1
|
Muscat S, Nichols AEC. Leveraging in vivo animal models of tendon loading to inform tissue engineering approaches. Front Bioeng Biotechnol 2024; 12:1449372. [PMID: 39434716 PMCID: PMC11491380 DOI: 10.3389/fbioe.2024.1449372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Tendon injuries disrupt successful transmission of force between muscle and bone, resulting in reduced mobility, increased pain, and significantly reduced quality of life for affected patients. There are currently no targeted treatments to improve tendon healing beyond conservative methods such as rest and physical therapy. Tissue engineering approaches hold great promise for designing instructive biomaterials that could improve tendon healing or for generating replacement graft tissue. More recently, engineered microphysiological systems to model tendon injuries have been used to identify therapeutic targets. Despite these advances, current tissue engineering efforts that aim to regenerate, replace, or model injured tendons have largely failed due in large part to a lack of understanding of how the mechanical environment of the tendon influences tissue homeostasis and how altered mechanical loading can promote or prevent disease progression. This review article draws inspiration from what is known about tendon loading from in vivo animal models and identifies key metrics that can be used to benchmark success in tissue engineering applications. Finally, we highlight important challenges and opportunities for the field of tendon tissue engineering that should be taken into consideration in designing engineered platforms to understand or improve tendon healing.
Collapse
Affiliation(s)
- Samantha Muscat
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
- Department of Orthopedics and Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Anne E. C. Nichols
- Department of Orthopedics and Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
2
|
Yue S, Zhai G, Zhao S, Liang X, Liu Y, Zheng J, Chen X, Dong Y. The biphasic role of the infrapatellar fat pad in osteoarthritis. Biomed Pharmacother 2024; 179:117364. [PMID: 39226725 DOI: 10.1016/j.biopha.2024.117364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative disease resulting in joint deterioration. It is a whole organ disease characterized by cartilage degeneration and varying degrees of synovitis, involving pathological changes in all joint tissues, such as cartilage, subchondral bone, ligaments, meniscus, synovium, and infrapatellar fat pad (IPFP). IPFP is the largest adipose tissue structure in the knee joint and is composed of fat cells, immune cells and blood vessels. Moreover, IPFP is located close to the cartilage and bone surface so that it may reduce the impact of loading and absorb forces generated through the knee joint, and may have a protective role in joint health. IPFP has been shown to release various cytokines and adipokines that play pro-inflammatory and pro-catabolic roles in cartilage, promoting OA progression. Intra-articular injections of IPFP-derived mesenchymal stem cells and exosomes have been shown to reduce pain and prevent OA progression in patients with knee OA. Previous studies have shown that IPFP has a biphasic effect on OA progression. This article reviews the latest research progress of IPFP, discusses the role and mechanism of IPFP in OA, provide new intervention strategies for the treatment of OA. This article will also discuss the handling of IPFP during the procedure of total knee arthroplasty.
Collapse
Affiliation(s)
- Songkai Yue
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Ganggang Zhai
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Siyu Zhao
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Xiaming Liang
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Yunke Liu
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Xiaoyang Chen
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Yonghui Dong
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China.
| |
Collapse
|
3
|
Shen S, Lin Y, Sun J, Liu Y, Chen Y, Lu J. A New Tissue Engineering Strategy to Promote Tendon-bone Healing: Regulation of Osteogenic and Chondrogenic Differentiation of Tendon-derived Stem Cells. Orthop Surg 2024; 16:2311-2325. [PMID: 39043618 PMCID: PMC11456719 DOI: 10.1111/os.14152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
In the field of sports medicine, repair surgery for anterior cruciate ligament (ACL) and rotator cuff (RC) injuries are remarkably common. Despite the availability of relatively effective treatment modalities, outcomes often fall short of expectations. This comprehensive review aims to thoroughly examine current strategies employed to promote tendon-bone healing and analyze pertinent preclinical and clinical research. Amidst ongoing investigations, tendon-derived stem cells (TDSCs), which have comparatively limited prior exploration, have garnered increasing attention in the context of tendon-bone healing, emerging as a promising cell type for regenerative therapies. This review article delves into the potential of combining TDSCs with tissue engineering methods, with ACL reconstruction as the main focus. It comprehensively reviews relevant research on ACL and RC healing to address the issues of graft healing and bone tunnel integration. To optimize tendon-bone healing outcomes, our emphasis lies in not only reconstructing the original microstructure of the tendon-bone interface but also achieving proper bone tunnel integration, encompassing both cartilage and bone formation. In this endeavor, we thoroughly analyze the transcriptional and molecular regulatory variables governing TDSCs differentiation, incorporating a retrospective analysis utilizing single-cell sequencing, with the aim of unearthing relevant signaling pathways and processes. By presenting a novel strategy rooted in TDSCs-driven osteogenic and chondrogenic differentiation for tendon-bone healing, this study paves the way for potential future research avenues and promising therapeutic applications. It is anticipated that the findings herein will contribute to advancing the field of tendon-bone healing and foster the exploration of TDSCs as a viable option for regenerative therapies in the future.
Collapse
Affiliation(s)
- Sinuo Shen
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yucheng Lin
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Jiachen Sun
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yuanhao Liu
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yuzhi Chen
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Jun Lu
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
4
|
Timmer KB, Killian ML, Harley BAC. Paracrine signals influence patterns of fibrocartilage differentiation in a lyophilized gelatin hydrogel for applications in rotator cuff repair. Biomater Sci 2024; 12:4806-4822. [PMID: 39150417 PMCID: PMC11404831 DOI: 10.1039/d4bm00543k] [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] [Indexed: 08/17/2024]
Abstract
Rotator cuff injuries present a clinical challenge for repair due to current limitations in functional regeneration of the native tendon-to-bone enthesis. A biomaterial that can regionally instruct unique tissue-specific phenotypes offers potential to promote enthesis repair. We have recently demonstrated the mechanical benefits of a stratified triphasic biomaterial made up of tendon- and bone-mimetic collagen scaffold compartments connected via a continuous hydrogel, and we now explore the potential of a biologically favorable enthesis hydrogel for this application. Here we report in vitro behavior of human mesenchymal stem cells (hMSCs) within thiolated gelatin (Gel-SH) hydrogels in response to chondrogenic stimuli as well as paracrine signals derived from MSC-seeded bone and tendon scaffold compartments. Chondrogenic differentiation media promoted upregulation of cartilage and entheseal fibrocartilage matrix markers COL2, COLX, and ACAN as well as the enthesis-associated transcription factors SCX, SOX9, and RUNX2 in hMSCs within Gel-SH. Similar effects were observed in response to TGF-β3 and BMP-4, enthesis-associated growth factors known to play a role in entheseal development and maintenance. Conditioned media generated by hMSCs seeded in tendon- and bone-mimetic collagen scaffolds influenced patterns of gene expression regarding enthesis-relevant growth factors, matrix markers, and tendon-to-bone transcription factors for hMSCs within the material. Together, these findings demonstrate that a Gel-SH hydrogel provides a permissive environment for enthesis tissue engineering and highlights the significance of cellular crosstalk between adjacent compartments within a spatially graded biomaterial.
Collapse
Affiliation(s)
- Kyle B Timmer
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.
| | - Megan L Killian
- Department of Orthopaedic Surgery, University of Michigan Ann Arbor, Ann Arbor, Michigan 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, Ann Arbor, Michigan 48109, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
5
|
Enayati M, Liu W, Madry H, Neisiany RE, Cucchiarini M. Functionalized hydrogels as smart gene delivery systems to treat musculoskeletal disorders. Adv Colloid Interface Sci 2024; 331:103232. [PMID: 38889626 DOI: 10.1016/j.cis.2024.103232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Despite critical advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy based on the delivery of therapeutic genetic sequences has strong value to offer effective, durable options to decisively manage such disorders. Furthermore, scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy, allowing for the spatiotemporal delivery of candidate genes to sites of injury. Among the many scaffolds for musculoskeletal research, hydrogels raised increasing attention in addition to other potent systems (solid, hybrid scaffolds) due to their versatility and competence as drug and cell carriers in tissue engineering and wound dressing. Attractive functionalities of hydrogels for musculoskeletal therapy include their injectability, stimuli-responsiveness, self-healing, and nanocomposition that may further allow to upgrade of them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. Such functionalized hydrogels may also be tuned to successfully transfer therapeutic genes in a minimally invasive manner in order to protect their cargos and allow for their long-term effects. In light of such features, this review focuses on functionalized hydrogels and demonstrates their competence for the treatment of musculoskeletal disorders using gene therapy procedures, from gene therapy principles to hydrogel functionalization methods and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are being discussed in the perspective of translation in patients. STATEMENT OF SIGNIFICANCE: Despite advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy has strong value in offering effective, durable options to decisively manage such disorders. Scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy. Among many scaffolds for musculoskeletal research, hydrogels raised increasing attention. Functionalities including injectability, stimuli-responsiveness, and self-healing, tune them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. This review introduces functionalized hydrogels for musculoskeletal disorder treatment using gene therapy procedures, from gene therapy principles to functionalized hydrogels and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are discussed from the perspective of translation in patients.
Collapse
Affiliation(s)
- Mohammadsaeid Enayati
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Rasoul Esmaeely Neisiany
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland; Department of Polymer Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany.
| |
Collapse
|
6
|
Emonts C, Bauer B, Pitts J, Roger Y, Hoffmann A, Menzel H, Gries T. Mechanical, Biological and In Vitro Degradation Investigation of Braided Scaffolds for Tendon and Ligament Tissue Engineering Based on Different Polycaprolactone Materials with Chitosan-Graft-PCL Surface Modification. Polymers (Basel) 2024; 16:2349. [PMID: 39204570 PMCID: PMC11360056 DOI: 10.3390/polym16162349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/08/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
Injuries to tendons and ligaments are highly prevalent in the musculoskeletal system. Current treatments involve autologous transplants with limited availability and donor site morbidity. Tissue engineering offers a new approach through temporary load-bearing scaffolds. These scaffolds have to fulfill numerous requirements, the majority of which can be met using braiding combined with high-strength polycaprolactone (PCL) fibers. Considering regulatory requirements, several medical-grade PCL materials were assessed regarding their mechanical, degradational and cell biological properties. In the course of the investigation, an excellent fiber tensile strength of up to 850 MPa was achieved. The fibers were braided into multilayer scaffolds and scaled to match the human ACL. These were characterized regarding their morphology and their mechanical and degradational properties. Two strategies were followed to provide biological cues: (a) applying a chitosan-graft-PCL surface modification and (b) using non-circular fiber morphologies as topographical stimuli. Cell vitality assays showed generally positive cytocompatibility and no impairments due to the surface modification or material grade. The best cell vitality was achieved with a scaffold consisting of snowflake-shaped monofilaments combined with a 25° braiding angle. The surface modification equips the scaffold with a release platform for function molecules (as recently demonstrated) so that a holistic approach to addressing the numerous requirements is provided.
