1
|
Tashjian RZ, Zitnay J, Kazmers NH, Veerabhadraiah SR, Zelada AC, Honeggar M, Smith MC, Chalmers PN, Henninger HB, Jurynec MJ. Tenascin C deletion impairs tendon healing and functional recovery after rotator cuff repair. J Orthop Res 2025; 43:483-491. [PMID: 39601211 PMCID: PMC11806989 DOI: 10.1002/jor.26025] [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: 04/15/2024] [Revised: 09/22/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024]
Abstract
The biological factors that affect healing after rotator cuff repair (RCR) are not well understood. Genetic variants in the extracellular matrix protein Tenascin C (TNC) are associated with impaired tendon healing and it is expressed in rotator cuff tendon tissue after injury, suggesting it may have a role in the repair process. The purpose of the current study was to determine the role of TNC on tendon healing after RCR in a murine model. The supraspinatus tendon was transected and repaired on the left shoulder of wild-type (WT-RCR), Tenascin C null (Tnc--RCR) and Tnc heterozygous (Tnc+/--RCR) mice. Controls included the unoperated, contralateral shoulder of WT-RCR, Tnc-RCR, Tnc+/--RCR mice and unoperated shoulders from age and genotype matched controls. We performed histologic, activity testing, bulk RNA-seq, and biomechanical analyses. At 8-weeks post-RCR, Tnc- and Tnc+/- mice had severe bone and tendon defects following RCR. Tnc--RCR mice had reduced activity after RCR including reduced wheel rotations, wheel duration, and wheel episode average velocity compared with WT-RCR. Loss of Tnc following RCR altered gene expression in the shoulder, including upregulation of sex hormone and WNT pathways and a downregulation of inflammation and cell cycle pathways. Tnc- mice had similar biomechanical properties after repair as WT. Further research is required to evaluate tissue specific alterations of Tnc, the interactions of Tnc and sex hormone and inflammation pathways as well as possible adjuvants to improve enthesis healing in the setting of reduced TNC function.
Collapse
Affiliation(s)
- Robert Z. Tashjian
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Jared Zitnay
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Nikolas H. Kazmers
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | | | - Antonio C. Zelada
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Matthew Honeggar
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Matthew C. Smith
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Peter N. Chalmers
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Heath B. Henninger
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Michael J. Jurynec
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
2
|
Yu X, Wu G, Cai P, Ding Y, Cui J, Wu J, Shen Y, Song J, Yuan Z, El-Newehy M, Abdulhameed MM, Chen H, Mo X, Sun B, Yu Y. Carbon Fiber-Mediated Electrospinning Scaffolds Can Conduct Electricity for Repairing Defective Tendon. ACS APPLIED MATERIALS & INTERFACES 2024; 16:52104-52115. [PMID: 39288100 DOI: 10.1021/acsami.4c12245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Partial or complete rupture of the tendon can damage the collagen structure, resulting in the disruption of the electrical signal pathway. It is a great challenge to reconstruct the original electrical signal pathway of the tendon and promote the regeneration and functional recovery of defective tendon. In this study, carbon fiber-mediated electrospinning scaffolds were fabricated by wrapping conductive, high-strength, loose single-bundle carbon fibers with nanofiber membranes. Due to the presence of nanofiber membranes, the maximum tensile force of the scaffolds was 2.4 times higher than that of carbon fibers, while providing excellent temporal and spatial prerequisites for tenocytes to adapt to electrical stimulation to accelerate proliferation and expression. The diameter of the carbon fiber monofilaments used in this study was 5.07 ± 1.20 μm, which matched the diameter of tendon collagen, allowing for quickly establishing the connection between the tendon tissue and the scaffold, and better promoting the recovery of the electrical signal pathway. In a rabbit Achilles tendon defect repair model, the carbon fiber-mediated electrospinning scaffold was almost filled with collagen fibers compared to a nonconductive polyethylene glycol terephthalate scaffold. Transcriptome sequencing revealed that fibromodulin and tenomodulin expression were upregulated, and their related proteoglycans and glycosaminoglycan binding proteins pathways were enhanced, which could regulate the TGF-β signaling pathway and optimize the extracellular matrix assembly, thus promoting tendon repair. Therefore, the scaffold in this study makes up for the shortage of conductive scaffolds for repairing tendon defects, revealing the potential impact of conductivity on the signaling pathway of tendon repair and providing a new approach for future clinical studies.
