palma M, Kilcoyne M, Pandit A, Biggs MJ. A Self-Powered Piezo-Bioelectric Device Regulates Tendon Repair-Associated Signaling Pathways through Modulation of Mechanosensitive Ion Channels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2008788. [PMID: 34423493 PMCID: PMC11468587 DOI: 10.1002/adma.202008788] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/17/2021] [Indexed: 06/13/2023]
Abstract
Tendon disease constitutes an unmet clinical need and remains a critical challenge in the field of orthopaedic surgery. Innovative solutions are required to overcome the limitations of current tendon grafting approaches, and bioelectronic therapies show promise in treating musculoskeletal diseases, accelerating functional recovery through the activation of tissue regeneration-specific signaling pathways. Self-powered bioelectronic devices, particularly piezoelectric materials, represent a paradigm shift in biomedicine, negating the need for battery or external powering and complementing existing mechanotherapy to accelerate the repair processes. Here, the dynamic response of tendon cells to a piezoelectric collagen-analogue scaffold comprised of aligned nanoscale fibers made of the ferroelectric material poly(vinylidene fluoride-co-trifluoroethylene) is shown. It is demonstrated that motion-powered electromechanical stimulation of tendon tissue through piezo-bioelectric device results in ion channel modulation in vitro and regulates specific tissue regeneration signaling pathways. Finally, the potential of the piezo-bioelectronic device in modulating the progression of tendinopathy-associated processes in vivo, using a rat Achilles acute injury model is shown. This study indicates that electromechanical stimulation regulates mechanosensitive ion channel sensitivity and promotes tendon-specific over non-tenogenic tissue repair processes.
Collapse
Affiliation(s)
- Marc A. Fernandez‐Yague
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Alexandre Trotier
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Secil Demir
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Sunny Akogwu Abbah
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Aitor Larrañaga
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
- University of the Basque CountryDepartment of Mining‐Metallurgy Engineering and Materials Science and POLYMATBarrio SarrienaBilbao48013Spain
| | - Arun Thirumaran
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Aimee Stapleton
- University of LimerickDepartment of PhysicsLimerickV94 T9PXIreland
| | | | - Matteo Palma
- Queen Mary University of LondonMaterials Research Institute and School of Biological and Chemical SciencesMile End RoadLondonE1 4NSUK
| | - Michelle Kilcoyne
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Manus J. Biggs
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| |
Collapse
|
22
|
Zhang S, Ju W, Chen X, Zhao Y, Feng L, Yin Z, Chen X. Hierarchical ultrastructure: An overview of what is known about tendons and future perspective for tendon engineering. Bioact Mater 2021; 8:124-139. [PMID: 34541391 PMCID: PMC8424392 DOI: 10.1016/j.bioactmat.2021.06.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022] Open
Abstract
Abnormal tendons are rarely ever repaired to the natural structure and morphology of normal tendons. To better guide the repair and regeneration of injured tendons through a tissue engineering method, it is necessary to have insights into the internal morphology, organization, and composition of natural tendons. This review summarized recent researches on the structure and function of the extracellular matrix (ECM) components of tendons and highlight the application of multiple detection methodologies concerning the structure of ECMs. In addition, we look forward to the future of multi-dimensional biomaterial design methods and the potential of structural repair for tendon ECM components. In addition, focus is placed on the macro to micro detection methods for tendons, and current techniques for evaluating the extracellular matrix of tendons at the micro level are introduced in detail. Finally, emphasis is given to future extracellular matrix detection methods, as well as to how future efforts could concentrate on fabricating the biomimetic tendons. Summarize recent research on the structure and function of the extracellular matrix (ECM) components of tendons. Comments on current research methods concerning the structure of ECMs. Perspective on the future of multi-dimensional detection techniques and structural repair of tendon ECM components.
Collapse
Affiliation(s)
- Shichen Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310052, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Wei Ju
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoyi Chen
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Guangxi Medical University, Guangxi, 530021, China
| | - Yanyan Zhao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310052, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Lingchong Feng
- State Key Laboratory of Clean Energy Utilization, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zi Yin
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine and Regenerative Medicine and Department of Orthopedic Surgery of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310052, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Guangxi Medical University, Guangxi, 530021, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| |
Collapse
|
23
|
Lim KT, Patel DK, Dutta SD, Ganguly K. Fluid Flow Mechanical Stimulation-Assisted Cartridge Device for the Osteogenic Differentiation of Human Mesenchymal Stem Cells. MICROMACHINES 2021; 12:927. [PMID: 34442549 PMCID: PMC8398302 DOI: 10.3390/mi12080927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022]
Abstract
Human mesenchymal stem cells (hMSCs) have the potential to differentiate into different types of mesodermal tissues. In vitro proliferation and differentiation of hMSCs are necessary for bone regeneration in tissue engineering. The present study aimed to design and develop a fluid flow mechanically-assisted cartridge device to enhance the osteogenic differentiation of hMSCs. We used the fluorescence-activated cell-sorting method to analyze the multipotent properties of hMSCs and found that the cultured cells retained their stemness potential. We also evaluated the cell viabilities of the cultured cells via water-soluble tetrazolium salt 1 (WST-1) assay under different rates of flow (0.035, 0.21, and 0.35 mL/min) and static conditions and found that the cell growth rate was approximately 12% higher in the 0.035 mL/min flow condition than the other conditions. Moreover, the cultured cells were healthy and adhered properly to the culture substrate. Enhanced mineralization and alkaline phosphatase activity were also observed under different perfusion conditions compared to the static conditions, indicating that the applied conditions play important roles in the proliferation and differentiation of hMSCs. Furthermore, we determined the expression levels of osteogenesis-related genes, including the runt-related protein 2 (Runx2), collagen type I (Col1), osteopontin (OPN), and osteocalcin (OCN), under various perfusion vis-à-vis static conditions and found that they were significantly affected by the applied conditions. Furthermore, the fluorescence intensities of OCN and OPN osteogenic gene markers were found to be enhanced in the 0.035 mL/min flow condition compared to the control, indicating that it was a suitable condition for osteogenic differentiation. Taken together, the findings of this study reveal that the developed cartridge device promotes the proliferation and differentiation of hMSCs and can potentially be used in the field of tissue engineering.