Collapse
Affiliation(s)
- Caroline Emonts
- Institut für Textiltechnik, RWTH Aachen University, 52074 Aachen, Germany
| | - Benedict Bauer
- Institut für Textiltechnik, RWTH Aachen University, 52074 Aachen, Germany
| | - Johannes Pitts
- Institute for Technical Chemistry, Braunschweig University of Technology, 38106 Braunschweig, Germany
| | - Yvonne Roger
- Hannover Medical School, Department of Orthopedic Surgery, Graded Implants and Regenerative Strategies, Laboratory of Biomechanics and Biomaterials, 30625 Hannover, Germany
- Niedersächsisches Zentrum für Biomedizintechnik, Implantatforschung und Entwicklung (NIFE), 30625 Hannover, Germany
| | - Andrea Hoffmann
- Hannover Medical School, Department of Orthopedic Surgery, Graded Implants and Regenerative Strategies, Laboratory of Biomechanics and Biomaterials, 30625 Hannover, Germany
- Niedersächsisches Zentrum für Biomedizintechnik, Implantatforschung und Entwicklung (NIFE), 30625 Hannover, Germany
| | - Henning Menzel
- Institute for Technical Chemistry, Braunschweig University of Technology, 38106 Braunschweig, Germany
| | - Thomas Gries
- Institut für Textiltechnik, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
7
|
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
|
8
|
Adjei-Sowah E, Chandrasiri I, Xiao B, Liu Y, Ackerman JE, Soto C, Nichols AEC, Nolan K, Benoit DSW, Loiselle AE. Development of a nanoparticle-based tendon-targeting drug delivery system to pharmacologically modulate tendon healing. SCIENCE ADVANCES 2024; 10:eadn2332. [PMID: 38896625 PMCID: PMC11186494 DOI: 10.1126/sciadv.adn2332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Satisfactory healing following acute tendon injury is marred by fibrosis. Despite the high frequency of tendon injuries and poor outcomes, there are no pharmacological therapies in use to enhance the healing process. Moreover, systemic treatments demonstrate poor tendon homing, limiting the beneficial effects of potential tendon therapeutics. To address this unmet need, we leveraged our existing tendon healing spatial transcriptomics dataset and identified an area enriched for expression of Acp5 (TRAP) and subsequently demonstrated robust TRAP activity in the healing tendon. This unexpected finding allowed us to refine and apply our existing TRAP binding peptide (TBP) functionalized nanoparticle (NP) drug delivery system (DDS) to facilitate improved delivery of systemic treatments to the healing tendon. To demonstrate the translational potential of this DDS, we delivered niclosamide (NEN), an S100a4 inhibitor. While systemic delivery of free NEN did not alter healing, TBP-NPNEN enhanced both functional and mechanical recovery, demonstrating the translational potential of this approach to enhance the tendon healing process.
Collapse
Affiliation(s)
- Emmanuela Adjei-Sowah
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Indika Chandrasiri
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Baixue Xiao
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yuxuan Liu
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14623, USA
| | - Jessica E. Ackerman
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Celia Soto
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Anne E. C. Nichols
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Orthopaedics and Physical Performance, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Katherine Nolan
- Department of Comparative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14623, USA
- Materials Science Program, University of Rochester, Rochester, NY 14623, USA
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA
| | - Alayna E. Loiselle
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Orthopaedics and Physical Performance, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
9
|
Li J, Ke H, Lei X, Zhang J, Wen Z, Xiao Z, Chen H, Yao J, Wang X, Wei Z, Zhang H, Pan W, Shao Y, Zhao Y, Xie D, Zeng C. Controlled-release hydrogel loaded with magnesium-based nanoflowers synergize immunomodulation and cartilage regeneration in tendon-bone healing. Bioact Mater 2024; 36:62-82. [PMID: 38440323 PMCID: PMC10909705 DOI: 10.1016/j.bioactmat.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Tendon-bone interface injuries pose a significant challenge in tissue regeneration, necessitating innovative approaches. Hydrogels with integrated supportive features and controlled release of therapeutic agents have emerged as promising candidates for the treatment of such injuries. In this study, we aimed to develop a temperature-sensitive composite hydrogel capable of providing sustained release of magnesium ions (Mg2+). We synthesized magnesium-Procyanidin coordinated metal polyphenol nanoparticles (Mg-PC) through a self-assembly process and integrated them into a two-component hydrogel. The hydrogel was composed of dopamine-modified hyaluronic acid (Dop-HA) and F127. To ensure controlled release and mitigate the "burst release" effect of Mg2+, we covalently crosslinked the Mg-PC nanoparticles through coordination bonds with the catechol moiety within the hydrogel. This crosslinking strategy extended the release window of Mg2+ concentrations for up to 56 days. The resulting hydrogel (Mg-PC@Dop-HA/F127) exhibited favorable properties, including injectability, thermosensitivity and shape adaptability, making it suitable for injection and adaptation to irregularly shaped supraspinatus implantation sites. Furthermore, the hydrogel sustained the release of Mg2+ and Procyanidins, which attracted mesenchymal stem and progenitor cells, alleviated inflammation, and promoted macrophage polarization towards the M2 phenotype. Additionally, it enhanced collagen synthesis and mineralization, facilitating the repair of the tendon-bone interface. By incorporating multilevel metal phenolic networks (MPN) to control ion release, these hybridized hydrogels can be customized for various biomedical applications.
Collapse
Affiliation(s)
- Jintao Li
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Haolin Ke
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangcheng Lei
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jiexin Zhang
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhicheng Wen
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhisheng Xiao
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Huabin Chen
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Juncheng Yao
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xuan Wang
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengnong Wei
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Hongrui Zhang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weilun Pan
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yan Shao
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yitao Zhao
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Denghui Xie
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Chun Zeng
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Liu YC, Chen SH, Kuan CH, Chen SH, Huang WY, Chen HX, Wang TW. Assembly of Interfacial Polyelectrolyte Complexation Fibers with Mineralization Gradient for Physiologically-Inspired Ligament Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314294. [PMID: 38572797 DOI: 10.1002/adma.202314294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/29/2024] [Indexed: 04/05/2024]
Abstract
Current synthetic grafts for ligament rupture repair often fail to integrate well with the surrounding biological tissue, leading to complications such as graft wear, fatigue, and subsequent re-rupture. To address this medical challenge, this study aims at advancing the development of a biological ligament through the integration of physiologically-inspired principles and tissue engineering strategies. In this study, interfacial polyelectrolyte complexation (IPC) spinning technique, along with a custom-designed collection system, to fabricate a hierarchical scaffold mimicking native ligament structure, is utilized. To emulate the bone-ligament interface and alleviate stress concentration, a hydroxyapatite (HAp) mineral gradient is strategically introduced near both ends of the scaffold to enhance interface integration and diminish the risk of avulsion rupture. Biomimetic viscoelasticity is successfully displayed to provide similar mechanical support to native ligamentous tissue under physiological conditions. By introducing the connective tissue growth factor (CTGF) and conducting mesenchymal stem cells transplantation, the regenerative potential of the synthetic ligament is significantly amplified. This pioneering study offers a multifaceted solution combining biomimetic materials, regenerative therapies, and advanced techniques to potentially transform ligament rupture treatment.
Collapse
Affiliation(s)
- Yu-Chung Liu
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| | - Shih-Heng Chen
- Division of Trauma Plastic Surgery, Department of Plastic & Reconstructive Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan City, 33305, Taiwan
| | - Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Shih-Hsien Chen
- Division of Trauma Plastic Surgery, Department of Plastic & Reconstructive Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan City, 33305, Taiwan
| | - Wei-Yuan Huang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| | - Hao-Xuan Chen
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| | - Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30044, Taiwan
| |
Collapse
|
11
|
Peniche Silva CJ, Balmayor ER, van Griensven M. Reprogramming tendon healing: a guide to novel molecular tools. Front Bioeng Biotechnol 2024; 12:1379773. [PMID: 38784762 PMCID: PMC11112497 DOI: 10.3389/fbioe.2024.1379773] [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: 01/31/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Tendons are a frequent site of injury, which greatly impairs the movement and locomotion of patients. Regrettably, injuries at the tendon frequently require surgical intervention, which leads to a long path to recovery. Moreover, the healing of tendons often involves the formation of scar tissue at the site of injury with poor mechanical properties and prone to re-injury. Tissue engineering carries the promise of better and more effective solutions to the improper healing of tendons. Lately, the field of regenerative medicine has seen a significant increase in the focus on the potential use of non-coding RNAs (e.g., siRNAs, miRNAs, and lncRNAs) as molecular tools for tendon tissue engineering. This class of molecules is being investigated due to their ability to act as epigenetic regulators of gene expression and protein production. Thus, providing a molecular instrument to fine-tune, reprogram, and modulate the processes of tendon differentiation, healing, and regeneration. This review focuses particularly on the latest advances involving the use of siRNAs, miRNAs, and lncRNAs in tendon tissue engineering applications.
Collapse
Affiliation(s)
- Carlos Julio Peniche Silva
- Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Elizabeth R. Balmayor
- Experimental Orthopaedics and Trauma Surgery, Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Martijn van Griensven
- Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
12
|
Sun Y, Tang Y. The relationship between lateral femoral condyle ratio measured by MRI and anterior cruciate ligament injury. Front Bioeng Biotechnol 2024; 12:1362110. [PMID: 38600950 PMCID: PMC11004325 DOI: 10.3389/fbioe.2024.1362110] [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: 12/27/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Background Previous studies have shown that the lateral femoral condyle ratio (LFCR) measured by X-ray has a significant relationship with the anterior cruciate ligament (ACL) injury. However, few relevant studies have been performed on LFCR measured by magnetic resonance imaging (MRI). Purpose (1) To evaluate the relationship between LFCR measured by MRI and ACL injury or rerupture. (2) To compare the LFCR measured by MRI with existing bony morphological risk factors and screen out the most predictive risk factors for primary ACL injury or rerupture. Study Design Cohort study; Level of evidence, 3. Methods Totally 147 patients who underwent knee arthroscopic surgery from 2015 to 2019 with minimum follow-up of 48 months were retrospectively evaluated. Patients were placed into three groups: 1) the control group of patients with simple meniscus tears without ligament injury; 2) the primary noncontact ACL injury group; 3) ACL rerupture group (ACL reconstruction failure). The LFCR measured by MRI and other previous known risk factors associated with MRI (notch width index, medial tibial slope, lateral tibial slope, medial tibial depth, lateral tibial height) were performed to evaluate their predictive value for ACL injury and rerupture. All the risk factors with p < 0.01 according to univariate analysis were included in the logistic regression models. Receiver operating characteristic (ROC) curves were analyzed for sensitivity, specificity, cut-off, and area under the curve (AUC). Z tests were used to compare the AUC values. Results The LFCR measured by MRI was obviously higher in primary ACL injury group (0.628 ± 0.020) and in ACL rerupture group (0.625 ± 0.021) than that in the control group (0.593 ± 0.030). The best risk factor was the LFCR with a cut-off of 0.602 (AUC, 0.818; 95% CI, 0.748-0.878; sensitivity, 90%; specificity, 66%). When combined with lateral tibial slope (cutoff, 7°) and lateral tibial height (cutoff, 3.6 mm), the diagnostic performance was improved significantly (AUC, 0.896; 95% CI, 0.890-0.950; sensitivity, 87%; specificity, 80%). Conclusion The increased LFCR measured by MRI was associated with a significantly higher risk for ACL injury or rerupture. The combination of LFCR, lateral tibial slope and lateral tibial height were the most predictive risk factors. This may help clinicians identify susceptible individuals and allow precision approaches for better prevention, treatment and management of this disease.
Collapse
Affiliation(s)
- Yang Sun
- Department of Sports Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Yun Tang
- Department of Sports Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
- Clinical Research Center, The First People’s Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
13
|
Bianchi E, Ruggeri M, Del Favero E, Pisano R, Artusio F, Ricci C, Vigani B, Ferraretto A, Boselli C, Icaro Cornaglia A, Rossi S, Sandri G. Chondroitin sulfate and caseinophosphopeptides doped polyurethane-based highly porous 3D scaffolds for tendon-to-bone regeneration. Int J Pharm 2024; 652:123822. [PMID: 38242257 DOI: 10.1016/j.ijpharm.2024.123822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Tendon disorders are common injuries, which can be greatly debilitating as they are often accompanied by great pain and inflammation. Moreover, several problems are also related to the laceration of the tendon-to-bone interface (TBI), a specific region subjected to great mechanical stresses. The techniques used nowadays for the treatment of tendon and TBI injuries often involve surgery. However, one critical aspect of this procedure involves the elevated risk of fail due to the tissues weakening and the postoperative alterations of the normal joint mechanics. Synthetic polymers, such as thermoplastic polyurethane, are of special interest in the tissue engineering field as they allow the production of scaffolds with tunable elastic and mechanical properties, that could guarantee an effective support during the new tissue formation. Based on these premises, the aim of this work was the design and the development of highly porous 3D scaffolds based on thermoplastic polyurethane, and doped with chondroitin sulfate and caseinophosphopeptides, able to mimic the structural, biomechanical, and biochemical functions of the TBI. The obtained scaffolds were characterized by a homogeneous microporous structure, and by a porosity optimal for cell nutrition and migration. They were also characterized by remarkable mechanical properties, reaching values comparable to the ones of the native tendons. The scaffolds promoted the tenocyte adhesion and proliferation when caseinophosphopetides and chondroitin sulfate are present in the 3D structure. In particular, caseinophosphopeptides' optimal concentration for cell proliferation resulted 2.4 mg/mL. Finally, the systems evaluation in vivo demonstrated the scaffolds' safety, since they did not cause any inflammatory effect nor foreign body response, representing interesting platforms for the regeneration of injured TBI.