Collapse
Affiliation(s)
- Xiao Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Genbin Wu
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Pengfei Cai
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yangfan Ding
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Jie Cui
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Jinglei Wu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yihong Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Jiahui Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Zhengchao Yuan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Meera Moydeen Abdulhameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Huifang Chen
- College of Materials Science and Engineering and State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai 201620, China
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Binbin Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yinxian Yu
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, 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
|
Tashjian RZ, Zitnay J, Kazmers NH, Veerabhadraiah SR, Zelada AC, Honeggar M, Smith MC, Chalmers PN, Henninger HB, Jurynec MJ. Tenascin C Deletion Impairs Tendon Healing and Functional Recovery After Rotator Cuff Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612543. [PMID: 39314362 PMCID: PMC11419033 DOI: 10.1101/2024.09.11.612543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The biological factors that affect healing after rotator cuff repair (RCR) are not well understood. Genetic variants in the extracellular matrix protein Tenascin C (TNC) are associated with impaired tendon healing and it is expressed in rotator cuff tendon tissue after injury, suggesting it may have a role in the repair process. The purpose of the current study was to determine the role of TNC on tendon healing after RCR in a murine model. The supraspinatus tendon was transected and repaired on the left shoulder of Wild-Type (WT-RCR), Tenascin C null (Tnc --RCR) and Tnc heterozygous (Tnc +/--RCR) mice. Controls included the unoperated, contralateral shoulder of WT-RCR, Tnc - RCR, Tnc +/--RCR mice and unoperated shoulders from age and genotype matched controls. We performed histologic, activity testing, RNA-seq, and biomechanical analyses. At 8-weeks post-RCR, Tnc - and Tnc +/- mice had severe bone and tendon defects following rotator cuff repair. Tnc --RCR mice had reduced activity after rotator cuff repair including reduced wheel rotations, wheel duration, and wheel episode average velocity compared with WT-RCR. Loss of Tnc following RCR altered gene expression in the shoulder, including upregulation of sex hormone and WNT pathways and a downregulation of inflammation and cell cycle pathways. Tnc - mice had similar biomechanical properties after repair as WT. Further research is required to evaluate tissue specific alterations of Tnc, the interactions of Tnc and sex hormone and inflammation pathways as well as possible adjuvants to improve enthesis healing in the setting of reduced TNC function.
Collapse
Affiliation(s)
- Robert Z. Tashjian
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Jared Zitnay
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Nikolas H. Kazmers
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | | | - Antonio C. Zelada
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Matthew Honeggar
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Matthew C. Smith
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Peter N. Chalmers
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Heath B. Henninger
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
| | - Michael J. Jurynec
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah 84108 USA
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
5
|
Wang Y, Qin Q, Wang Z, Negri S, Sono T, Tower RJ, Li Z, Xing X, Archer M, Thottappillil N, Zhu M, Suarez A, Kim DH, Harvey T, Fan CM, James AW. The Mohawk homeobox gene represents a marker and osteo-inhibitory factor in calvarial suture osteoprogenitor cells. Cell Death Dis 2024; 15:420. [PMID: 38886383 PMCID: PMC11183145 DOI: 10.1038/s41419-024-06813-4] [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: 02/03/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
The regeneration of the mammalian skeleton's craniofacial bones necessitates the action of intrinsic and extrinsic inductive factors from multiple cell types, which function hierarchically and temporally to control the differentiation of osteogenic progenitors. Single-cell transcriptomics of developing mouse calvarial suture recently identified a suture mesenchymal progenitor population with previously unappreciated tendon- or ligament-associated gene expression profile. Here, we developed a Mohawk homeobox (MkxCG; R26RtdT) reporter mouse and demonstrated that this reporter identifies an adult calvarial suture resident cell population that gives rise to calvarial osteoblasts and osteocytes during homeostatic conditions. Single-cell RNA sequencing (scRNA-Seq) data reveal that Mkx+ suture cells display a progenitor-like phenotype with expression of teno-ligamentous genes. Bone injury with Mkx+ cell ablation showed delayed bone healing. Remarkably, Mkx gene played a critical role as an osteo-inhibitory factor in calvarial suture cells, as knockdown or knockout resulted in increased osteogenic differentiation. Localized deletion of Mkx in vivo also resulted in robustly increased calvarial defect repair. We further showed that mechanical stretch dynamically regulates Mkx expression, in turn regulating calvarial cell osteogenesis. Together, we define Mkx+ cells within the suture mesenchyme as a progenitor population for adult craniofacial bone repair, and Mkx acts as a mechanoresponsive gene to prevent osteogenic differentiation within the stem cell niche.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Ziyi Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Orthopaedic and Trauma Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology of the University of Verona, 37134, Verona, Italy