Collapse
Affiliation(s)
- Ki-Taek Lim
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
- Biomechagen Co., Ltd., Chuncheon 24341, Korea
| | - Dinesh-K. Patel
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| | - Sayan-Deb Dutta
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| | - Keya Ganguly
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| |
Collapse
|
24
|
Nie D, Zhou Y, Wang W, Zhang J, Wang JHC. Mechanical Overloading Induced-Activation of mTOR Signaling in Tendon Stem/Progenitor Cells Contributes to Tendinopathy Development. Front Cell Dev Biol 2021; 9:687856. [PMID: 34322484 PMCID: PMC8311934 DOI: 10.3389/fcell.2021.687856] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/18/2021] [Indexed: 01/08/2023] Open
Abstract
Despite the importance of mechanical loading in tendon homeostasis and pathophysiology, the molecular responses involved in the mechanotransduction in tendon cells remain unclear. In this study, we found that in vitro mechanical loading activated the mammalian target of rapamycin (mTOR) in rat patellar tendon stem/progenitor cells (TSCs) in a stretching magnitude-dependent manner. Application of rapamycin, a specific inhibitor of mTOR, attenuated the phosphorylation of S6 and 4E-BP1 and as such, largely inhibited the mechanical activation of mTOR. Moreover, rapamycin significantly decreased the proliferation and non-tenocyte differentiation of PTSCs as indicated by the reduced expression levels of LPL, PPARγ, SOX-9, collagen II, Runx-2, and osteocalcin genes. In the animal studies, mice subjected to intensive treadmill running (ITR) developed tendon degeneration, as evidenced by the formation of round-shaped cells, accumulation of proteoglycans, and expression of SOX-9 and collagen II proteins. However, daily injections of rapamycin in ITR mice reduced all these tendon degenerative changes. Collectively, these findings suggest that mechanical loading activates the mTOR signaling in TSCs, and rapamycin may be used to prevent tendinopathy development by blocking non-tenocyte differentiation due to mechanical over-activation of mTOR in TSCs.
Collapse
Affiliation(s)
- Daibang Nie
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yiqin Zhou
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wang Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - James H.-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
25
|
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
|
26
|
Klatte-Schulz F, Bormann N, Voss I, Melzer J, Schmock A, Bucher CH, Thiele K, Moroder P, Haffner-Luntzer M, Ignatius A, Duda GN, Wildemann B. Bursa-Derived Cells Show a Distinct Mechano-Response to Physiological and Pathological Loading in vitro. Front Cell Dev Biol 2021; 9:657166. [PMID: 34136480 PMCID: PMC8201779 DOI: 10.3389/fcell.2021.657166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
The mechano-response of highly loaded tissues such as bones or tendons is well investigated, but knowledge regarding the mechano-responsiveness of adjacent tissues such as the subacromial bursa is missing. For a better understanding of the physiological role of the bursa as a friction-reducing structure in the joint, the study aimed to analyze whether and how bursa-derived cells respond to physiological and pathological mechanical loading. This might help to overcome some of the controversies in the field regarding the role of the bursa in the development and healing of shoulder pathologies. Cells of six donors seeded on collagen-coated silicon dishes were stimulated over 3 days for 1 or 4 h with 1, 5, or 10% strain. Orientation of the actin cytoskeleton, YAP nuclear translocation, and activation of non-muscle myosin II (NMM-II) were evaluated for 4 h stimulations to get a deeper insight into mechano-transduction processes. To investigate the potential of bursa-derived cells to adapt their matrix formation and remodeling according to mechanical loading, outcome measures included cell viability, gene expression of extracellular matrix and remodeling markers, and protein secretions. The orientation angle of the actin cytoskeleton increased toward a more perpendicular direction with increased loading and lowest variations for the 5% loading group. With 10% tension load, cells were visibly stressed, indicated by loss in actin density and slightly reduced cell viability. A significantly increased YAP nuclear translocation occurred for the 1% loading group with a similar trend for the 5% group. NMM-II activation was weak for all stimulation conditions. On the gene expression level, only the expression of TIMP2 was down-regulated in the 1 h group compared to control. On the protein level, collagen type I and MMP2 increased with higher/longer straining, respectively, whereas TIMP1 secretion was reduced, resulting in an MMP/TIMP imbalance. In conclusion, this study documents for the first time a clear mechano-responsiveness in bursa-derived cells with activation of mechano-transduction pathways and thus hint to a physiological function of mechanical loading in bursa-derived cells. This study represents the basis for further investigations, which might lead to improved treatment options of subacromial bursa-related pathologies in the future.
Collapse
Affiliation(s)
- Franka Klatte-Schulz
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nicole Bormann
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Isabel Voss
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Josephine Melzer
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Aysha Schmock
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kathi Thiele
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Moroder
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Ulm University, Ulm, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Britt Wildemann
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental Trauma Surgery, Department of Trauma-, Hand- and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
27
|
Wei B, Lu J. Characterization of Tendon-Derived Stem Cells and Rescue Tendon Injury. Stem Cell Rev Rep 2021; 17:1534-1551. [PMID: 33651334 DOI: 10.1007/s12015-021-10143-9] [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] [Accepted: 02/21/2021] [Indexed: 12/12/2022]
Abstract
The natural healing ability of tendon is limited, and it cannot restore the native structure and function of tendon injuries. Tendon-derived stem cells (TDSCs) are a new type of pluripotent stem cells with multi-directional differentiation potential and are expected to become a promising cell-seed for the treatment of tendon injuries in the future. In this review, we outline the latest advances in the culture and identification of TDSCs. In addition, the influencing factors on the differentiation of TDSCs are discussed. Moreover, we aim to discuss recent studies to enhance TDSCs treatment of injured tendons. Finally, we identify the limitations of the current understanding of TDSCs biology, the main challenges of using their use, and potential therapeutic strategies to inform cell-based tendon repair.