Collapse
Affiliation(s)
- Eleonora Bianchi
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Marco Ruggeri
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Elena Del Favero
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Roberto Pisano
- Department of Applied Science and Technology (DISAT), Polytechnic of Torino, Torino, Italy
| | - Fiora Artusio
- Department of Applied Science and Technology (DISAT), Polytechnic of Torino, Torino, Italy
| | - Caterina Ricci
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Anita Ferraretto
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, via Mangiagalli 31, Milan, Italy; Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Galeazzi-Sant'Ambrogio, Via Cristina Belgioioso 173, 20157 Milan, Italy
| | - Cinzia Boselli
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Antonia Icaro Cornaglia
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 2, 27100 Pavia, Italy
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
14
|
Li Z, Hou D, Tang Z, Xiong L, Yan Y. The potential role of stem cells-derived extracellular vesicles in the treatment of musculoskeletal system diseases. Cell Biol Int 2024; 48:237-252. [PMID: 38100269 DOI: 10.1002/cbin.12107] [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: 05/17/2022] [Revised: 09/29/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023]
Abstract
The therapeutic potential of stem cells-derived extracellular vesicles (EVs) has shown a great progress in the regenerative medicine. EVs are rich in a variety of bioactive substances, which are important carriers of signal transmission and interactions between cells, and they play an important role in the processes of tissue repair and regeneration. Several studies have shown that stem cells-derived EVs regulate immunity, promote cell proliferation and differentiation, enhance bone and vascular regeneration, and play an increasingly important role in musculoskeletal system. This review aimed to describe the biological characteristics of stem cells-derived EVs and discuss their potential role in the therapy of musculoskeletal system diseases.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Demiao Hou
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Zijin Tang
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Lishun Xiong
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yiguo Yan
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
15
|
Guo C, Zhang Y, Dong W, Huang B, Liu Y. Risk factors and clinical characteristics of surgical site infections in athletes undergoing Achilles tendon repair surgery. Int Wound J 2024; 21:e14666. [PMID: 38420668 PMCID: PMC10902687 DOI: 10.1111/iwj.14666] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 03/02/2024] Open
Abstract
Achilles tendon ruptures are common in athletes, requiring surgical intervention. However, the risk of surgical site infections (SSIs) post-surgery poses significant challenges. This study aims to analyse the risk factors and microbial aetiology associated with SSIs in athletes undergoing Achilles tendon repair. A comprehensive retrospective analysis was conducted from May 2021 to July 2023. The study included 25 patients with SSIs (case group) and 50 patients without SSIs (control group) post Achilles tendon repair surgery. Inclusion criteria encompassed patients with medically confirmed Achilles tendon ruptures who underwent surgical repair. Exclusion criteria included prior tendon pathologies and significant chronic illnesses. Diagnostic criteria for SSIs involved symptoms like elevated body temperature and localized tenderness, along with laboratory confirmations such as positive microbiological cultures. The study utilized VITEK® 2 for bacterial identification and involved statistical analyses like univariate and multivariate logistic regression. The study identified Staphylococcus aureus as the primary pathogen in SSIs. Significant risk factors included lack of prophylactic antibiotic use, presence of diabetes, open wounds and prolonged surgery duration. Univariate analysis revealed stark contrasts in these factors between infected and non-infected groups, while multivariate analysis underscored their importance in SSI development. S. aureus emerged as the predominant pathogen in SSIs post Achilles tendon repair. Critical risk factors such as absence of prophylactic antibiotics, diabetes, open wounds and extended surgery duration play a vital role in SSIs. Addressing these factors is essential for better postoperative outcomes in Achilles tendon repair surgeries.
Collapse
Affiliation(s)
- Chenhao Guo
- College of Physical EducationShanxi UniversityTaiyuanChina
| | - Yuze Zhang
- College of Physical EducationShanxi UniversityTaiyuanChina
| | - Wenhan Dong
- College of Physical EducationTaiyuan University of TechnologyTaiyuanChina
| | - Bo Huang
- Department of Rehabilitation MedicineTaiyuan Seventh People's HospitalTaiyuanChina
| | - Yinghai Liu
- College of Physical EducationShanxi UniversityTaiyuanChina
| |
Collapse
|
16
|
Ganesh S B, Sabu A, Kaarthikeyan G, Eswaramoorthy R, P T P. Development of a Cissus quadrangularis-Doped Extracellular Matrix and a Hyaluronic Acid-Incorporated Scaffold for Periodontal Regeneration: An In Vitro Study. Cureus 2024; 16:e56507. [PMID: 38646344 PMCID: PMC11026305 DOI: 10.7759/cureus.56507] [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: 01/12/2024] [Accepted: 03/19/2024] [Indexed: 04/23/2024] Open
Abstract
PURPOSE The study aimed to analyze whether adding Cissus quadrangularis (CQ) extract and the extracellular matrix of ovine tendon (TENDON) increases the regenerative potential of mesenchymal stem cells produced in hyaluronic acid (HA) scaffolds for tenogenesis. MATERIALS AND METHODS Fifty grams of powdered CQ was mixed with 250 mL of ethanol to prepare the extract. Two grams of hyaluronic acid powder was added to 100 mL of distilled water to make the HA solution. The ovine tendon was decellularized using a mixture of 10% phosphate-buffered saline (PBS), sodium dodecyl sulfate (SDS), and Triton-X. The hydrogel samples were prepared by mixing the extracellular matrix of tendon, HA, and CQ, after which they were divided into study groups such as HA, HA + CQ, HA + TENDON, and HA + CQ + TENDON. Scanning electron microscopy (SEM) analysis, swelling analysis, differentiation analysis, compression test, compatibility assay, and tenogenesis assay were later conducted. RESULTS The morphology of the samples was analyzed using SEM. Low levels of swelling of the hydrogels were observed. Cells were found to be viable and showed good differentiation and tenogenesis. Optimal compression levels were observed, and the properties of the prepared hydrogels were satisfactory. CONCLUSION The results suggest that the addition of CQ considerably increases the tenogenic potential of the extracellular matrix/HA scaffold. Hence, it can be used as a regenerative material for periodontal tissue regeneration.
Collapse
Affiliation(s)
- Balaji Ganesh S
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Abraham Sabu
- Dentistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - G Kaarthikeyan
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Rajalakshmanan Eswaramoorthy
- Biomaterials, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Priyangha P T
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
17
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
18
|
Li Y, Deng T, Aili D, Chen Y, Zhu W, Liu Q. Cell Sheet Technology: An Emerging Approach for Tendon and Ligament Tissue Engineering. Ann Biomed Eng 2024; 52:141-152. [PMID: 37731091 DOI: 10.1007/s10439-023-03370-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/09/2023] [Indexed: 09/22/2023]
Abstract
Tendon and ligament injuries account for a substantial proportion of disorders in the musculoskeletal system. While non-operative and operative treatment strategies have advanced, the restoration of native tendon and ligament structures after injury is still challenging due to its innate limited regenerative ability. Cell sheet technology is an innovative tool for tissue fabrication and cell transplantation in regenerative medicine. In this review, we first summarize different harvesting procedures and advantages of cell sheet technology, which preserves intact cell-to-cell connections and extracellular matrix. We then describe the recent progress of cell sheet technology from preclinical studies, focusing on the application of stem cell-derived sheets in treating tendon and ligament injuries, as well as highlighting its effects on mitigating inflammation and promoting tendon/graft-bone interface healing. Finally, we discuss several prerequisites for future clinical translation including the selection of appropriate cell source, optimization of preparation process, establishment of suitable animal model, and the fabrication of vascularized complex tissue. We believe this review could potentially provoke new ideas and drive the development of more functional biomimetic tissues using cell sheet technology to meet the needs of clinical patients.
Collapse
Affiliation(s)
- Yexin Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ting Deng
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Dilihumaer Aili
- Department of Orthopedic Surgery, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi, People's Republic of China
| | - Yang Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Qian Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| |
Collapse
|
19
|
Türkoğlu GC, Khomarloo N, Mohsenzadeh E, Gospodinova DN, Neznakomova M, Salaün F. PVA-Based Electrospun Materials-A Promising Route to Designing Nanofiber Mats with Desired Morphological Shape-A Review. Int J Mol Sci 2024; 25:1668. [PMID: 38338946 PMCID: PMC10855838 DOI: 10.3390/ijms25031668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Poly(vinyl alcohol) is one of the most attractive polymers with a wide range of uses because of its water solubility, biocompatibility, low toxicity, good mechanical properties, and relatively low cost. This review article focuses on recent advances in poly(vinyl alcohol) electrospinning and summarizes parameters of the process (voltage, distance, flow rate, and collector), solution (molecular weight and concentration), and ambient (humidity and temperature) in order to comprehend the influence on the structural, mechanical, and chemical properties of poly(vinyl alcohol)-based electrospun matrices. The importance of poly(vinyl alcohol) electrospinning in biomedical applications is emphasized by exploring a literature review on biomedical applications including wound dressings, drug delivery, tissue engineering, and biosensors. The study also highlights a new promising area of particles formation through the electrospraying of poly(vinyl alcohol). The limitations and advantages of working with different poly(vinyl alcohol) matrices are reviewed, and some recommendations for the future are made to advance this field of study.
Collapse
Affiliation(s)
- Gizem Ceylan Türkoğlu
- Department of Textile Engineering, Dokuz Eylul University, İzmir 35397, Turkey;
- Univ. Lille, ENSAIT, ULR 2461-GEMTEX-Génie et Matériaux Textiles, F-59000 Lille, France; (N.K.); (E.M.)
| | - Niloufar Khomarloo
- Univ. Lille, ENSAIT, ULR 2461-GEMTEX-Génie et Matériaux Textiles, F-59000 Lille, France; (N.K.); (E.M.)
- Univ. Lille, ENSAIT, ULR 2461-GEMTEX-Génie et Matériaux Textiles, Junia, F-59000 Lille, France
| | - Elham Mohsenzadeh
- Univ. Lille, ENSAIT, ULR 2461-GEMTEX-Génie et Matériaux Textiles, F-59000 Lille, France; (N.K.); (E.M.)