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Robert J Tower
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Manyu Zhu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Allister Suarez
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Tyler Harvey
- Department of Embryology, Carnegie Institution of Washington, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution of Washington, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
6
|
Shi G, Koichi N, Wan R, Wang Y, Reisdorf R, Wilson A, Huang TC, Amadio PC, Meves A, Zhao C, Moran SL. Pentamidine-loaded gelatin decreases adhesion formation of flexor tendon. J Orthop Translat 2024; 45:75-87. [PMID: 38511123 PMCID: PMC10950576 DOI: 10.1016/j.jot.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/14/2023] [Accepted: 10/26/2023] [Indexed: 03/22/2024] Open
Abstract
Background Prevention of adhesion formation following flexor tendon repair is essential for restoration of normal finger function. Although many medications have been studied in the experimental setting to prevent adhesions, clinical application is limited due to the complexity of application and delivery in clinical translation. Methods In this study, optimal dosages of gelatin and pentamidine were validated by gelatin concentration test. Following cell viability, cell migration, live and dead cell, and cell adhesion assay of the Turkey tenocytes, a model of Turkey tendon repair was established to evaluate the effectiveness of the Pentamidine-Gelatin sheet. Results Pentamidine carried with gelatin, a Food and drug administration (FDA) approved material for drug delivery, showed good dynamic release, biocompatibility, and degradation. The optimal dose of pentamidine (25ug) was determined in the in vivo study using tenocyte viability, migration, and cell adhesion assays. Further biochemical analyses demonstrated that this positive effect may be due to pentamidine downregulating the Wnt signaling pathway without affecting collagen expression. Conclusions We tested a FDA-approved antibiotic, pentamidine, for reducing adhesion formation after flexor tendon repair in both in vitro and in vivo using a novel turkey animal model. Compared with the non-pentamidine treatment group, pentamidine treated turkeys had significantly reduced adhesions and improved digit function after six weeks of tendon healing. The translational potential of this article This study for the first time showed that a common clinical drug, pentamidine, has a potential for clinical application to reduce tendon adhesions and improve tendon gliding function without interfering with tendon healing.
Collapse
Affiliation(s)
- Guidong Shi
- Department of Orthopaedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Nakagawa Koichi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Rou Wan
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Yicun Wang
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ramona Reisdorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Abigayle Wilson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Tony C.T. Huang
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Peter C. Amadio
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Steven L. Moran
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
7
|
Liu Z, Han W, Meng J, Pi Y, Wu T, Fan Y, Guo Q, Hu X, Chen Y, Jiang W, Zhao F. Mohawk protects against tendon damage via suppressing Wnt/β-catenin pathway. Heliyon 2024; 10:e25658. [PMID: 38370202 PMCID: PMC10867664 DOI: 10.1016/j.heliyon.2024.e25658] [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: 12/31/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024] Open
Abstract
Degenerative tendon injuries are common clinical problems associated with overuse or aging, and understanding the mechanisms of tendon injury and regeneration can contribute to the study of tendon healing and repair. As a transcription factor, Mohawk (Mkx) is responsible for tendons development, yet, the roles of which in tendon damage remain mostly elusive. In this study, using Mkx overexpressed mice on long treadmill as an in vivo model and MkxOE Achilles tenocytes stimulated by equiaxial stretch as an in vitro model, we anaylsed the effects of Mkx overexpression on the tendon. Mkx and tendon tension strength were decreased after the expose to excessive mechanical forces, and Mkx overexpression protected the tendon from damage. Moreover, we revealed that the Wnt/β-catenin activation, inflammation, and Runx2 expression were increased at the injured Achilles tendon, upregulated Mkx significantly reversed the increased Wnt/β-catenin pathway, Tnf-α, Il-1β, and Il-6 levels, and reduced tendon cell damage. However, Wnt3a, IWR and BIO had not significantly affected the Mkx expression in achilles tenocytes. In conclusion, Mkx is involved in tendon healing and protects the tendon from damage through suppressing Wnt/β-catenin pathway, suggesting Mkx/Wnt/β-catenin pathway may be potential therapeutic targets for tendon damage.