Collapse
Affiliation(s)
- Bing Wei
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jun Lu
- Department of Orthopaedic Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Bochon K, Zielniok K, Gawlak M, Zawada K, Zarychta-Wiśniewska W, Siennicka K, Struzik S, Pączek L, Burdzińska A. The Effect of L-Ascorbic Acid and Serum Reduction on Tenogenic Differentiation of Human Mesenchymal Stromal Cells. Int J Stem Cells 2021; 14:33-46. [PMID: 33122467 PMCID: PMC7904532 DOI: 10.15283/ijsc20023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 08/02/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background and Objectives Despite significant improvement in the treatment of tendon injuries, the full tissue recovery is often not possible because of its limited ability to auto-repair. The transplantation of mesenchymal stromal cells (MSCs) is considered as a novel approach in the treatment of tendinopathies. The question about the optimal culture conditions remains open. In this study we aimed to investigate if serum reduction, L-ascorbic acid supplementation or a combination of both factors can induce tenogenic differentiation of human adipose-derived MSCs (ASCs). Methods and Results Human ASCs from 3 healthy donors were used in the study. The tested conditions were: 0.5 mM of ascorbic acid 2-phosphate (AA-2P), reduced serum content (2% FBS) or combination of these two factors. The combination of AA-2P and 2% FBS was the only experimental condition that caused a significant increase of the expression of all analyzed genes related to tenogenesis (SCLERAXIS, MOHAWK, COLLAGEN_1, COLLAGEN_3, DECORIN) in comparison to the untreated control (evaluated by RT-PCR, 5th day of experiment). Moreover, this treatment significantly increased the synthesis of SCLERAXIS, MOHAWK, COLLAGEN_1, COLLAGEN_3 proteins at the same time point (evaluated by Western blot method). Double immunocytochemical staining revealed that AA-2P significantly increased the extracellular deposition of both types of collagens. Semi-quantitative Electron Spin Resonance analysis of ascorbyl free radical revealed that AA-2P do not induce harmful transition metals-driven redox reactions in cell culture media. Conclusions Obtained results justify the use of reduced content of serum with the addition of 0.5 mM of AA-2P in tenogenic inducing media.
Collapse
Affiliation(s)
- Karolina Bochon
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Zielniok
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Gawlak
- Department of Pharmacodynamics and Pathophysiology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Zawada
- Department of Physical Chemistry, Faculty of Pharmacy with the Laboratory Medicine Division, Medical University of Warsaw, Warsaw, Poland
| | | | - Katarzyna Siennicka
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Sławomir Struzik
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland.,Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Burdzińska
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
29
|
Bianchi E, Ruggeri M, Rossi S, Vigani B, Miele D, Bonferoni MC, Sandri G, Ferrari F. Innovative Strategies in Tendon Tissue Engineering. Pharmaceutics 2021; 13:89. [PMID: 33440840 PMCID: PMC7827834 DOI: 10.3390/pharmaceutics13010089] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022] Open
Abstract
The tendon is a highly aligned connective tissue that transmits force from muscle to bone. Each year, more than 32 million tendon injuries have been reported, in fact, tendinopathies represent at least 50% of all sports injuries, and their incidence rates have increased in recent decades due to the aging population. Current clinical grafts used in tendon treatment are subject to several restrictions and there is a significant demand for alternative engineered tissue. For this reason, innovative strategies need to be explored. Tendon replacement and regeneration are complex since scaffolds need to guarantee an adequate hierarchical structured morphology and mechanical properties to stand the load. Moreover, to guide cell proliferation and growth, scaffolds should provide a fibrous network that mimics the collagen arrangement of the extracellular matrix in the tendons. This review focuses on tendon repair and regeneration. Particular attention has been devoted to the innovative approaches in tissue engineering. Advanced manufacturing techniques, such as electrospinning, soft lithography, and three-dimensional (3D) printing, have been described. Furthermore, biological augmentation has been considered, as an emerging strategy with great therapeutic potential.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (E.B.); (M.R.); (S.R.); (B.V.); (D.M.); (M.C.B.); (F.F.)
| | | |
Collapse
|
30
|
Im GI, Kim TK. Stem Cells for the Regeneration of Tendon and Ligament: A Perspective. Int J Stem Cells 2020; 13:335-341. [PMID: 33122471 PMCID: PMC7691853 DOI: 10.15283/ijsc20091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/12/2020] [Accepted: 08/16/2020] [Indexed: 12/12/2022] Open
Abstract
Tendons are structures that connect muscles to the bones in our body and transmit the force generated by contraction of the muscles to the bones. Ligaments are structures that connect bones to bones, with histological properties similar to tendons. In tendon and ligament tissue, there are very small amounts of cells similar to mesenchymal stem cells (MSCs) called tendon stem/progenitor cells (TSPCs), or tenogenic stem cells. While the role of specific growth factors and transcription factors is well established in the osteogenic and chondrogenic differentiation of stem cells, a consensus has not been established for tenogenic differentiation. Injuries to tendons and ligaments are very common, but natural healing is very slow and inefficient due to limited vascularization. Currently, there is no adequate method for restoring extensive tendon or ligament defects. Procedures addressing the unmet need for regeneration of these tissues are needed. In this review, the current knowledge, as well as the authors’ ideas and perspective on stem cell and regenerative medicine for tendon and ligament defects are presented.
Collapse
Affiliation(s)
- Gun-Il Im
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Korea
| | - Tae-Kyung Kim
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Korea
| |
Collapse
|
31
|
Dyment NA, Barrett JG, Awad H, Bautista CA, Banes A, Butler DL. A brief history of tendon and ligament bioreactors: Impact and future prospects. J Orthop Res 2020; 38:2318-2330. [PMID: 32579266 PMCID: PMC7722018 DOI: 10.1002/jor.24784] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/28/2020] [Accepted: 06/12/2020] [Indexed: 02/04/2023]
Abstract
Bioreactors are powerful tools with the potential to model tissue development and disease in vitro. For nearly four decades, bioreactors have been used to create tendon and ligament tissue-engineered constructs in order to define basic mechanisms of cell function, extracellular matrix deposition, tissue organization, injury, and tissue remodeling. This review provides a historical perspective of tendon and ligament bioreactors and their contributions to this advancing field. First, we demonstrate the need for bioreactors to improve understanding of tendon and ligament function and dysfunction. Next, we detail the history and evolution of bioreactor development and design from simple stretching of explants to fabrication and stimulation of two- and three-dimensional constructs. Then, we demonstrate how research using tendon and ligament bioreactors has led to pivotal basic science and tissue-engineering discoveries. Finally, we provide guidance for new basic, applied, and clinical research utilizing these valuable systems, recognizing that fundamental knowledge of cell-cell and cell-matrix interactions combined with appropriate mechanical and chemical stimulation of constructs could ultimately lead to functional tendon and ligament repairs in the coming decades.