- Univ. Lille, ENSAIT, ULR 2461-GEMTEX-Génie et Matériaux Textiles, Junia, F-59000 Lille, France
| | - Dilyana Nikolaeva Gospodinova
- Faculty of Electrical Engineering, Department of Electrical Apparatus, Technical University of Sofia, 1156 Sofia, Bulgaria;
| | - Margarita Neznakomova
- Faculty of Industrial Technology, Department of Material Science and Technology of Materials, Technical University of Sofia, 1000 Sofia, Bulgaria;
| | - Fabien Salaün
- Univ. Lille, ENSAIT, ULR 2461-GEMTEX-Génie et Matériaux Textiles, F-59000 Lille, France; (N.K.); (E.M.)
| |
Collapse
|
20
|
Zhong S, Lan Y, Liu J, Seng Tam M, Hou Z, Zheng Q, Fu S, Bao D. Advances focusing on the application of decellularization methods in tendon-bone healing. J Adv Res 2024:S2090-1232(24)00033-X. [PMID: 38237768 DOI: 10.1016/j.jare.2024.01.020] [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: 09/18/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND The tendon or ligament is attached to the bone by a triphasic but continuous area of heterogeneous tissue called the tendon-bone interface (TBI). The rapid and functional regeneration of TBI is challenging owing to its complex composition and difficulty in self-healing. The development of new technologies, such as decellularization, has shown promise in the regeneration of TBI. Several ex vivo and in vivo studies have shown that decellularized grafts and decellularized biomaterial scaffolds achieved better efficacy in enhancing TBI healing. However further information on the type of review that is available is needed. AIM OF THE REVIEW In this review, we discuss the current application of decellularization biomaterials in promoting TBI healing and the possible mechanisms involved. With this work, we would like to reveal how tissues or biomaterials that have been decellularized can improve tendon-bone healing and to provide a theoretical basis for future related studies. KEY SCIENTIFIC CONCEPTS OF THE REVIEW Decellularization is an emerging technology that utilizes various chemical, enzymatic and/or physical strategies to remove cellular components from tissues while retaining the structure and composition of the extracellular matrix (ECM). After decellularization, the cellular components of the tissue that cause an immune response are removed, while various biologically active biofactors are retained. This review further explores how tissues or biomaterials that have been decellularized improve TBI healing.
Collapse
Affiliation(s)
- Sheng Zhong
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yujian Lan
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jinyu Liu
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | | | - Zhipeng Hou
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qianghua Zheng
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shijie Fu
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Dingsu Bao
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China; Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| |
Collapse
|
21
|
von Witzleben M, Hahn J, Richter RF, de Freitas B, Steyer E, Schütz K, Vater C, Bernhardt A, Elschner C, Gelinsky M. Tailoring the pore design of embroidered structures by melt electrowriting to enhance the cell alignment in scaffold-based tendon reconstruction. BIOMATERIALS ADVANCES 2024; 156:213708. [PMID: 38029698 DOI: 10.1016/j.bioadv.2023.213708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Tissue engineering of ligaments and tendons aims to reproduce the complex and hierarchical tissue structure while meeting the biomechanical and biological requirements. For the first time, the additive manufacturing methods of embroidery technology and melt electrowriting (MEW) were combined to mimic these properties closely. The mechanical benefits of embroidered structures were paired with a superficial micro-scale structure to provide a guide pattern for directional cell growth. An evaluation of several previously reported MEW fiber architectures was performed. The designs with the highest cell orientation of primary dermal fibroblasts were then applied to embroidery structures and subsequently evaluated using human adipose-derived stem cells (AT-MSC). The addition of MEW fibers resulted in the formation of a mechanically robust layer on the embroidered scaffolds, leading to composite structures with mechanical properties comparable to those of the anterior cruciate ligament. Furthermore, the combination of embroidered and MEW structures supports a higher cell orientation of AT-MSC compared to embroidered structures alone. Collagen coating further promoted cell attachment. Thus, these investigations provide a sound basis for the fabrication of heterogeneous and hierarchical synthetic tendon and ligament substitutes.
Collapse
Affiliation(s)
- Max von Witzleben
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany
| | - Judith Hahn
- Leibniz-Institut für Polymerforschung Dresden e. V. (IPF), Institute of Polymer Materials, Hohe Str. 6, 01069 Dresden, Germany
| | - Ron F Richter
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany
| | - Bianca de Freitas
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany
| | - Emily Steyer
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany
| | - Kathleen Schütz
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany
| | - Corina Vater
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany
| | - Anne Bernhardt
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany
| | - Cindy Elschner
- Leibniz-Institut für Polymerforschung Dresden e. V. (IPF), Institute of Polymer Materials, Hohe Str. 6, 01069 Dresden, Germany
| | - Michael Gelinsky
- Technische Universität Dresden, University Hospital Carl Gustav Carus and Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Fetscherstr. 74, 01307 Dresden, Germany.
| |
Collapse
|
22
|
Zhu Y, Yan J, Zhang H, Cui G. Bone marrow mesenchymal stem cell‑derived exosomes: A novel therapeutic agent for tendon‑bone healing (Review). Int J Mol Med 2023; 52:121. [PMID: 37937691 PMCID: PMC10635703 DOI: 10.3892/ijmm.2023.5324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
In sports medicine, injuries related to the insertion of tendons into bones, including rotator cuff injuries, anterior cruciate ligament injuries and Achilles tendon ruptures, are commonly observed. However, traditional therapies have proven to be insufficient in achieving satisfactory outcomes due to the intricate anatomical structure associated with these injuries. Adult bone marrow mesenchymal stem cells possess self‑renewal and multi‑directional differentiation potential and can generate various mesenchymal tissues to aid in the recovery of bone, cartilage, adipose tissue and bone marrow hematopoietic tissue. In addition, extracellular vesicles derived from bone marrow mesenchymal stem cells known as exosomes, contain lipids, proteins and nucleic acids that govern the tissue microenvironment, facilitate tissue repair and perform various biological functions. Studies have demonstrated that bone marrow mesenchymal stem cell‑derived exosomes can function as natural nanocapsules for drug delivery and can enhance tendon‑bone healing strength. The present review discusses the latest research results on the role of exosomes released by bone marrow mesenchymal stem cells in tendon‑bone healing and provides valuable information for implementing these techniques in regenerative medicine and sports health.
Collapse
Affiliation(s)
- Yongjia Zhu
- Department of Arthritis, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Jiapeng Yan
- Department of Arthritis, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Hongfei Zhang
- Department of Arthritis, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Guanxing Cui
- Department of Arthritis, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
23
|
Adjei-Sowah E, Chandrasiri I, Xiao B, Liu Y, Ackerman JE, Soto C, Nichols AEC, Nolan K, Benoit DSW, Loiselle AE. Development of a Nanoparticle-Based Tendon-Targeting Drug Delivery System to Pharmacologically Modulate Tendon Healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569204. [PMID: 38076889 PMCID: PMC10705411 DOI: 10.1101/2023.11.29.569204] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Tendon regeneration following acute injury is marred by a fibrotic healing response that prevents complete functional recovery. Despite the high frequency of tendon injuries and the poor outcomes, including functional deficits and elevated risk of re-injury, there are currently no pharmacological therapies in clinical use to enhance the healing process. Several promising pharmacotherapies have been identified; however, systemic treatments lack tendon specificity, resulting in poor tendon biodistribution and perhaps explaining the largely limited beneficial effects of these treatments on the tendon healing process. To address this major unmet need, we leveraged our existing spatial transcriptomics dataset of the tendon healing process to identify an area of the healing tendon that is enriched for expression of Acp5. Acp5 encodes tartrate-resistant acid phosphatase (TRAP), and we demonstrate robust TRAP activity in the healing tendon. This unexpected finding allowed us to refine and apply our existing TRAP binding peptide (TBP) functionalized nanoparticle (NP) drug delivery system (DDS) to facilitate improved delivery of systemic treatments to the healing tendon. To demonstrate the translational potential of this drug delivery system, we delivered the S100a4 inhibitor, Niclosamide to the healing tendon. We have previously shown that genetic knockdown of S100a4 enhances tendon healing. While systemic delivery of Niclosamide did not affect the healing process, relative to controls, TBP-NP delivery of Niclosamide enhanced both functional and mechanical outcome measures. Collectively, these data identify a novel tendon-targeting drug delivery system and demonstrate the translational potential of this approach to enhance the tendon healing process.
Collapse
|
24
|
Hu J, Jiang Z, Zhang J, Yang G. Application of silk fibroin coatings for biomaterial surface modification: a silk road for biomedicine. J Zhejiang Univ Sci B 2023; 24:943-956. [PMID: 37961798 PMCID: PMC10646393 DOI: 10.1631/jzus.b2300003] [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: 01/02/2023] [Accepted: 04/18/2023] [Indexed: 11/15/2023]
Abstract
Silk fibroin (SF) as a natural biopolymer has become a popular material for biomedical applications due to its minimal immunogenicity, tunable biodegradability, and high biocompatibility. Nowadays, various techniques have been developed for the applications of SF in bioengineering. Most of the literature reviews focus on the SF-based biomaterials and their different forms of applications such as films, hydrogels, and scaffolds. SF is also valuable as a coating on other substrate materials for biomedicine; however, there are few reviews related to SF-coated biomaterials. Thus, in this review, we focused on the surface modification of biomaterials using SF coatings, demonstrated their various preparation methods on substrate materials, and introduced the latest procedures. The diverse applications of SF coatings for biomedicine are discussed, including bone, ligament, skin, mucosa, and nerve regeneration, and dental implant surface modification. SF coating is conducive to inducing cell adhesion and migration, promoting hydroxyapatite (HA) deposition and matrix mineralization, and inhibiting the Notch signaling pathway, making it a promising strategy for bone regeneration. In addition, SF-coated composite scaffolds can be considered prospective candidates for ligament regeneration after injury. SF coating has been proven to enhance the mechanical properties of the substrate material, and render integral stability to the dressing material during the regeneration of skin and mucosa. Moreover, SF coating is a potential strategy to accelerate nerve regeneration due to its dielectric properties, mechanical flexibility, and angiogenesis promotion effect. In addition, SF coating is an effective and popular means for dental implant surface modification to promote osteogenesis around implants made of different materials. Thus, this review can be of great benefit for further improvements in SF-coated biomaterials, and will undoubtedly contribute to clinical transformation in the future.
Collapse
Affiliation(s)
- Jinxing Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Jing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
25
|
Lp MRL, Agrawal DK. Biomechanical Forces in the Tissue Engineering and Regeneration of Shoulder, Hip, Knee, and Ankle Joints. JOURNAL OF BIOTECHNOLOGY AND BIOMEDICINE 2023; 6:491-500. [PMID: 38037618 PMCID: PMC10688570 DOI: 10.26502/jbb.2642-91280111] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Tear on the tendon, ligament and articular cartilage of the joints do not heal by itself and new modalities of treatment are required to address the need for full restoration of joint functions. Accompanied by degenerative diseases, the healing of these tissues does not occur naturally and hence requires surgical interventions, but with associated morbidity. Tissue engineering strategies are now focusing on the effective incorporation of biomechanical stimulation by the application of biomechanical forces relevant to the tissue of interest to regenerate and engineer functional tissues. Bioreactors are being continuously developed to accomplish this goal. Although bioreactors have been developed, the advancement in the field of biomaterial, basic science, and cell engineering warrant further refinement for their effective use. In this article we reviewed the application of biomechanical forces in the tissue engineering and regeneration of the joints such as rotator cuff of shoulder, ball and socket joint of the hip, articular cartilage of knee, and the ankle joints.
Collapse
Affiliation(s)
- Merlin Rajesh Lal Lp
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| | - Devendra K Agrawal
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| |
Collapse
|
26
|
Ferrell JL, Dodson A, Martin J. Microfragmented adipose tissue in the treatment of a full-thickness supraspinatus tear: a case report. Regen Med 2023; 18:773-780. [PMID: 37727974 DOI: 10.2217/rme-2023-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
A 70-year-old female presented with an 8-month history of right anterior shoulder pain and weakness, unresolved with conservative management. Among other shoulder pathology, the patient was diagnosed with a full-thickness supraspinatus tear and elected to proceed with the microfragmented adipose tissue procedure to treat the injured tendon and nearby relevant structures. Improvements in pain and function were documented along with progressive healing of the supraspinatus on ultrasound and MRI following the procedure. This case demonstrates the efficacy of microfragmented adipose tissue as a relatively novel approach to treating non-retracted, full-thickness rotator cuff tears.