Collapse
Affiliation(s)
- Ziming Liu
- Department of Sports Medicine, Sports Medicine Institute, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Wenfeng Han
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Jiao Meng
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou, China
| | - Yanbing Pi
- Department of Sports Medicine, Sports Medicine Institute, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Tong Wu
- Department of Sports Medicine, Sports Medicine Institute, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Yifei Fan
- Department of Sports Medicine, Sports Medicine Institute, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Qinwei Guo
- Department of Sports Medicine, Sports Medicine Institute, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Sports Medicine Institute, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Yuhua Chen
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou, China
| | - Wenxiao Jiang
- Department of Sports Medicine, Qilu Hospital of Shandong University (Qingdao Campus), Qingdao, Shandong, China
| | - Feng Zhao
- Department of Sports Medicine, Sports Medicine Institute, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Peking University Third Hospital, Beijing, China
| |
Collapse
|
8
|
Cherief M, Xu J, Li Z, Tower RJ, Ramesh S, Qin Q, Gomez-Salazar M, Yea JH, Lee S, Negri S, Xu M, Price T, Kendal AR, Fan CM, Clemens TL, Levi B, James AW. TrkA-mediated sensory innervation of injured mouse tendon supports tendon sheath progenitor cell expansion and tendon repair. Sci Transl Med 2023; 15:eade4619. [PMID: 38117901 DOI: 10.1126/scitranslmed.ade4619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
Peripheral neurons terminate at the surface of tendons partly to relay nociceptive pain signals; however, the role of peripheral nerves in tendon injury and repair remains unclear. Here, we show that after Achilles tendon injury in mice, there is new nerve growth near tendon cells that express nerve growth factor (NGF). Conditional deletion of the Ngf gene in either myeloid or mesenchymal mouse cells limited both innervation and tendon repair. Similarly, inhibition of the NGF receptor tropomyosin receptor kinase A (TrkA) abrogated tendon healing in mouse tendon injury. Sural nerve transection blocked the postinjury increase in tendon sensory innervation and the expansion of tendon sheath progenitor cells (TSPCs) expressing tubulin polymerization promoting protein family member 3. Single cell and spatial transcriptomics revealed that disruption of sensory innervation resulted in dysregulated inflammatory signaling and transforming growth factor-β (TGFβ) signaling in injured mouse tendon. Culture of mouse TSPCs with conditioned medium from dorsal root ganglia neuron further supported a role for neuronal mediators and TGFβ signaling in TSPC proliferation. Transcriptomic and histologic analyses of injured human tendon biopsy samples supported a role for innervation and TGFβ signaling in human tendon regeneration. Last, treating mice after tendon injury systemically with a small-molecule partial agonist of TrkA increased neurovascular response, TGFβ signaling, TSPC expansion, and tendon tissue repair. Although further studies should investigate the potential effects of denervation on mechanical loading of tendon, our results suggest that peripheral innervation is critical for the regenerative response after acute tendon injury.