Collapse
Affiliation(s)
- Nathaniel A. Dyment
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, PA
| | - Jennifer G. Barrett
- Department of Large Animal Clinical Sciences, Marion duPont Scott Equine Medical Center, Virginia Tech, Leesburg, VA
| | - Hani Awad
- Department of Biomedical Engineering, The Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14627
| | | | - Albert Banes
- Flexcell International Corp., 2730 Tucker St., Suite 200, Burlington, 27215, NC
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC
| | - David L. Butler
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221
| |
Collapse
|
32
|
Wang HN, Huang YC, Ni GX. Mechanotransduction of stem cells for tendon repair. World J Stem Cells 2020; 12:952-965. [PMID: 33033557 PMCID: PMC7524696 DOI: 10.4252/wjsc.v12.i9.952] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/06/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tendon is a mechanosensitive tissue that transmits force from muscle to bone. Physiological loading contributes to maintaining the homeostasis and adaptation of tendon, but aberrant loading may lead to injury or failed repair. It is shown that stem cells respond to mechanical loading and play an essential role in both acute and chronic injuries, as well as in tendon repair. In the process of mechanotransduction, mechanical loading is detected by mechanosensors that regulate cell differentiation and proliferation via several signaling pathways. In order to better understand the stem-cell response to mechanical stimulation and the potential mechanism of the tendon repair process, in this review, we summarize the source and role of endogenous and exogenous stem cells active in tendon repair, describe the mechanical response of stem cells, and finally, highlight the mechanotransduction process and underlying signaling pathways.
Collapse
Affiliation(s)
- Hao-Nan Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Yong-Can Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Guo-Xin Ni
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China.
| |
Collapse
|
33
|
Pei D, Wang M, Li W, Li M, Liu Q, Ding R, Zhao J, Li A, Li J, Xu F, Jin G. Remodeling of aligned fibrous extracellular matrix by encapsulated cells under mechanical stretching. Acta Biomater 2020; 112:202-212. [PMID: 32470526 DOI: 10.1016/j.actbio.2020.05.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022]
Abstract
Extracellular matrix (ECM) remodeling is essential for the development and functions of connective tissues (e.g., heart, muscle and the periodontal ligament), and entails the highly anisotropic response of cells and their organized ECM molecules to mechanical stimulation. However, the nature of how cells remodel their surrounding ECM under mechanical stimulation remains elusive. Here, we encapsulated human periodontal ligament stem cells (hPDLSCs) within an aligned rat collagen scaffold labeled with fluorescein isothiocyanate (FITC) and applied mechanical stimulation on the scaffold using magnetic stretching. Through tracking the FITC-labeled rat collagen scaffold and the newly secreted human type I collagen, we studied the effect of magnetic stretching on the mechanism of aligned ECM remodeling by the encapsulated cells. We found that the aligned topography combined with magnetic stretching could significantly promote initial ECM degradation and new ECM secretion: expression of matrix metalloproteinases 1 and 9 is increased markedly, and the elastic modulus of the stretched scaffold (75 kPa) is significantly higher than that of the random scaffold (50 kPa). The data support a model whereby the cells remodel their surrounding ECM under continuous stretching through degradation and then secretion of new ECM to integrate with the aligned ECM and maintain tissue function. Our study offers a valuable basis for future optimized design of biomaterial scaffolds for clinical translation. STATEMENT OF SIGNIFICANCE: Extracellular matrix (ECM) remodeling is essential for the development and functions of connective tissues. However, the nature of how cells remodel their surrounding aligned ECM under mechanical stimulation remains elusive. Herein, we developed a method to reveal the remodeling of aligned rat collagen scaffold by the encapsulated human periodontal ligament stem cells (hPDLSCs) using fluorescence imaging. We found that the aligned topography combined with magnetic stretching could significantly promote initial ECM degradation and new ECM secretion: the expression of matrix metalloproteinase 1 and 9 are significantly higher, and the elastic modulus increases from 50 kPa to 75 kPa as compared to the random collagen scaffold encapsulating hPDLSCs. Our study holds great potential in optimization of bio-scaffold design for clinical translation.
Collapse
|
34
|
No YJ, Castilho M, Ramaswamy Y, Zreiqat H. Role of Biomaterials and Controlled Architecture on Tendon/Ligament Repair and Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904511. [PMID: 31814177 DOI: 10.1002/adma.201904511] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 11/10/2019] [Indexed: 06/10/2023]
Abstract
Engineering synthetic scaffolds to repair and regenerate ruptured native tendon and ligament (T/L) tissues is a significant engineering challenge due to the need to satisfy both the unique biological and biomechanical properties of these tissues. Long-term clinical outcomes of synthetic scaffolds relying solely on high uniaxial tensile strength are poor with high rates of implant rupture and synovitis. Ideal biomaterials for T/L repair and regeneration need to possess the appropriate biological and biomechanical properties necessary for the successful repair and regeneration of ruptured tendon and ligament tissues.
Collapse
Affiliation(s)
- Young Jung No
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Yogambha Ramaswamy
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Training Centre for Innovative BioEngineering, Sydney, NSW, 2006, Australia
- Radcliffe Institute for Advanced Study, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
35
|
Wang D, Pun CCM, Huang S, Tang TCM, Ho KKW, Rothrauff BB, Yung PSH, Blocki AM, Ker EDF, Tuan RS. Tendon-derived extracellular matrix induces mesenchymal stem cell tenogenesis via an integrin/transforming growth factor-β crosstalk-mediated mechanism. FASEB J 2020; 34:8172-8186. [PMID: 32301551 DOI: 10.1096/fj.201902377rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/26/2022]
Abstract
Treatment of tendon injuries is challenging. To develop means to augment tendon regeneration, we have previously prepared a soluble, low immunogenic (DNA-free), tendon extracellular matrix fraction (tECM) by urea extraction of juvenile bovine tendons, which is capable of enhancing transforming growth factor-β (TGF-β) mediated tenogenesis in human adipose-derived stem cells (hASCs). Here, we aimed to elucidate the mechanism of tECM-driven hASC tenogenic differentiation in vitro, focusing on the integrin and TGF-β/SMAD pathways. Our results showed that tECM promoted hASC proliferation and tenogenic differentiation in vitro based on tenogenesis-associated markers. tECM also induced higher expression of several integrin subunits and TGF-β receptors, and nuclear translocation of p-SMAD2 in hASCs. Pharmacological inhibition of integrin-ECM binding, focal adhesion kinase (FAK) signaling, or TGF-β signaling independently led to compromised pro-tenogenic effects of tECM and actin fiber polymerization. Additionally, integrin blockade inhibited tECM-driven TGFBR2 expression, while inhibiting TGF-β signaling decreased tECM-mediated expression of integrin α1, α2, and β1 in hASCs. Together, these findings suggest that the strong pro-tenogenic bioactivity of tECM is regulated via integrin/TGF-β signaling crosstalk. Understanding how integrins interact with signaling by TGF-β and/or other growth factors (GFs) within the tendon ECM microenvironment will provide a rational basis for an ECM-based approach for tendon repair.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Charmaine C M Pun
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Thomas C M Tang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kevin K W Ho
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Benjamin B Rothrauff
- Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick S H Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Anna M Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Elmer D F Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
36
|
Tendon and ligament mechanical loading in the pathogenesis of inflammatory arthritis. Nat Rev Rheumatol 2020; 16:193-207. [PMID: 32080619 DOI: 10.1038/s41584-019-0364-x] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Mechanical loading is an important factor in musculoskeletal health and disease. Tendons and ligaments require physiological levels of mechanical loading to develop and maintain their tissue architecture, a process that is achieved at the cellular level through mechanotransduction-mediated fine tuning of the extracellular matrix by tendon and ligament stromal cells. Pathological levels of force represent a biological (mechanical) stress that elicits an immune system-mediated tissue repair pathway in tendons and ligaments. The biomechanics and mechanobiology of tendons and ligaments form the basis for understanding how such tissues sense and respond to mechanical force, and the anatomical extent of several mechanical stress-related disorders in tendons and ligaments overlaps with that of chronic inflammatory arthritis in joints. The role of mechanical stress in 'overuse' injuries, such as tendinopathy, has long been known, but mechanical stress is now also emerging as a possible trigger for some forms of chronic inflammatory arthritis, including spondyloarthritis and rheumatoid arthritis. Thus, seemingly diverse diseases of the musculoskeletal system might have similar mechanisms of immunopathogenesis owing to conserved responses to mechanical stress.