Collapse
Affiliation(s)
- John L Ferrell
- Regenerative Orthopedics and Sports Medicine, Washington DC, 20036, USA
| | - Alanna Dodson
- Regenerative Orthopedics and Sports Medicine, Washington DC, 20036, USA
| | - Joshua Martin
- Regenerative Orthopedics and Sports Medicine, Washington DC, 20036, USA
| |
Collapse
|
27
|
Schynkel L, Meeremans M, Meyer AA, Schoolaert E, Geltmeyer J, Omidinia-Anarkoli A, Van Vlierberghe S, Daelemans L, De Laporte L, De Schauwer C, Hoogenboom R, De Clerck K. Cell Guiding Multicomponent Nanoyarn Tendon Scaffolds with Tunable Morphology and Flexibility. ACS APPLIED MATERIALS & INTERFACES 2023; 15:42241-42250. [PMID: 37650520 DOI: 10.1021/acsami.3c08241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Nanofibrous scaffolds are widely investigated for tendon tissue engineering due to their porous structure, high flexibility, and the ability to guide cells in a preferred direction. Previous research has shown that providing a microenvironment similar to in vivo settings improves tissue regeneration. Therefore, in this work, ingenious multicomponent nanoyarn scaffolds that mimic the fibrillar and tubular structures of tendons are developed for the first time through electrospinning and bundling nanoyarns followed by electrospinning of a nanofibrous shell around the bundle. Multicomponent nanoyarn scaffolds out of poly(ε-caprolactone) with varying porosity, density, and diameter were successfully produced by coelectrospinning with water-soluble poly(2-ethyl-2-oxazoline) as a sacrificial component. The diameter and fiber orientation of the nanoyarns were successfully tuned based on parameter-morphology models obtained by the design of experiments. Cyclic bending tests were performed, indicating that the flexibility of the multicomponent nanoyarn scaffolds depends on the morphology and can be tuned through controlling the number of nanoyarns in the bundle and the porosity. Indirect and direct cell culture tests using mouse and equine tendon cells revealed excellent cytocompatibility of the nanofibrous products and demonstrated the potential of the nanoyarns to guide the growing cells along the nanofiber direction, which is crucial for tendon tissue engineering.
Collapse
Affiliation(s)
- Lucas Schynkel
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | - Marguerite Meeremans
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Anna A Meyer
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstrasse. 50, 52074 Aachen, Germany
- Institute of Technical and Macromolecular Chemistry (ITMC), RWTH University Aachen, Worringerweg 2, 52074 Aachen ,Germany
| | - Ella Schoolaert
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | - Jozefien Geltmeyer
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | | | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281 - Building S4, 9000 Ghent, Belgium
| | - Lode Daelemans
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| | - Laura De Laporte
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstrasse. 50, 52074 Aachen, Germany
- Institute of Technical and Macromolecular Chemistry (ITMC), RWTH University Aachen, Worringerweg 2, 52074 Aachen ,Germany
- Advanced Materials for Biomedicine (AMB), Institute of Applied Medical Engineering (AME), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074 Aachen ,Germany
| | - Catharina De Schauwer
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281 - Building S4, 9000 Ghent, Belgium
| | - Karen De Clerck
- Centre for Textile Science and Engineering, Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Tech Lane Science Park 70A, 9052 Ghent, Belgium
| |
Collapse
|
28
|
Degradation behavior of ZE21C magnesium alloy suture anchors and their effect on ligament-bone junction repair. Bioact Mater 2023; 26:128-141. [PMID: 36891259 PMCID: PMC9986500 DOI: 10.1016/j.bioactmat.2023.02.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/18/2023] [Accepted: 02/18/2023] [Indexed: 03/04/2023] Open
Abstract
Current materials comprising suture anchors used to reconstruct ligament-bone junctions still have limitation in biocompatibility, degradability or mechanical properties. Magnesium alloys are potential bone implant materials, and Mg2+ has been shown to promote ligament-bone healing. Here, we used Mg-2 wt.% Zn-0.5 wt.% Y-1 wt.% Nd-0.5 wt.% Zr (ZE21C) alloy and Ti6Al4V (TC4) alloy to prepare suture anchors to reconstruct the patellar ligament-tibia in SD rats. We studied the degradation behavior of the ZE21C suture anchor via in vitro and in vivo experiments and assessed its reparative effect on the ligament-bone junction. In vitro, the ZE21C suture anchor degraded gradually, and calcium and phosphorus products accumulated on its surface during degradation. In vivo, the ZE21C suture anchor could maintain its mechanical integrity within 12 weeks of implantation in rats. The tail of the ZE21C suture anchor in high stress concentration degraded rapidly during the early implantation stage (0-4weeks), while bone healing accelerated the degradation of the anchor head in the late implantation stage (4-12weeks). Radiological, histological, and biomechanical assays indicated that the ZE21C suture anchor promoted bone healing above the suture anchor and fibrocartilaginous interface regeneration in the ligament-bone junction, leading to better biomechanical strength than the TC4 group. Hence, this study provides a basis for further research on the clinical application of degradable magnesium alloy suture anchors.
Collapse
|
29
|
Son YH, Yang DH, Uricoli B, Park SJ, Jeong GJ, Chun HJ. Three-Dimensional Cell Culture System for Tendon Tissue Engineering. Tissue Eng Regen Med 2023; 20:553-562. [PMID: 37278865 PMCID: PMC10313620 DOI: 10.1007/s13770-023-00550-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: 02/07/2023] [Revised: 04/07/2023] [Accepted: 05/01/2023] [Indexed: 06/07/2023] Open
Abstract
Tendon, connective tissue between bone and muscle has unique component of the musculoskeletal system. It plays important role for transporting mechanical stress from muscle to bone and enabling locomotive motion of the body. There are some restoration capacities in the tendon tissue, but the injured tendons are not completely regenerated after acute and chronic tendon injury. At this point, the treatment options for tendon injuries are limited and not that successful. Therefore, biomedical engineering approaches are emerged to cope with this issue. Among them, three-dimensional cell culture platforms provided similarity to in vivo conditions and suggested opportunities for new therapeutic approaches for treatment of tendon injuries. In this review, we focus on the characteristics of tendon tissue and tendon pathologies which can be targets for tendon tissue engineering strategies. Then proof-of-concept and pre-clinical studies leveraging advanced 3-dimensional cell culture platforms for tendon tissue regeneration have been discussed.
Collapse
Affiliation(s)
- Young Hoon Son
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dae Hyeok Yang
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, the Republic of Korea
| | - Biaggio Uricoli
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sung-Jin Park
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Gun-Jae Jeong
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, the Republic of Korea.
| | - Heung Jae Chun
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, the Republic of Korea.
| |
Collapse
|
30
|
Liang W, Zhou C, Meng Y, Fu L, Zeng B, Liu Z, Ming W, Long H. An overview of the material science and knowledge of nanomedicine, bioscaffolds, and tissue engineering for tendon restoration. Front Bioeng Biotechnol 2023; 11:1199220. [PMID: 37388772 PMCID: PMC10306281 DOI: 10.3389/fbioe.2023.1199220] [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: 04/03/2023] [Accepted: 05/29/2023] [Indexed: 07/01/2023] Open
Abstract
Tendon wounds are a worldwide health issue affecting millions of people annually. Due to the characteristics of tendons, their natural restoration is a complicated and lengthy process. With the advancement of bioengineering, biomaterials, and cell biology, a new science, tissue engineering, has developed. In this field, numerous ways have been offered. As increasingly intricate and natural structures resembling tendons are produced, the results are encouraging. This study highlights the nature of the tendon and the standard cures that have thus far been utilized. Then, a comparison is made between the many tendon tissue engineering methodologies proposed to date, concentrating on the ingredients required to gain the structures that enable appropriate tendon renewal: cells, growth factors, scaffolds, and scaffold formation methods. The analysis of all these factors enables a global understanding of the impact of each component employed in tendon restoration, thereby shedding light on potential future approaches involving the creation of novel combinations of materials, cells, designs, and bioactive molecules for the restoration of a functional tendon.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, Zhejiang, China
| | - Yanfeng Meng
- Department of Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Lifeng Fu
- Department of Orthopedics, Shaoxing City Keqiao District Hospital of Traditional Chinese Medicine, Shaoxing, Zhejiang, China
| | - Bin Zeng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Zunyong Liu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Wenyi Ming
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Hengguo Long
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| |
Collapse
|
31
|
Yun HW, Jin YJ, Shin DI, Noh S, Kim KM, Park JY, Lim S, Park DY. Fibrocartilage extracellular matrix augmented demineralized bone matrix graft repairs tendon-to-bone interface in a rabbit tendon reconstruction model. BIOMATERIALS ADVANCES 2023; 152:213522. [PMID: 37343332 DOI: 10.1016/j.bioadv.2023.213522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023]
Abstract
Current tendon/ligament reconstructions integrate via scar tissue rather than proper bone-tendon interface regeneration, which affects graft longevity, changes in bone tunnel size, and functional outcomes. The purpose of this study was to develop a functional demineralized bone matrix (DBM) + fibrocartilage extracellular matrix (FCECM) composite scaffold, characterize its physicochemical properties, and evaluate its efficacy in repairing tendon-bone interface in a rabbit tendon reconstruction model. Solubilized FCECM was loaded and crosslinked on to DBM scaffolds via gamma-irradiation to create DBM + FCECM scaffolds. The resulting scaffold showed interconnected pores coated with FCECM and protein cargo similar to FCECM. The addition of FCECM modified the physicochemical properties of the DBM scaffold, including microstructure, biochemical composition, mechanical strength, thermodynamic properties, and degradation period. The DBM + FCECM scaffold was biocompatible for mesenchymal stem cells (MSCs) and resulted in elevation of fibrochondrogenic gene markers compared to DBM scaffolds in vitro. In vivo implantation of DBM + FCECM scaffold resulted in neofibrocartilage formation, better pullout strength, and less bone tunnel widening compared to DBM only group in a rabbit tendon reconstruction model. In conclusion, the FCECM augmented DBM scaffold repairs the tendon-bone interface with osseous-fibrocartilage tissue, which may be utilized to improve current tendon reconstruction surgeries.
Collapse
Affiliation(s)
- Hee-Woong Yun
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Yong Jun Jin
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Dong Il Shin
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Sujin Noh
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Kyu Min Kim
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Jae-Young Park
- Department of Orthopedic Surgery, CHA Bundang Medical Center, School of Medicine, CHA University, Pocheon 13496, Gyeonggi-do, Republic of Korea
| | - Sumin Lim
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Do Young Park
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea.
| |
Collapse
|
32
|
Huang L, Chen L, Chen H, Wang M, Jin L, Zhou S, Gao L, Li R, Li Q, Wang H, Zhang C, Wang J. Biomimetic Scaffolds for Tendon Tissue Regeneration. Biomimetics (Basel) 2023; 8:246. [PMID: 37366841 DOI: 10.3390/biomimetics8020246] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Tendon tissue connects muscle to bone and plays crucial roles in stress transfer. Tendon injury remains a significant clinical challenge due to its complicated biological structure and poor self-healing capacity. The treatments for tendon injury have advanced significantly with the development of technology, including the use of sophisticated biomaterials, bioactive growth factors, and numerous stem cells. Among these, biomaterials that the mimic extracellular matrix (ECM) of tendon tissue would provide a resembling microenvironment to improve efficacy in tendon repair and regeneration. In this review, we will begin with a description of the constituents and structural features of tendon tissue, followed by a focus on the available biomimetic scaffolds of natural or synthetic origin for tendon tissue engineering. Finally, we will discuss novel strategies and present challenges in tendon regeneration and repair.