Collapse
Affiliation(s)
- Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert J Tower
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Sowmya Ramesh
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Ji-Hye Yea
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Orthopaedics and Traumatology, University of Verona, Verona 37129, Italy
| | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Theodore Price
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, TX 75080, USA
| | - Adrian R Kendal
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford OX3 7LD, UK
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21208, USA
| | - Thomas L Clemens
- Department of Orthopaedics, University of Maryland, Baltimore, MD 21205, USA
- Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
9
|
Pancheri NM, Daw JT, Ditton D, Schiele NR, Birks S, Uzer G, Jones CL, Penney BT, Theodossiou SK. The LINC complex regulates Achilles tendon elastic modulus, Achilles and tail tendon collagen crimp, and Achilles and tail tendon lateral expansion during early postnatal development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566892. [PMID: 38014288 PMCID: PMC10680625 DOI: 10.1101/2023.11.13.566892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
There is limited understanding of how mechanical signals regulate tendon development. The nucleus has emerged as a major regulator of cellular mechanosensation, via the linker of nucleoskeleton and cytoskeleton (LINC) protein complex. Specific roles of LINC in tenogenesis have not been explored. In this study, we investigate how LINC regulates tendon development by disabling LINC-mediated mechanosensing via dominant negative (dn) expression of the Klarsicht, ANC-1, and Syne Homology (KASH) domain, which is necessary for LINC to function. We hypothesized that LINC regulates mechanotransduction in developing tendon, and that disabling LINC would impact tendon mechanical properties and structure in a mouse model of dnKASH. We used Achilles (AT) and tail (TT) tendons as representative energy-storing and limb-positioning tendons, respectively. Mechanical testing at postnatal day 10 showed that disabling the LINC complex via dnKASH significantly impacted tendon mechanical properties and cross-sectional area, and that effects differed between ATs and TTs. Collagen crimp distance was also impacted in dnKASH tendons, and was significantly decreased in ATs, and increased in TTs. Overall, we show that disruption to the LINC complex specifically impacts tendon mechanics and collagen crimp structure, with unique responses between an energy-storing and limb-positioning tendon. This suggests that nuclear mechanotransduction through LINC plays a role in regulating tendon formation during neonatal development.
Collapse
|
10
|
Lu J, Chen H, Lyu K, Jiang L, Chen Y, Long L, Wang X, Shi H, Li S. The Functions and Mechanisms of Tendon Stem/Progenitor Cells in Tendon Healing. Stem Cells Int 2023; 2023:1258024. [PMID: 37731626 PMCID: PMC10509002 DOI: 10.1155/2023/1258024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Tendon injury is one of the prevalent disorders of the musculoskeletal system in orthopedics and is characterized by pain and limitation of joint function. Due to the difficulty of spontaneous tendon healing, and the scar tissue and low mechanical properties that usually develops after healing. Therefore, the healing of tendon injury remains a clinical challenge. Although there are a multitude of approaches to treating tendon injury, the therapeutic effects have not been satisfactory to date. Recent studies have shown that stem cell therapy has a facilitative effect on tendon healing. In particular, tendon stem/progenitor cells (TSPCs), a type of stem cell from tendon tissue, play an important role not only in tendon development and tendon homeostasis, but also in tendon healing. Compared to other stem cells, TSPCs have the potential to spontaneously differentiate into tenocytes and express higher levels of tendon-related genes. TSPCs promote tendon healing by three mechanisms: modulating the inflammatory response, promoting tenocyte proliferation, and accelerating collagen production and balancing extracellular matrix remodeling. However, current investigations have shown that TSPCs also have a negative effect on tendon healing. For example, misdifferentiation of TSPCs leads to a "failed healing response," which in turn leads to the development of chronic tendon injury (tendinopathy). The focus of this paper is to describe the characteristics of TSPCs and tenocytes, to demonstrate the roles of TSPCs in tendon healing, while discussing the approaches used to culture and differentiate TSPCs. In addition, the limitations of TSPCs in clinical application and their potential therapeutic strategies are elucidated.
Collapse
Affiliation(s)
- Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Hui Chen
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Longhai Long
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoqiang Wang
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Houyin Shi
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
11
|
Yuan X, Zhu Z, Xia P, Wang Z, Zhao X, Jiang X, Wang T, Gao Q, Xu J, Shan D, Guo B, Yao Q, He Y. Tough Gelatin Hydrogel for Tissue Engineering. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301665. [PMID: 37353916 PMCID: PMC10460895 DOI: 10.1002/advs.202301665] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/18/2023] [Indexed: 06/25/2023]
Abstract
Tough hydrogel has attracted considerable interest in various fields, however, due to poor biocompatibility, nondegradation, and pronounced compositional differences from natural tissues, it is difficult to be used for tissue regeneration. Here, a gelatin-based tough hydrogel (GBTH) is proposed to fill this gap. Inspired by human exercise to improve muscle strength, the synergistic effect is utilized to generate highly functional crystalline domains for resisting crack propagation. The GBTH exhibits excellent tensile strength of 6.67 MPa (145-fold that after untreated gelation). Furthermore, it is directly sutured to a ruptured tendon of adult rabbits due to its pronounced toughness and biocompatibility, self-degradability in vivo, and similarity to natural tissue components. Ruptured tendons can compensate for mechanotransduction by GBTH and stimulate tendon differentiation to quickly return to the initial state, that is, within eight weeks. This strategy provides a new avenue for preparation of highly biocompatible tough hydrogel for tissue regeneration.