Collapse
|
37
|
In Vivo and In Vitro Mechanical Loading of Mouse Achilles Tendons and Tenocytes-A Pilot Study. Int J Mol Sci 2020; 21:ijms21041313. [PMID: 32075290 PMCID: PMC7072865 DOI: 10.3390/ijms21041313] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
Mechanical force is a key factor for the maintenance, adaptation, and function of tendons. Investigating the impact of mechanical loading in tenocytes and tendons might provide important information on in vivo tendon mechanobiology. Therefore, the study aimed at understanding if an in vitro loading set up of tenocytes leads to similar regulations of cell shape and gene expression, as loading of the Achilles tendon in an in vivo mouse model. In vivo: The left tibiae of mice (n = 12) were subject to axial cyclic compressive loading for 3 weeks, and the Achilles tendons were harvested. The right tibiae served as the internal non-loaded control. In vitro: tenocytes were isolated from mice Achilles tendons and were loaded for 4 h or 5 days (n = 6 per group) based on the in vivo protocol. Histology showed significant differences in the cell shape between in vivo and in vitro loading. On the molecular level, quantitative real-time PCR revealed significant differences in the gene expression of collagen type I and III and of the matrix metalloproteinases (MMP). Tendon-associated markers showed a similar expression profile. This study showed that the gene expression of tendon markers was similar, whereas significant changes in the expression of extracellular matrix (ECM) related genes were detected between in vivo and in vitro loading. This first pilot study is important for understanding to which extent in vitro stimulation set-ups of tenocytes can mimic in vivo characteristics.
Collapse
|
38
|
Matos AM, Gonçalves AI, El Haj AJ, Gomes ME. Magnetic biomaterials and nano-instructive tools as mediators of tendon mechanotransduction. NANOSCALE ADVANCES 2020; 2:140-148. [PMID: 36133967 PMCID: PMC9417540 DOI: 10.1039/c9na00615j] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/29/2019] [Indexed: 05/29/2023]
Abstract
Tendon tissues connect muscle to bone allowing the transmission of forces resulting in joint movement. Tendon injuries are prevalent in society and the impact on public health is of utmost concern. Thus, clinical options for tendon treatments are in demand, and tissue engineering aims to provide reliable and successful long-term regenerative solutions. Moreover, the possibility of regulating cell fate by triggering intracellular pathways is a current challenge in regenerative medicine. In the last decade, the use of magnetic nanoparticles as nano-instructive tools has led to great advances in diagnostics and therapeutics. Recent advances using magnetic nanomaterials for regenerative medicine applications include the incorporation of magnetic biomaterials within 3D scaffolds resulting in mechanoresponsive systems with unprecedented properties and the use of nanomagnetic actuators to control cell signaling. Mechano-responsive scaffolds and nanomagnetic systems can act as mechanostimulation platforms to apply forces directly to single cells and multicellular biological tissues. As transmitters of forces in a localized manner, the approaches enable the downstream activation of key tenogenic signaling pathways. In this minireview, we provide a brief outlook on the tenogenic signaling pathways which are most associated with the conversion of mechanical input into biochemical signals, the novel bio-magnetic approaches which can activate these pathways, and the efforts to translate magnetic biomaterials into regenerative platforms for tendon repair.
Collapse
Affiliation(s)
- Ana M Matos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Alicia J El Haj
- Healthcare Technologies Institute, Birmingham University B15 2TT Birmingham UK
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at the University of Minho Avepark, 4805-017 Barco Guimarães Portugal
| |
Collapse
|
39
|
Evrova O, Kellenberger D, Calcagni M, Vogel V, Buschmann J. Supporting Cell-Based Tendon Therapy: Effect of PDGF-BB and Ascorbic Acid on Rabbit Achilles Tenocytes in Vitro. Int J Mol Sci 2020; 21:ijms21020458. [PMID: 31936891 PMCID: PMC7014238 DOI: 10.3390/ijms21020458] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/21/2022] Open
Abstract
Cell-based tendon therapies with tenocytes as a cell source need effective tenocyte in vitro expansion before application for tendinopathies and tendon injuries. Supplementation of tenocyte culture with biomolecules that can boost proliferation and matrix synthesis is one viable option for supporting cell expansion. In this in vitro study, the impacts of ascorbic acid or PDGF-BB supplementation on rabbit Achilles tenocyte culture were studied. Namely, cell proliferation, changes in gene expression of several ECM and tendon markers (collagen I, collagen III, fibronectin, aggrecan, biglycan, decorin, ki67, tenascin-C, tenomodulin, Mohawk, α-SMA, MMP-2, MMP-9, TIMP1, and TIMP2) and ECM deposition (collagen I and fibronectin) were assessed. Ascorbic acid and PDGF-BB enhanced tenocyte proliferation, while ascorbic acid significantly accelerated the deposition of collagen I. Both biomolecules led to different changes in the gene expression profile of the cultured tenocytes, where upregulation of collagen I, Mohawk, decorin, MMP-2, and TIMP-2 was observed with ascorbic acid, while these markers were downregulated by PDGF-BB supplementation. Vice versa, there was an upregulation of fibronectin, biglycan and tenascin-C by PDGF-BB supplementation, while ascorbic acid led to a downregulation of these markers. However, both biomolecules are promising candidates for improving and accelerating the in vitro expansion of tenocytes, which is vital for various tendon tissue engineering approaches or cell-based tendon therapy.