Collapse
Affiliation(s)
- Lvxing Huang
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Le Chen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310000, China
| | - Hengyi Chen
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Manju Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310000, China
| | - Letian Jin
- School of Medical Imaging, Hangzhou Medical College, Hangzhou 310000, China
| | - Shenghai Zhou
- School of Medical Imaging, Hangzhou Medical College, Hangzhou 310000, China
| | - Lexin Gao
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Ruwei Li
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Quan Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310000, China
| | - Hanchang Wang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou 310000, China
| | - Can Zhang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Junjuan Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310000, China
| |
Collapse
|
33
|
Papalamprou A, Yu V, Chen A, Stefanovic T, Kaneda G, Salehi K, Castaneda CM, Gertych A, Glaeser JD, Sheyn D. Directing iPSC differentiation into iTenocytes using combined scleraxis overexpression and cyclic loading. J Orthop Res 2023; 41:1148-1161. [PMID: 36203346 PMCID: PMC10076443 DOI: 10.1002/jor.25459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 02/04/2023]
Abstract
Regenerative therapies for tendon are falling behind other tissues due to the lack of an appropriate and potent cell therapeutic candidate. This study aimed to induce tenogenesis using stable Scleraxis (Scx) overexpression in combination with uniaxial mechanical stretch of iPSC-derived mesenchymal stromal-like cells (iMSCs). Scx is the single direct molecular regulator of tendon differentiation known to date. Bone marrow-derived (BM-)MSCs were used as reference. Scx overexpression alone resulted in significantly higher upregulation of tenogenic markers in iMSCs compared to BM-MSCs. Mechanoregulation is known to be a central element guiding tendon development and healing. Mechanical stimulation combined with Scx overexpression resulted in morphometric and cytoskeleton-related changes, upregulation of early and late tendon markers, and increased extracellular matrix deposition and alignment, and tenomodulin perinuclear localization in iMSCs. Our findings suggest that these cells can be differentiated into tenocytes and might be a better candidate for tendon cell therapy applications than BM-MSCs.
Collapse
Affiliation(s)
- Angela Papalamprou
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Victoria Yu
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Angel Chen
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tina Stefanovic
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Giselle Kaneda
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Khosrowdad Salehi
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Chloe M. Castaneda
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Arkadiusz Gertych
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Juliane D. Glaeser
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
34
|
Zou M, Wang J, Shao Z. Therapeutic Potential of Exosomes in Tendon and Tendon-Bone Healing: A Systematic Review of Preclinical Studies. J Funct Biomater 2023; 14:299. [PMID: 37367263 PMCID: PMC10299056 DOI: 10.3390/jfb14060299] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Exosomes have been proven to play a positive role in tendon and tendon-bone healing. Here, we systematically review the literature to evaluate the efficacy of exosomes in tendon and tendon-bone healing. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a systematic and comprehensive review of the literature was performed on 21 January 2023. The electronic databases searched included Medline (through PubMed), Web of Science, Embase, Scopus, Cochrane Library and Ovid. In the end, a total of 1794 articles were systematically reviewed. Furthermore, a "snowball" search was also carried out. Finally, forty-six studies were included for analysis, with the total sample size being 1481 rats, 416 mice, 330 rabbits, 48 dogs, and 12 sheep. In these studies, exosomes promoted tendon and tendon-bone healing and displayed improved histological, biomechanical and morphological outcomes. Some studies also suggested the mechanism of exosomes in promoting tendon and tendon-bone healing, mainly through the following aspects: (1) suppressing inflammatory response and regulating macrophage polarization; (2) regulating gene expression, reshaping cell microenvironment and reconstructing extracellular matrix; (3) promoting angiogenesis. The risk of bias in the included studies was low on the whole. This systematic review provides evidence of the positive effect of exosomes on tendon and tendon-bone healing in preclinical studies. The unclear-to-low risk of bias highlights the significance of standardization of outcome reporting. It should be noted that the most suitable source, isolation methods, concentration and administration frequency of exosomes are still unknown. Additionally, few studies have used large animals as subjects. Further studies may be required on comparing the safety and efficacy of different treatment parameters in large animal models, which would be conducive to the design of clinical trials.
Collapse
Affiliation(s)
- Mingrui Zou
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (M.Z.); (J.W.)
- Beijing Key Laboratory of Sports Injuries, Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Jingzhou Wang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (M.Z.); (J.W.)
- Beijing Key Laboratory of Sports Injuries, Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Zhenxing Shao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (M.Z.); (J.W.)
- Beijing Key Laboratory of Sports Injuries, Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| |
Collapse
|
35
|
Bianchi E, Ruggeri M, Vigani B, Del Favero E, Ricci C, Boselli C, Icaro Cornaglia A, Viseras C, Rossi S, Sandri G. Cerium Oxide and Chondroitin Sulfate Doped Polyurethane Scaffold to Bridge Tendons. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37220144 DOI: 10.1021/acsami.3c06144] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Tendon disorders are common medical conditions, which can be greatly debilitating as they are often accompanied by great pain and inflammation. The techniques used nowadays for the treatment of chronic tendon injuries often involve surgery. However, one critical aspect of this procedure involves the scar tissue, characterized by mechanical properties that vary from healthy tissue, rendering the tendons inclined to reinjury or rupture. Synthetic polymers, such as thermoplastic polyurethane, are of special interest in the tissue engineering field as they allow the production of scaffolds with controlled elastic and mechanical properties, which could guarantee an effective support during the new tissue formation. The aim of this work was the design and the development of tubular nanofibrous scaffolds based on thermoplastic polyurethane and enriched with cerium oxide nanoparticles and chondroitin sulfate. The scaffolds were characterized by remarkable mechanical properties, especially when tubular aligned, reaching values comparable to the ones of the native tendons. A weight loss test was performed, suggesting a degradation in prolonged times. In particular, the scaffolds maintained their morphology and also remarkable mechanical properties after 12 weeks of degradation. The scaffolds promoted the cell adhesion and proliferation, in particular when in aligned conformation. Finally, the systems in vivo did not cause any inflammatory effect, representing interesting platforms for the regeneration of injured tendons.
Collapse
Affiliation(s)
- Eleonora Bianchi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Marco Ruggeri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Elena Del Favero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, LITA Viale Fratelli Cervi 93, Segrate 20090, Italy
| | - Caterina Ricci
- Department of Medical Biotechnology and Translational Medicine, University of Milan, LITA Viale Fratelli Cervi 93, Segrate 20090, Italy
| | - Cinzia Boselli
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Antonia Icaro Cornaglia
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 2, Pavia 27100 , Italy
| | - César Viseras
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Campus of Cartuja, Granada 18071, Spain
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| |
Collapse
|
36
|
Hojabri M, Tayebi T, Kasravi M, Aghdaee A, Ahmadi A, Mazloomnejad R, Tarasi R, Shaabani A, Bahrami S, Niknejad H. Wet-spinnability and crosslinked Fiber properties of alginate/hydroxyethyl cellulose with varied proportion for potential use in tendon tissue engineering. Int J Biol Macromol 2023; 240:124492. [PMID: 37072060 DOI: 10.1016/j.ijbiomac.2023.124492] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/20/2023]
Abstract
Researchers have examined different bio-inspired materials in tissue engineering and regenerative medicine to fabricate scaffolds to address tendon regeneration requirements. We developed fibers based on alginate (Alg) and hydroxyethyl cellulose (HEC) by wet-spinning technique to mimic the fibrous sheath of ECM. Various proportions (25:75, 50:50, 75:25) of 1 % Alg and 4 % HEC were blended to this aim. Two steps of crosslinking with different concentrations of CaCl2 (2.5 and 5 %) and glutaraldehyde (2.5 %) were used to improve physical and mechanical properties. The fibers were characterized by FTIR, SEM, swelling, degradation, and tensile tests. The in vitro proliferation, viability, and migration of tenocytes on the fibers were also evaluated. Moreover, the biocompatibility of implanted fibers was investigated in an animal model. The results showed ionic and covalent molecular interactions between the components. In addition, by properly maintaining surface morphology, fiber alignment, and swelling, lower concentrations of HEC in the blending provided good degradability and mechanical features. The mechanical strength of fibers was in the range of collagenous fibers. Increasing the crosslinking led to significantly different mechanical behaviors in terms of tensile strength and elongation at break. Because of good in vitro and in vivo biocompatibility, tenocyte proliferation, and migration, the biological macromolecular fibers could serve as desirable tendon substitutes. This study provides more practical insight into tendon tissue engineering in translational medicine.
Collapse
Affiliation(s)
- Mahsa Hojabri
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Tayebi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Kasravi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Aghdaee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armin Ahmadi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Radman Mazloomnejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghayeh Tarasi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Shaabani
- Department of Polymer and Materials Chemistry, Faculty of Chemistry and Petroleum Science, Shahid Beheshti University, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Assessing the biocompatibility of bovine tendon scaffold, a step forward in tendon tissue engineering. Cell Tissue Bank 2023; 24:11-24. [PMID: 35596907 DOI: 10.1007/s10561-022-10012-w] [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: 10/01/2021] [Accepted: 04/25/2022] [Indexed: 11/02/2022]
Abstract
Tendon is a collagen-enriched, tough, and intricately arranged connective tissue that connects muscle to the bone and transmits forces, resulting in joint movement. High mechanical demands can affect normal tissues and may lead to severe disorders, which usually require replacement of the damaged tendon. In recent decades, various decellularization methods have been studied for tissue engineering applications. One of the major challenges in tendon decellularization is preservation of the tendon extracellular matrix (ECM) architecture to maintain natural tissue characteristics. The aim of the present study was to create a decellularized bovine Achilles tendon scaffold to investigate its cytocompatibility with seeded hAd-MSCs (human adipose derived-mesenchymal stem cells) and blastema tissue in vitro. Here, we describe a reliable procedure to decellularize bovine Achilles tendon using a combination of physical and chemical treatments including repetitive freeze-thaw cycles and the ionic detergent SDS, respectively. The decellularization effectiveness and cytocompatibility of the tendon scaffolds were verified by histological studies and scanning electron microscopy for up to 30 days after culture. Histological studies revealed hAd-MSC attachment and penetration into the scaffolds at 5, 10, 15 and 20 days of culture. However, a decrease in cell number was observed on days 25 and 30 after culture in vitro. Moreover, migration of the blastema tissue cells into the scaffold were shown at 10 to 25 days post culture, however, destruction of the scaffolds and reduction in cell number were observed on 30th day after culture. Our results suggest that this decellularization protocol is an effective and biocompatible procedure which supports the maintenance and growth of both hAd-MSCs and blastema cells, and thus might be promising for tendon tissue engineering.
Collapse
|
38
|
Ning C, Li P, Gao C, Fu L, Liao Z, Tian G, Yin H, Li M, Sui X, Yuan Z, Liu S, Guo Q. Recent advances in tendon tissue engineering strategy. Front Bioeng Biotechnol 2023; 11:1115312. [PMID: 36890920 PMCID: PMC9986339 DOI: 10.3389/fbioe.2023.1115312] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Tendon injuries often result in significant pain and disability and impose severe clinical and financial burdens on our society. Despite considerable achievements in the field of regenerative medicine in the past several decades, effective treatments remain a challenge due to the limited natural healing capacity of tendons caused by poor cell density and vascularization. The development of tissue engineering has provided more promising results in regenerating tendon-like tissues with compositional, structural and functional characteristics comparable to those of native tendon tissues. Tissue engineering is the discipline of regenerative medicine that aims to restore the physiological functions of tissues by using a combination of cells and materials, as well as suitable biochemical and physicochemical factors. In this review, following a discussion of tendon structure, injury and healing, we aim to elucidate the current strategies (biomaterials, scaffold fabrication techniques, cells, biological adjuncts, mechanical loading and bioreactors, and the role of macrophage polarization in tendon regeneration), challenges and future directions in the field of tendon tissue engineering.
Collapse
Affiliation(s)
- Chao Ning
- Chinese PLA Medical School, Beijing, China
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Pinxue Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Cangjian Gao
- Chinese PLA Medical School, Beijing, China
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Liwei Fu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Zhiyao Liao
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Guangzhao Tian
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Han Yin
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Muzhe Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xiang Sui
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Zhiguo Yuan
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuyun Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Quanyi Guo
- Chinese PLA Medical School, Beijing, China
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
39
|
Wang H, Zhang H, Liu L, Ma K, Huang J, Zhang J. Design and experimental study on closed-loop process of preparing chitosan from crab shells. Biotechnol Appl Biochem 2023. [PMID: 36807387 DOI: 10.1002/bab.2450] [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: 10/19/2022] [Revised: 01/31/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023]
Abstract
The purpose of this article is to design a green and comprehensive utilization process for preparing chitosan from crab shells. Glutamate acid was used as a decalcifying agent for crab shells, and the mixed solution of potassium hydroxide/isopropanol was used for deproteinization and deacetylation to prepare chitosan. Glutamic acid and isopropanol could be recovered for recycling. At the same time, calcium carbonate and protein in crab shells were converted into calcium hydrogen phosphate and compound fertilizer containing nitrogen, phosphorus, and potassium, respectively. The prepared chitosan was characterized by Fourier-transform infrared (FT-IR), differential scanning calorimetry (DSC), x-ray diffraction (XRD), and scanning electron microscopy (SEM), and its deacetylation degree and viscosity average molecular weight were 88.7% ± 0.68% and 792.1 ± 10.82 kDa, respectively. The recoveries of glutamic acid and isopropanol were 95.56% ± 1.39% and 88.14% ± 1.13%, respectively. The prepared chitosan has large molecular weight and deacetylation degree, controllable production cost, comprehensive utilization of crab shell components, and greatly reduced waste emissions.