Collapse
Affiliation(s)
- Ximin Yuan
- State Key Laboratory of Advanced Welding and JoiningHarbin Institute of TechnologyHarbin150001P. R. China
- National Innovation Center for Advanced Medical DevicesShenzhen457001P. R. China
| | - Zhou Zhu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologyChengdu610041P. R. China
| | - Pengcheng Xia
- Institute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006P. R. China
| | - Zhenjia Wang
- State Key Laboratory of Advanced Welding and JoiningHarbin Institute of TechnologyHarbin150001P. R. China
- National Innovation Center for Advanced Medical DevicesShenzhen457001P. R. China
| | - Xiao Zhao
- Institute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006P. R. China
| | - Xiao Jiang
- Institute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006P. R. China
| | - Tianming Wang
- Institute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006P. R. China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic SystemsSchool of Mechanical EngineeringZhejiang UniversityHangzhou310027P. R. China
| | - Jie Xu
- State Key Laboratory of Advanced Welding and JoiningHarbin Institute of TechnologyHarbin150001P. R. China
- National Innovation Center for Advanced Medical DevicesShenzhen457001P. R. China
| | - Debin Shan
- State Key Laboratory of Advanced Welding and JoiningHarbin Institute of TechnologyHarbin150001P. R. China
- National Innovation Center for Advanced Medical DevicesShenzhen457001P. R. China
| | - Bin Guo
- State Key Laboratory of Advanced Welding and JoiningHarbin Institute of TechnologyHarbin150001P. R. China
- National Innovation Center for Advanced Medical DevicesShenzhen457001P. R. China
| | - Qingqiang Yao
- Institute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006P. R. China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic SystemsSchool of Mechanical EngineeringZhejiang UniversityHangzhou310027P. R. China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang ProvinceCollege of Mechanical EngineeringZhejiang UniversityHangzhou310027P. R. China
- Cancer CenterZhejiang UniversityHangzhou310058P. R. China
| |
Collapse
|
12
|
Zhu M, Metzen F, Hopkinson M, Betz J, Heilig J, Sodhi J, Imhof T, Niehoff A, Birk DE, Izu Y, Krüger M, Pitsillides AA, Altmüller J, van Osch GJ, Straub V, Schreiber G, Paulsson M, Koch M, Brachvogel B. Ablation of collagen XII disturbs joint extracellular matrix organization and causes patellar subluxation. iScience 2023; 26:107225. [PMID: 37485359 PMCID: PMC10362267 DOI: 10.1016/j.isci.2023.107225] [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: 08/04/2022] [Revised: 05/05/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Collagen XII, belonging to the fibril-associated collagens, is a homotrimeric secreted extracellular matrix (ECM) protein encoded by the COL12A1 gene. Mutations in the human COL12A1 gene cause an Ehlers-Danlos/myopathy overlap syndrome leading to skeletal abnormalities and muscle weakness. Here, we studied the role of collagen XII in joint pathophysiology by analyzing collagen XII deficient mice and human patients. We found that collagen XII is widely expressed across multiple connective tissue of the developing joint. Lack of collagen XII in mice destabilizes tendons and the femoral trochlear groove to induce patellar subluxation in the patellofemoral joint. These changes are associated with an ECM damage response in tendon and secondary quadriceps muscle degeneration. Moreover, patellar subluxation was also identified as a clinical feature of human patients with collagen XII deficiency. The results provide an explanation for joint hyperlaxity in mice and human patients with collagen XII deficiency.