Collapse
Affiliation(s)
- Olivera Evrova
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (O.E.); (M.C.)
- Laboratory of Applied Mechanobiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland; (D.K.); (V.V.)
| | - Damian Kellenberger
- Laboratory of Applied Mechanobiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland; (D.K.); (V.V.)
| | - Maurizio Calcagni
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (O.E.); (M.C.)
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland; (D.K.); (V.V.)
| | - Johanna Buschmann
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (O.E.); (M.C.)
- Correspondence: ; Tel.: +41-44-255-9895
| |
Collapse
|
40
|
Lin S, Li J, Dong L, Cheng K, Lin J, Weng W. Periodic-Mechanical-Stimulus Enhanced Osteogenic Differentiation of Mesenchymal Stem Cells on Fe 3O 4/Mineralized Collagen Coatings. ACS Biomater Sci Eng 2019; 5:6446-6453. [PMID: 33417797 DOI: 10.1021/acsbiomaterials.9b00833] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mechanical stimulus has been demonstrated to be critical to stem cell fate commitment and tissue repair. However, it still remains a challenge to remote control of the mechanical stimulus acting on cells. Here, we designed a magnetic Fe3O4/mineralized collagen coating on titanium substrate to regulate the osteogenic differentiation of mesenchymal stem cells (MSCs). The mode and intensity of the mechanical stimulus acting on cells could be controlled by adjusting the remote applied magnetic field. We demonstrated that the adhesion, proliferation, and differentiation of MSCs were strongly dependent on the mode and intensity of the mechanical stimuli. Strikingly, the periodic mechanical stimulus (12 h every other day, PMS) showed the significantly up-regulated expression of osteogenesis-related markers, ALP, compared to that of the static mechanical stimulus mode. The reason is proposed as (1) initially, PMS mode enables the coatings to have appropriate surface mechanical properties for promoting focal adhesion, integrin expression, and cytoskeleton development of MSCs, letting MSCs have good capability of accepting as well as transferring mechanical stimuli; (2) during MSCs growth, PMS mode may effectively manipulate MSCs cytoskeleton development and movement, and mechanotranduction mechanism could be well activated; thus, MSCs osteogenic differentiation is enhanced. This work therefore provides a novel strategy to engineer bioactive coatings with remote control over the intensity and mode of the mechanical stimulus acting on cells, and would have an impact on the design of smart biomaterial surfaces for orthopedic applications.
Collapse
Affiliation(s)
- Suya Lin
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | | | | | - Kui Cheng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | | | - Wenjian Weng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
41
|
Janjic Rankovic M, Docheva D, Wichelhaus A, Baumert U. Effect of static compressive force on in vitro cultured PDL fibroblasts: monitoring of viability and gene expression over 6 days. Clin Oral Investig 2019; 24:2497-2511. [PMID: 31728735 DOI: 10.1007/s00784-019-03113-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The aim was to investigate the impact of static compressive force (CF) application on human PDL-derived fibroblasts (HPDF) in vitro for up to 6 days on the expression of specific genes and to monitor cell growth and cell viability. MATERIALS AND METHODS CF of 2 g/cm2 was applied on HPDFs for 1-6 days. On each day, gene expression (cFOS, HB-GAM, COX2, IL6, TNFα, RUNX2, and P2RX2) and secretion (TNFα, PGE2) were determined by RT-qPCR and ELISA, respectively. Cell growth and cell viability were monitored daily. RESULTS In comparison with controls, significant upregulation of cFOS in compressed HPDFs was observed. HB-GAM showed no changes in expression, except on day 5 (P < 0.001). IL6 expression was significantly upregulated from day 2-5, reaching the maximum on day 3 (P < 0.001). TNFα expression was upregulated on all but day 2. COX2 showed upregulation, reaching the plateau from day 3 (P < 0.001) until day 4 (P < 0.001), and returning to the initial state till day 6. P2RX7 was downregulated on days 2 and 4 to 6 (P < 0.001). RUNX2 was downregulated on days 2 and 5 (both P < 0.001). Cells in both groups were proliferating, and no negative effect on cell viability was observed. CONCLUSION Results suggest high molecular activity up to 6 days, therefore introducing further need for in vitro studies with a longer duration that would explain other genes and metabolites involved in orthodontic tooth movement (OTM). CLINICAL RELEVANCE Extension of an established in vitro force application system for prolonged force application (6 days) simulating the initial phase of OTM.
Collapse
Affiliation(s)
- Mila Janjic Rankovic
- Department of Orthodontics and Dentofacial Orthopedics, University Hospital, LMU Munich, Goethestrasse 70, 80336, Munich, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Andrea Wichelhaus
- Department of Orthodontics and Dentofacial Orthopedics, University Hospital, LMU Munich, Goethestrasse 70, 80336, Munich, Germany
| | - Uwe Baumert
- Department of Orthodontics and Dentofacial Orthopedics, University Hospital, LMU Munich, Goethestrasse 70, 80336, Munich, Germany.
| |
Collapse
|
42
|
Wan W, He C, Du C, Wang Y, Wu S, Wang T, Zou R. Effect of ILK on small-molecule metabolism of human periodontal ligament fibroblasts with mechanical stretching. J Periodontal Res 2019; 55:229-237. [PMID: 31630411 DOI: 10.1111/jre.12706] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 09/16/2019] [Accepted: 09/22/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Mechanical stimuli can cause periodontal tissue reconstruction. Studies have found that changes in metabolites can be the terminal effect of integrin-mediated mechanical signaling. As a key kinase in integrin regulation, integrin-linked kinase (ILK) mediates mechanical signal transduction, which may contribute to metabolite changes. Defining the components of small-molecule metabolites can optimize mechanical stimuli and periodontal tissue reconstruction. Our purpose is to detect the effect of ILK-mediated mechanical signaling on intracellular small-molecule metabolites (amino acids and organic acids) in human periodontal ligament fibroblasts (HPDLFs). METHODS Primary HPDLFs were isolated by enzyme digestion method. Tensile stresses were applied on HPDLFs in vitro using a Flexcell system. ILK gene in HPDLFs was knocked down by RNA interference (RNAi). Twenty common amino acids and seven organic acids in HPDLFs were analyzed by gas chromatography/mass spectrometry technique. RESULTS Five amino acids (ie, alanine, glutamine, glutamate, glycine, and threonine) and three organic acids (ie, pyruvate, lactate, and citric acid) were found to be changed remarkably after mechanical stretching. In addition, baseline levels of four amino acids (ie, glutamate, glutamine, threonine, and glycine) and two organic acids (ie, lactate and citric acid) were significantly different in ILK knockdown compared with wild-type HPDLFs. CONCLUSION This study suggests that five amino acids (ie, alanine, glutamine, glutamate, glycine, and threonine) and three organic acids (ie, pyruvate, lactate, and citric acid) may act as cellular mediators for mechanical signals in HPDLFs. Among them, four amino acids (ie, glutamate, glutamine, threonine, and glycine) and two organic acids (ie, lactate and citric acid) may be closely linked to ILK.