Collapse
Affiliation(s)
- Huiming Wang
- College of Biology and Environment, Zhejiang Wanli University, Ningbo, Zhejiang, China
| | - Huien Zhang
- College of Biology and Environment, Zhejiang Wanli University, Ningbo, Zhejiang, China
| | - Liping Liu
- College of Biology and Environment, Zhejiang Wanli University, Ningbo, Zhejiang, China
| | - Kunqin Ma
- College of Biology and Environment, Zhejiang Wanli University, Ningbo, Zhejiang, China
| | - Jinqin Huang
- College of Biology and Environment, Zhejiang Wanli University, Ningbo, Zhejiang, China
| | - Jian Zhang
- College of Biology and Environment, Zhejiang Wanli University, Ningbo, Zhejiang, China
| |
Collapse
|
40
|
Bian N, Chu C, Rung S, Huangphattarakul V, Man Y, Lin J, Hu C. Immunomodulatory Biomaterials and Emerging Analytical Techniques for Probing the Immune Micro-Environment. Tissue Eng Regen Med 2023; 20:11-24. [PMID: 36241939 PMCID: PMC9852373 DOI: 10.1007/s13770-022-00491-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 02/01/2023] Open
Abstract
After implantation of a biomaterial, both the host immune system and properties of the material determine the local immune response. Through triggering or modulating the local immune response, materials can be designed towards a desired direction of promoting tissue repair or regeneration. High-throughput sequencing technologies such as single-cell RNA sequencing (scRNA-seq) emerging as a powerful tool for dissecting the immune micro-environment around biomaterials, have not been fully utilized in the field of soft tissue regeneration. In this review, we first discussed the procedures of foreign body reaction in brief. Then, we summarized the influences that physical and chemical modulation of biomaterials have on cell behaviors in the micro-environment. Finally, we discussed the application of scRNA-seq in probing the scaffold immune micro-environment and provided some reference to designing immunomodulatory biomaterials. The foreign body response consists of a series of biological reactions. Immunomodulatory materials regulate immune cell activation and polarization, mediate divergent local immune micro-environments and possess different tissue engineering functions. The manipulation of physical and chemical properties of scaffolds can modulate local immune responses, resulting in different outcomes of fibrosis or tissue regeneration. With the advancement of technology, emerging techniques such as scRNA-seq provide an unprecedented understanding of immune cell heterogeneity and plasticity in a scaffold-induced immune micro-environment at high resolution. The in-depth understanding of the interaction between scaffolds and the host immune system helps to provide clues for the design of biomaterials to optimize regeneration and promote a pro-regenerative local immune micro-environment.
Collapse
Affiliation(s)
- Nanyan Bian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chenyu Chu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Shengan Rung
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Vicha Huangphattarakul
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Jie Lin
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| | - Chen Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
41
|
Collagen Fibril Diameter Distribution of Sheep Anterior Cruciate Ligament. Polymers (Basel) 2023; 15:polym15030752. [PMID: 36772054 PMCID: PMC9920528 DOI: 10.3390/polym15030752] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
The anterior cruciate ligament (ACL) tissue is a soft tissue connecting the femur and tibia at the knee joint and demonstrates a limited capacity for self-regeneration due to its low vascularity. The currently available clinical procedures are unable to fully restore damaged ACL tissue, and tissue engineering can offer options with a potential of restoring the torn/ruptured ACL by using biomimetic constructs that are similar to native tissue in terms of structure, composition, and functions. However, a model substrate to understand how the ACL cells regenerate the injured tissue is still not available. In this study, it is hypothesized that the nanofiber-based model substrate with bimodal and unimodal fiber diameter distributions will mimic the diameter distribution of collagen fibrils seen in healthy and injured sheep ACL, respectively. The aims were to (i) create an ACL injury in a sheep ACL by applying extensional force to rupture the healthy ACL tissue, (ii) measure the collagen fibril diameter distributions of healthy and injured ACL, (iii) fabricate polycaprolactone (PCL) nanofiber-based model constructs using electrospinning with diameter distributions similar to healthy and injured ACL tissue, and (iv) measure mechanical properties of ACL tissue and PCL electrospun constructs. The results showed that the fiber diameter distributions of PCL electrospun constructs and those of the healthy and injured ACL tissues were similar. The novelty in this investigation is that the collagen fibril diameter distribution of healthy and injured sheep ACL tissues was reported for the first time. The study is significant because it aims to create a model construct to solve an important orthopedic-related clinical problem affecting millions of people globally. The model construct fabricated in this work is expected to have an important impact on ACL regeneration efforts.
Collapse
|
42
|
Zou J, Yang W, Cui W, Li C, Ma C, Ji X, Hong J, Qu Z, Chen J, Liu A, Wu H. Therapeutic potential and mechanisms of mesenchymal stem cell-derived exosomes as bioactive materials in tendon-bone healing. J Nanobiotechnology 2023; 21:14. [PMID: 36642728 PMCID: PMC9841717 DOI: 10.1186/s12951-023-01778-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
Tendon-bone insertion (TBI) injuries, such as anterior cruciate ligament injury and rotator cuff injury, are the most common soft tissue injuries. In most situations, surgical tendon/ligament reconstruction is necessary for treating such injuries. However, a significant number of cases failed because healing of the enthesis occurs through scar tissue formation rather than the regeneration of transitional tissue. In recent years, the therapeutic potential of mesenchymal stem cells (MSCs) has been well documented in animal and clinical studies, such as chronic paraplegia, non-ischemic heart failure, and osteoarthritis of the knee. MSCs are multipotent stem cells, which have self-renewability and the ability to differentiate into a wide variety of cells such as chondrocytes, osteoblasts, and adipocytes. Numerous studies have suggested that MSCs could promote angiogenesis and cell proliferation, reduce inflammation, and produce a large number of bioactive molecules involved in the repair. These effects are likely mediated by the paracrine mechanisms of MSCs, particularly through the release of exosomes. Exosomes, nano-sized extracellular vesicles (EVs) with a lipid bilayer and a membrane structure, are naturally released by various cell types. They play an essential role in intercellular communication by transferring bioactive lipids, proteins, and nucleic acids, such as mRNAs and miRNAs, between cells to influence the physiological and pathological processes of recipient cells. Exosomes have been shown to facilitate tissue repair and regeneration. Herein, we discuss the prospective applications of MSC-derived exosomes in TBI injuries. We also review the roles of MSC-EVs and the underlying mechanisms of their effects on promoting tendon-bone healing. At last, we discuss the present challenges and future research directions.
Collapse
Affiliation(s)
- Jiaxuan Zou
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Weinan Yang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Wushi Cui
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Congsun Li
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Chiyuan Ma
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Xiaoxiao Ji
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Jianqiao Hong
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Zihao Qu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China
| | - Jing Chen
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People's Republic of China.
| | - An Liu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China.
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China.
| | - Haobo Wu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, People's Republic of China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310002, People's Republic of China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310002, People's Republic of China.
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310002, People's Republic of China.
| |
Collapse
|
43
|
Micropattern Silk Fibroin Film Facilitates Tendon Repair In Vivo and Promotes Tenogenic Differentiation of Tendon Stem/Progenitor Cells through the α2 β1/FAK/PI3K/AKT Signaling Pathway In Vitro. Stem Cells Int 2023; 2023:2915826. [PMID: 36684388 PMCID: PMC9859702 DOI: 10.1155/2023/2915826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 01/15/2023] Open
Abstract
Background Tendon injuries are common clinical disorders. Due to the limited regeneration ability of tendons, tissue engineering technology is often used as an adjuvant treatment. This study explored the molecular pathways underlying micropattern SF film-regulated TSPC propensity and their repairing effects to highlight the application value of micropattern SF films. Methods First, we characterized the physical properties of the micropattern SF films and explored their repairing effects on the injured tendons in vivo. Then, we seeded TSPCs on SF films in vitro and determined the micropattern SF film-induced gene expression and activation of signaling pathways in TSPCs through high-throughput RNA sequencing and proteomics assays. Results The results of in vivo studies suggested that micropattern SF films can promote remodeling of the injured tendon. In addition, immunohistochemistry (IHC) results showed that tendon marker genes were significantly increased in the micropattern SF film repair group. Transcriptomic and proteomic analyses demonstrated that micropattern SF film-induced genes and proteins in TSPCs were mainly enriched in the focal adhesion kinase (FAK)/actin and phosphoinositide 3-kinase (PI3K)/AKT pathways. Western blot analysis showed that the expression of integrins α2β1, tenascin-C (TNC), and tenomodulin (TNMD) and the phosphorylation of AKT were significantly increased in the micropattern SF film group, which could be abrogated by applying PI3K/AKT inhibitors. Conclusion Micropattern SF films modified by water annealing can promote remodeling of the injured tendon in vivo and regulate the tendon differentiation of TSPCs through the α2β1/FAK/PI3K/AKT signaling pathway in vitro. Therefore, they have great medical value in tendon repair.
Collapse
|
44
|
Biological and Mechanical Factors and Epigenetic Regulation Involved in Tendon Healing. Stem Cells Int 2023; 2023:4387630. [PMID: 36655033 PMCID: PMC9842431 DOI: 10.1155/2023/4387630] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/18/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
Tendons are an important part of the musculoskeletal system. Connecting muscles to bones, tendons convert force into movement. Tendon injury can be acute or chronic. Noticeably, tendon healing requires a long time span and includes inflammation, proliferation, and remodeling processes. The mismatch between endogenous and exogenous healing may lead to adhesion causing further negative effects. Management of tendon injuries and complications such as subsequent adhesion formation are still challenges for clinicians. Due to numerous factors, tendon healing is a complex process. This review introduces the role of various biological and mechanical factors and epigenetic regulation processes involved in tendon healing.
Collapse
|
45
|
Yu L, Zeng L, Zhang Z, Zhu G, Xu Z, Xia J, Weng J, Li J, Pathak JL. Cannabidiol Rescues TNF-α-Inhibited Proliferation, Migration, and Osteogenic/Odontogenic Differentiation of Dental Pulp Stem Cells. Biomolecules 2023; 13:biom13010118. [PMID: 36671503 PMCID: PMC9856031 DOI: 10.3390/biom13010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
Strategies to promote dental pulp stem cells (DPSCs) functions including proliferation, migration, pro-angiogenic effects, and odontogenic/osteogenic differentiation are in urgent need to restore pulpitis-damaged dentin/pulp regeneration and DPSCs-based bone tissue engineering applications. Cannabidiol (CBD), an active component of Cannabis sativa has shown anti-inflammation, chemotactic, anti-microbial, and tissue regenerative potentials. Based on these facts, this study aimed to analyze the effect of CBD on DPSCs proliferation, migration, and osteogenic/odontogenic differentiation in basal and inflammatory conditions. Highly pure DPSCs with characteristics of mesenchymal stem cells (MSCs) were successfully isolated, as indicated by the results of flowcytometry and multi-lineage (osteogenic, adipogenic, and chondrogenic) differentiation potentials. Among the concentration tested (0.1-12.5 µM), CBD (2.5 μM) showed the highest anabolic effect on the proliferation and osteogenic/odontogenic differentiation of DPSCs. Pro-angiogenic growth factor VEGF mRNA expression was robustly higher in CBD-treated DPSCs. CBD also prompted the migration of DPSCs and CBD receptor CB1 and CB2 expression in DPSCs. TNF-α inhibited the viability, migration, and osteogenic/odontogenic differentiation of DPSCs and CBD reversed these effects. CBD alleviated the TNF-α-upregulated expression of pro-inflammatory cytokines TNF-α, interleukin (IL)-1β, and IL-6 in DPSCs. In conclusion, our results indicate the possible application of CBD on DPSCs-based dentin/pulp and bone regeneration.