Collapse
Affiliation(s)
- Mengjie Zhu
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Fabian Metzen
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Mark Hopkinson
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, UK
| | - Janina Betz
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Juliane Heilig
- Institute of Biomechanics & Orthopaedics, German Sport University Cologne, Cologne, Germany
- Center for Musculoskeletal Biomechanics (CCMB), Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jassi Sodhi
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | - Thomas Imhof
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics & Orthopaedics, German Sport University Cologne, Cologne, Germany
- Center for Musculoskeletal Biomechanics (CCMB), Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - David E. Birk
- College of Medicine, University of South Florida, Morsani, Tampa, FL, USA
| | - Yayoi Izu
- Department of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Marcus Krüger
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Andrew A. Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, UK
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
- Berlin Institute of Health at Charité, Core Facility Genomics, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center, CN Rotterdam, the Netherlands
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | | | - Mats Paulsson
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Bent Brachvogel
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Gonzalez RD, Small GW, Green AJ, Akhtari FS, Motsinger-Reif AA, Quintanilha JCF, Havener TM, Reif DM, McLeod HL, Wiltshire T. MKX-AS1 Gene Expression Associated with Variation in Drug Response to Oxaliplatin and Clinical Outcomes in Colorectal Cancer Patients. Pharmaceuticals (Basel) 2023; 16:ph16050757. [PMID: 37242540 DOI: 10.3390/ph16050757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Oxaliplatin (OXAL) is a commonly used chemotherapy for treating colorectal cancer (CRC). A recent genome wide association study (GWAS) showed that a genetic variant (rs11006706) in the lncRNA gene MKX-AS1 and partnered sense gene MKX could impact the response of genetically varied cell lines to OXAL treatment. This study found that the expression levels of MKX-AS1 and MKX in lymphocytes (LCLs) and CRC cell lines differed between the rs11006706 genotypes, indicating that this gene pair could play a role in OXAL response. Further analysis of patient survival data from the Cancer Genome Atlas (TCGA) and other sources showed that patients with high MKX-AS1 expression status had significantly worse overall survival (HR = 3.2; 95%CI = (1.17-9); p = 0.024) compared to cases with low MKX-AS1 expression status. Alternatively, high MKX expression status had significantly better overall survival (HR = 0.22; 95%CI = (0.07-0.7); p = 0.01) compared to cases with low MKX expression status. These results suggest an association between MKX-AS1 and MKX expression status that could be useful as a prognostic marker of response to OXAL and potential patient outcomes in CRC.
Collapse
Affiliation(s)
- Ricardo D Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - George W Small
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Adrian J Green
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27606, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27606, USA
| | - Farida S Akhtari
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Alison A Motsinger-Reif
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | - Tammy M Havener
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David M Reif
- Predictive Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Howard L McLeod
- Center for Precision Medicine and Functional Genomics, Utah Tech University, St. George, UT 84770, USA
| | - Tim Wiltshire
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
14
|
Vo L, Schmidtke MW, Da Rosa-Junior NT, Ren M, Schlame M, Greenberg ML. Cardiolipin metabolism regulates expression of muscle transcription factor MyoD1 and muscle development. J Biol Chem 2023; 299:102978. [PMID: 36739949 PMCID: PMC9999232 DOI: 10.1016/j.jbc.2023.102978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The mitochondrial phospholipid cardiolipin (CL) is critical for numerous essential biological processes, including mitochondrial dynamics and energy metabolism. Mutations in the CL remodeling enzyme TAFAZZIN cause Barth syndrome, a life-threatening genetic disorder that results in severe physiological defects, including cardiomyopathy, skeletal myopathy, and neutropenia. To study the molecular mechanisms whereby CL deficiency leads to skeletal myopathy, we carried out transcriptomic analysis of the TAFAZZIN-knockout (TAZ-KO) mouse myoblast C2C12 cell line. Our data indicated that cardiac and muscle development pathways are highly decreased in TAZ-KO cells, consistent with a previous report of defective myogenesis in this cell line. Interestingly, the muscle transcription factor myoblast determination protein 1 (MyoD1) is significantly repressed in TAZ-KO cells and TAZ-KO mouse hearts. Exogenous expression of MyoD1 rescued the myogenesis defects previously observed in TAZ-KO cells. Our data suggest that MyoD1 repression is caused by upregulation of the MyoD1 negative regulator, homeobox protein Mohawk, and decreased Wnt signaling. Our findings reveal, for the first time, that CL metabolism regulates muscle differentiation through MyoD1 and identify the mechanism whereby MyoD1 is repressed in CL-deficient cells.