Collapse
Affiliation(s)
- Wanting Wan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Chuan He
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | | | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Shiyang Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Tairan Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
43
|
Zhang C, Zhu J, Zhou Y, Thampatty BP, Wang JHC. Tendon Stem/Progenitor Cells and Their Interactions with Extracellular Matrix and Mechanical Loading. Stem Cells Int 2019; 2019:3674647. [PMID: 31737075 PMCID: PMC6815631 DOI: 10.1155/2019/3674647] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/04/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
Tendons are unique connective tissues in the sense that their biological properties are largely determined by their tendon-specific stem cells, extracellular matrix (ECM) surrounding the stem cells, mechanical loading conditions placed on the tendon, and the complex interactions among them. This review is aimed at providing an overview of recent advances in the identification and characterization of tendon stem/progenitor cells (TSPCs) and their interactions with ECM and mechanical loading. In addition, the effects of such interactions on the maintenance of tendon homeostasis and the initiation of tendon pathological conditions are discussed. Moreover, the challenges in further investigations of TSPC mechanobiology in vitro and in vivo are outlined. Finally, future research efforts are suggested, which include using specific gene knockout models and single-cell transcription profiling to enable a broad and deep understanding of the physiology and pathophysiology of tendons.
Collapse
Affiliation(s)
- Chuanxin Zhang
- Joint Surgery and Sports Medicine Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jun Zhu
- Joint Surgery and Sports Medicine Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yiqin Zhou
- Joint Surgery and Sports Medicine Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bhavani P. Thampatty
- MechanoBiology Laboratory, Departments of Orthopaedic Surgery, Bioengineering, and Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James H-C. Wang
- MechanoBiology Laboratory, Departments of Orthopaedic Surgery, Bioengineering, and Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
44
|
Walters M, Crew M, Fyfe G. Bone Surface Micro‐Topography at Craniofacial Entheses: Insights on Osteogenic Adaptation at Muscle Insertions. Anat Rec (Hoboken) 2019; 302:2140-2155. [DOI: 10.1002/ar.24215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 02/04/2019] [Accepted: 03/06/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Mark Walters
- School of Human SciencesThe University of Western Australia Crawley Perth Western Australia
- Department of Plastic and Reconstructive SurgeryPerth Children's Hospital Nedlands Perth Western Australia
| | - Michael Crew
- Health Department of Western Australia and Faculty of Health SciencesCurtin University Western Australia
| | - Georgina Fyfe
- Faculty of Health SciencesCurtin University Perth Western Australia
| |
Collapse
|
45
|
Walia B, Huang AH. Tendon stem progenitor cells: Understanding the biology to inform therapeutic strategies for tendon repair. J Orthop Res 2019; 37:1270-1280. [PMID: 30270569 PMCID: PMC6823601 DOI: 10.1002/jor.24156] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
Tendon and ligament injuries are a leading cause of healthcare visits with significant impact in terms of economic cost and reduced quality of life. To date, reparative strategies remain largely restricted to conservative treatment or surgical repair. However, these therapies fail to restore native tendon structure and function; thus, the tissue may re-rupture or degenerate with time. To improve tendon healing, one promising strategy may be harnessing the innate potential of resident tendon stem/progenitor cells (TSPCs) to guide tenogenic regeneration. In this review, we outline recent advances in the identification and characterization of putative TSPC populations, and discuss biochemical, biomechanical, and biomaterial methods employed for their culture and differentiation. Finally, we identify limitations in our current understanding of TSPC biology, key challenges for their use, and potential therapeutic strategies to inform cell-based tendon repair. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1270-1280, 2019.
Collapse
Affiliation(s)
- Bhavita Walia
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice H. Huang
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
46
|
Subramanian A, Kanzaki LF, Galloway JL, Schilling TF. Mechanical force regulates tendon extracellular matrix organization and tenocyte morphogenesis through TGFbeta signaling. eLife 2018; 7:e38069. [PMID: 30475205 PMCID: PMC6345564 DOI: 10.7554/elife.38069] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/26/2018] [Indexed: 12/28/2022] Open
Abstract
Mechanical forces between cells and extracellular matrix (ECM) influence cell shape and function. Tendons are ECM-rich tissues connecting muscles with bones that bear extreme tensional force. Analysis of transgenic zebrafish expressing mCherry driven by the tendon determinant scleraxis reveals that tendon fibroblasts (tenocytes) extend arrays of microtubule-rich projections at the onset of muscle contraction. In the trunk, these form a dense curtain along the myotendinous junctions at somite boundaries, perpendicular to myofibers, suggesting a role as force sensors to control ECM production and tendon strength. Paralysis or destabilization of microtubules reduces projection length and surrounding ECM, both of which are rescued by muscle stimulation. Paralysis also reduces SMAD3 phosphorylation in tenocytes and chemical inhibition of TGFβ signaling shortens tenocyte projections. These results suggest that TGFβ, released in response to force, acts on tenocytes to alter their morphology and ECM production, revealing a feedback mechanism by which tendons adapt to tension.