Collapse
Affiliation(s)
- Lina Yu
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
| | - Liting Zeng
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
| | - Zeyu Zhang
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
| | - Guanxiong Zhu
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
| | - Zidan Xu
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
| | - Junyi Xia
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
| | - Jinlong Weng
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
| | - Jiang Li
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou 510182, China
- Correspondence: (J.L.); (J.L.P.); Tel.: +(020)-8050-0893 (J.L.); +(020)-8192-7729 (J.L.P.)
| | - Janak Lal Pathak
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou 510182, China
- Correspondence: (J.L.); (J.L.P.); Tel.: +(020)-8050-0893 (J.L.); +(020)-8192-7729 (J.L.P.)
| |
Collapse
|
46
|
Xu X, Zhang Y, Ha P, Chen Y, Li C, Yen E, Bai Y, Chen R, Wu BM, Da Lio A, Ting K, Soo C, Zheng Z. A novel injectable fibromodulin-releasing granular hydrogel for tendon healing and functional recovery. Bioeng Transl Med 2023; 8:e10355. [PMID: 36684085 PMCID: PMC9842059 DOI: 10.1002/btm2.10355] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 01/25/2023] Open
Abstract
A crucial component of the musculoskeletal system, the tendon is one of the most commonly injured tissues in the body. In severe cases, the ruptured tendon leads to permanent dysfunction. Although many efforts have been devoted to seeking a safe and efficient treatment for enhancing tendon healing, currently existing treatments have not yet achieved a major clinical improvement. Here, an injectable granular hyaluronic acid (gHA)-hydrogel is engineered to deliver fibromodulin (FMOD)-a bioactive extracellular matrix (ECM) that enhances tenocyte mobility and optimizes the surrounding ECM assembly for tendon healing. The FMOD-releasing granular HA (FMOD/gHA)-hydrogel exhibits unique characteristics that are desired for both patients and health providers, such as permitting a microinvasive application and displaying a burst-to-sustained two-phase release of FMOD, which leads to a prompt FMOD delivery followed by a constant dose-maintaining period. Importantly, the generated FMOD-releasing granular HA hydrogel significantly augmented tendon-healing in a fully-ruptured rat's Achilles tendon model histologically, mechanically, and functionally. Particularly, the breaking strength of the wounded tendon and the gait performance of treated rats returns to the same normal level as the healthy controls. In summary, a novel effective FMOD/gHA-hydrogel is developed in response to the urgent demand for promoting tendon healing.
Collapse
Affiliation(s)
- Xue Xu
- Department of Oral and Maxillofacial Plastic and Traumatic SurgeryBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| | - Yulong Zhang
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Pin Ha
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| | - Yao Chen
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chenshuang Li
- Department of OrthodonticsSchool of Dental Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Emily Yen
- Arcadia High SchoolArcadiaCaliforniaUSA
| | - Yuxing Bai
- Department of OrthodonticsBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
| | - Renji Chen
- Department of Oral and Maxillofacial Plastic and Traumatic SurgeryBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
| | - Benjamin M. Wu
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Andrew Da Lio
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Kang Ting
- Forsyth Research InstituteHarvard UniversityCambridgeMassachusettsUSA
- Samueli School of EngineeringUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic SurgeryThe Orthopaedic Hospital Research Center, University of CaliforniaLos AngelesCaliforniaUSA
| | - Zhong Zheng
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
47
|
Wang L, Yang T, Ding L, Ye X, Wu L. Platelet-derived growth factor AA-modified electrospun fibers promote tendon healing. J Biomater Appl 2023; 37:1018-1028. [PMID: 36411499 DOI: 10.1177/08853282221139274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Platelet-derived growth factor AA (PDGF-AA) is an important promoter of tissue injury repair and might be a candidate for improving the mechanical properties of repaired tendons. Here, we designed a PDGF-AA-modified poly(lactide-co-glycolide) acid (PLGA) electrospun fibers to promote tendon rehabilitation after injury. In the present study, we grafted PDGF-AA on the surface of PLGA. In structural experiments, we found that the hydrophilicity of PLGA containing PDGF-AA (PLGA-PDGF-AA) increased, but the strength of the material did not change significantly. Moreover, no significant changes in tendon cell proliferation and viability were observed in the PLGA-PDGF-AA treatment compared with the control group. The mouse tendon injury model (n = 9) experiment illustrated that PLGA-PDGF-AA effectively promoted tendon healing, and we confirmed that PLGA-PDGF-AA promoted collagen synthesis and deposition by immunohistochemistry and RT-PCR. Moreover, the mechanical strength of PLGA-PDGF-AA-treated mouse (n = 9) tendon tissue was also higher than that of the PLGA-treated group alone. In conclusion, PLGA-PDGF-AA promoted regeneration after tendon injury and serves as a potential adjuvant material for surgical tendon injury repair.
Collapse
Affiliation(s)
- Linyou Wang
- Department of Radiology, 590733Taizhou Municipal Hospital, Taizhou City, Zhejiang Province, China
| | - Tiejun Yang
- Department of Radiology, 590733Taizhou Municipal Hospital, Taizhou City, Zhejiang Province, China
| | - Li Ding
- Department of Radiology, 590733Taizhou Municipal Hospital, Taizhou City, Zhejiang Province, China
| | - Xiao Ye
- Department of Radiology, 590733Taizhou Municipal Hospital, Taizhou City, Zhejiang Province, China
| | - Liang Wu
- Department of Radiology, 590733Taizhou Municipal Hospital, Taizhou City, Zhejiang Province, China
| |
Collapse
|
48
|
Amini M, Venkatesan JK, Liu W, Leroux A, Nguyen TN, Madry H, Migonney V, Cucchiarini M. Advanced Gene Therapy Strategies for the Repair of ACL Injuries. Int J Mol Sci 2022; 23:ijms232214467. [PMID: 36430947 PMCID: PMC9695211 DOI: 10.3390/ijms232214467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
The anterior cruciate ligament (ACL), the principal ligament for stabilization of the knee, is highly predisposed to injury in the human population. As a result of its poor intrinsic healing capacities, surgical intervention is generally necessary to repair ACL lesions, yet the outcomes are never fully satisfactory in terms of long-lasting, complete, and safe repair. Gene therapy, based on the transfer of therapeutic genetic sequences via a gene vector, is a potent tool to durably and adeptly enhance the processes of ACL repair and has been reported for its workability in various experimental models relevant to ACL injuries in vitro, in situ, and in vivo. As critical hurdles to the effective and safe translation of gene therapy for clinical applications still remain, including physiological barriers and host immune responses, biomaterial-guided gene therapy inspired by drug delivery systems has been further developed to protect and improve the classical procedures of gene transfer in the future treatment of ACL injuries in patients, as critically presented here.
Collapse
Affiliation(s)
- Mahnaz Amini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Amélie Leroux
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Tuan Ngoc Nguyen
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Véronique Migonney
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
- Correspondence: or
| |
Collapse
|
49
|
Janvier AJ, Pendleton EG, Mortensen LJ, Green DC, Henstock JR, Canty-Laird EG. Multimodal analysis of the differential effects of cyclic strain on collagen isoform composition, fibril architecture and biomechanics of tissue engineered tendon. J Tissue Eng 2022; 13:20417314221130486. [PMID: 36339372 PMCID: PMC9629721 DOI: 10.1177/20417314221130486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/18/2022] [Indexed: 11/07/2022] Open
Abstract
Tendon is predominantly composed of aligned type I collagen, but additional isoforms are known to influence fibril architecture and maturation, which contribute to the tendon’s overall biomechanical performance. The role of the less well-studied collagen isoforms on fibrillogenesis in tissue engineered tendons is currently unknown, and correlating their relative abundance with biomechanical changes in response to cyclic strain is a promising method for characterising optimised bioengineered tendon grafts. In this study, human mesenchymal stem cells (MSCs) were cultured in a fibrin scaffold with 3%, 5% or 10% cyclic strain at 0.5 Hz for 3 weeks, and a comprehensive multimodal analysis comprising qPCR, western blotting, histology, mechanical testing, fluorescent probe CLSM, TEM and label-free second-harmonic imaging was performed. Molecular data indicated complex transcriptional and translational regulation of collagen isoforms I, II, III, V XI, XII and XIV in response to cyclic strain. Isoforms (XII and XIV) associated with embryonic tenogenesis were deposited in the formation of neo-tendons from hMSCs, suggesting that these engineered tendons form through some recapitulation of a developmental pathway. Tendons cultured with 3% strain had the smallest median fibril diameter but highest resistance to stress, whilst at 10% strain tendons had the highest median fibril diameter and the highest rate of stress relaxation. Second harmonic generation exposed distinct structural arrangements of collagen fibres in each strain group. Fluorescent probe images correlated increasing cyclic strain with increased fibril alignment from 40% (static strain) to 61.5% alignment (10% cyclic strain). These results indicate that cyclic strain rates stimulate differential cell responses via complex regulation of collagen isoforms which influence the structural organisation of developing fibril architectures.
Collapse
Affiliation(s)
- Adam J Janvier
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Emily G Pendleton
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| | - Luke J Mortensen
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| | - Daniel C Green
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Liverpool, UK
| | - James R Henstock
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Liverpool, UK,Elizabeth G Canty-Laird, Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - Elizabeth G Canty-Laird
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK,Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| |
Collapse
|
50
|
Quantitative Analysis of Contrast-Enhanced Ultrasound That Can Be Used to Evaluate Angiogenesis during Patellar Tendon Healing in Rats. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:6867743. [PMID: 36313964 PMCID: PMC9584743 DOI: 10.1155/2022/6867743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/15/2022] [Indexed: 01/26/2023]
Abstract
Objective To investigate the efficacy of contrast-enhanced ultrasound (CEUS) in quantitatively evaluating angiogenesis during patellar tendon healing in rats. Methods A total of 40 Sprague-Dawley rats were used in this study. The patellar tendons of 30 rats (60 limbs) that underwent incision and suture were treated as the operation group and monitored after 7, 14, and 28 days. The normal patellar tendons of 10 rats (20 limbs) were treated as the control group and monitored on day 0. The ultrasound examination was used to evaluate the structure and blood perfusion of the patellar tendon. Immunohistochemistry was used to assess angiogenesis, and the biomechanical test was used to verify functional recovery of the patellar tendon. Results The tendons in the operation group were significantly thickened compared with those in the control group (p < 0.01). The peak intensity (PI) in CEUS of the tendons showed a clear difference at each time point after the surgery (p < 0.01). PI increased in the operation group with a maximum on day 7, and then gradually decreased until day 28 when PI was close to the basic intensity (BI) in the control group (p > 0.05). It was consistent with the change of the CD31-positive staining areas representing angiogenesis of the injured patellar tendons. The PI was positively correlated with the CD31-positive staining area fraction (R = 0.849, p < 0.001). The failure load and tensile strength of the repaired patellar tendons in the operation group increased over time. The PI showed negative correlations with the failure load (R = -0.787, p < 0.001) and tensile strength (R = -0.714, p < 0.001). Conclusion The PI in CEUS could quantitatively reflect the time-dependent change in the blood supply of the healing site, and the PI correlated with histologic and biomechanical properties of the healing tendon. Quantitative analysis of contrast-enhanced ultrasound could be a useful method to evaluate angiogenesis in healing tendons.
Collapse
|