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineUnited States
| | - Lauren Fallon Kanzaki
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineUnited States
| | - Jenna Lauren Galloway
- Center for Regenerative Medicine, Department of Orthopaedic SurgeryMassachusetts General Hospital, Harvard Stem Cell InstituteBostonUnited States
| | | |
Collapse
|
47
|
Dursun G, Tohidnezhad M, Markert B, Stoffel M. Effects of uniaxial stretching on tenocyte migration behaviour. CURRENT DIRECTIONS IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1515/cdbme-2018-0076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractIt is widely known that tendon tissues are subjected to repeated cyclic mechanical load which influences cellular processes. The involvement of principles of mechanics in tissue engineering contributes to the investigations of the connection between mechanical and biological parameters in cellular processes and as well as to the development of new approaches for specific treatment methods. The healing process of injured tendons includes tenocyte migration which occurs from intact regions of tendon into the wound site. The aim of the present study is to investigate and enhance the migration characteristics of tenocytes under uniaxial mechanical stretching using an in-house tensile bioreactor system. Uniaxial mechanical stretching is applied to tenocyte-seeded silicone as well as collagen membranes, which possess different material properties. Tenocyte-seeded silicone membranes were investigated under three different loading conditions, including unstimulated (control), 3% and 5% strain, at frequency of 0.5 Hz. Tenocyte-seeded collagen membranes were investigated using three different frequencies, including unstimulated (control), 0.1 Hz and 0.5 Hz at strain of 4%. The main finding in this study is that uniaxially mechanical stretching at 3% strain enhances the cell migration more than 5% strain on silicone membranes.
Collapse
Affiliation(s)
- Gözde Dursun
- 1Institute of General Mechanics, RWTH Aachen University,Aachen, Germany
| | | | - Bernd Markert
- 1Institute of General Mechanics, RWTH Aachen University,Aachen, Germany
| | - Marcus Stoffel
- 1Institute of General Mechanics, RWTH Aachen University,Aachen, Germany
| |
Collapse
|
48
|
In Vitro Comparison of 2D-Cell Culture and 3D-Cell Sheets of Scleraxis-Programmed Bone Marrow Derived Mesenchymal Stem Cells to Primary Tendon Stem/Progenitor Cells for Tendon Repair. Int J Mol Sci 2018; 19:ijms19082272. [PMID: 30072668 PMCID: PMC6121892 DOI: 10.3390/ijms19082272] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/23/2018] [Accepted: 07/27/2018] [Indexed: 01/26/2023] Open
Abstract
The poor and slow healing capacity of tendons requires novel strategies to speed up the tendon repair process. Hence, new and promising developments in tendon tissue engineering have become increasingly relevant. Previously, we have established a tendon progenitor cell line via ectopic expression of the tendon-related basic helix-loop-helix (bHLH) transcription factor Scleraxis (Scx) in human bone marrow mesenchymal stem cells (hMSC-Scx). The aim of this study was to directly compare the characteristics of hMSC-Scx cells to that of primary human tendon stem/progenitors cells (hTSPCs) via assessment of self-renewal and multipotency, gene marker expression profiling, in vitro wound healing assay and three-dimensional cell sheet formation. As expected, hTSPCs were more naive than hMSC-Scx cells because of higher clonogenicity, trilineage differentiation potential, and expression of stem cell markers, as well as higher mRNA levels of several gene factors associated with early tendon development. Interestingly, with regards to wound healing, both cell types demonstrate a comparable speed of scratch closure, as well as migratory velocity and distance in various migration experiments. In the three-dimensional cell sheet model, hMSC-Scx cells and hTSPCs form compact tendinous sheets as histological staining, and transmission electron microscopy shows spindle-shaped cells and collagen type I fibrils with similar average diameter size and distribution. Taken together, hTSPCs exceed hMSC-Scx cells in several characteristics, namely clonogenicity, multipotentiality, gene expression profile and rates of tendon-like sheet formation, whilst in three-dimensional cell sheets, both cell types have comparable in vitro healing potential and collagenous composition of their three-dimensional cell sheets, making both cell types a suitable cell source for tendon tissue engineering and healing.
Collapse
|
49
|
Bell R, Robles-Harris M, Anderson M, Laudier D, Schaffler M, Flatow E, Andarawis-Puri N. Inhibition of apoptosis exacerbates fatigue-damage tendon injuries in an in vivo rat model. Eur Cell Mater 2018; 36:44-56. [PMID: 30058060 PMCID: PMC6350530 DOI: 10.22203/ecm.v036a04] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Tendinopathy is a common and progressive musculoskeletal disease. Increased apoptosis is an end-stage tendinopathy manifestation, but its contribution to the pathology of the disease is unknown. A previously established in vivo model of fatigue damage accumulation shows that increased apoptosis is correlated with the severity of induced tendon damage, even in early onset of the disease, supporting its implication in the pathogenesis of the disease. Consequently, this study aimed to determine: (1) whether apoptosis could be inhibited after fatigue damage and (2) whether its inhibition could lead to remodeling of the extracellular matrix (ECM) and pericellular matrix (PCM), to ultimately improve the mechanical properties of fatigue-damaged tendons. The working hypothesis was that, despite the low vascular nature of the tendon, apoptosis would be inhibited, prompting increased production of matrix proteins and restoring tendon mechanical properties. Rats received 2 or 5 d of systemic pan-caspase inhibitor (Q-VD-OPh) or dimethyl sulfoxide (DMSO) carrier control injections starting immediately prior to fatigue loading and were sacrificed at days 7 and 14 post-fatigue-loading. Systemic pan-caspase inhibition for 2 d led to a surprising increase in apoptosis, but inhibition for 5 d increased the population of live cells that could repair the fatigue damage. Further analysis of the 5 d group showed that effective inhibition led to an increased population of cells producing ECM and PCM proteins, although typically in conjunction with oxidative stress markers. Ultimately, inhibition of apoptosis led to further deterioration in mechanical properties of fatigue-damaged tendons.
Collapse
Affiliation(s)
- R. Bell
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - M.A. Robles-Harris
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - M. Anderson
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D. Laudier
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M.B. Schaffler
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - E.L. Flatow
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - N. Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Hospital for Special Surgery, New York, NY, USA,Address for correspondence: Nelly Andarawis-Puri, PhD, Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, 14850, NY, USA.
| |
Collapse
|
50
|
Pesqueira T, Costa‐Almeida R, Gomes ME. Magnetotherapy: The quest for tendon regeneration. J Cell Physiol 2018; 233:6395-6405. [DOI: 10.1002/jcp.26637] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Tamagno Pesqueira
- 3B's Research Group − Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Zona Industrial da Gandra Barco Guimarães Portugal
- ICVS/3B's − PT Government Associate Laboratory Guimarães Portugal
| | - Raquel Costa‐Almeida
- 3B's Research Group − Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Zona Industrial da Gandra Barco Guimarães Portugal
- ICVS/3B's − PT Government Associate Laboratory Guimarães Portugal
| | - Manuela E. Gomes
- 3B's Research Group − Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Zona Industrial da Gandra Barco Guimarães Portugal
- ICVS/3B's − PT Government Associate Laboratory Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision Medicine Headquarters at University of Minho Barco Guimarães Portugal
| |
Collapse
|