Collapse
Affiliation(s)
- Linh Vo
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA
| | - Michael W Schmidtke
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA
| | | | - Mindong Ren
- Department of Anesthesiology, Perioperative Care, and Pain Medicine at New York University Grossman School of Medicine, New York, New York, USA; Department of Cell Biology at New York University Grossman School of Medicine, New York, New York, USA
| | - Michael Schlame
- Department of Anesthesiology, Perioperative Care, and Pain Medicine at New York University Grossman School of Medicine, New York, New York, USA; Department of Cell Biology at New York University Grossman School of Medicine, New York, New York, USA
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA.
| |
Collapse
|
15
|
Peserico A, Barboni B, Russo V, Bernabò N, El Khatib M, Prencipe G, Cerveró-Varona A, Haidar-Montes AA, Faydaver M, Citeroni MR, Berardinelli P, Mauro A. Mammal comparative tendon biology: advances in regulatory mechanisms through a computational modeling. Front Vet Sci 2023; 10:1175346. [PMID: 37180059 PMCID: PMC10174257 DOI: 10.3389/fvets.2023.1175346] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023] Open
Abstract
There is high clinical demand for the resolution of tendinopathies, which affect mainly adult individuals and animals. Tendon damage resolution during the adult lifetime is not as effective as in earlier stages where complete restoration of tendon structure and property occurs. However, the molecular mechanisms underlying tendon regeneration remain unknown, limiting the development of targeted therapies. The research aim was to draw a comparative map of molecules that control tenogenesis and to exploit systems biology to model their signaling cascades and physiological paths. Using current literature data on molecular interactions in early tendon development, species-specific data collections were created. Then, computational analysis was used to construct Tendon NETworks in which information flow and molecular links were traced, prioritized, and enriched. Species-specific Tendon NETworks generated a data-driven computational framework based on three operative levels and a stage-dependent set of molecules and interactions (embryo-fetal or prepubertal) responsible, respectively, for signaling differentiation and morphogenesis, shaping tendon transcriptional program and downstream modeling of its fibrillogenesis toward a mature tissue. The computational network enrichment unveiled a more complex hierarchical organization of molecule interactions assigning a central role to neuro and endocrine axes which are novel and only partially explored systems for tenogenesis. Overall, this study emphasizes the value of system biology in linking the currently available disjointed molecular data, by establishing the direction and priority of signaling flows. Simultaneously, computational enrichment was critical in revealing new nodes and pathways to watch out for in promoting biomedical advances in tendon healing and developing targeted therapeutic strategies to improve current clinical interventions.
Collapse
|
16
|
Iizumi R, Honda M. Wnt/β-Catenin Signaling Inhibits Osteogenic Differentiation in Human Periodontal Ligament Fibroblasts. Biomimetics (Basel) 2022; 7:biomimetics7040224. [PMID: 36546925 PMCID: PMC9776043 DOI: 10.3390/biomimetics7040224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022] Open
Abstract
The periodontal ligament is a collagenous tissue that is important for maintaining the homeostasis of cementum and alveolar bone. In tendon cells, Wnt/β-catenin signaling has been reported to regulate the expression level of Scleraxis (Scx) and Mohawk Homeobox (Mkx) gene and maintain the tissue homeostasis, while its role in the periodontal ligament is unclear. The aim of this study was to investigate the effects of Wnt/β-catenin signaling induced by Wnt-3a stimulation on the inhibition of osteogenic differentiation of human periodontal ligament fibroblasts (HPLFs). During osteogenic differentiation of HPLFs, they formed bone nodules independently of alkaline phosphatase (ALP) activity. After stimulation of Wnt-3a, the expression of β-catenin increased, and nuclear translocation of β-catenin was observed. These data indicate that Wnt-3a activated Wnt/β-catenin signaling. Furthermore, the stimulation of Wnt-3a inhibited the bone nodule formation and suppressed the expression of osteogenic differentiation-related genes such as Runx2, Osteopontin and Osteocalcin, and upregulated the gene expression of Type-I collagen and Periostin (Postn). Scx may be involved in the suppression of osteogenic differentiation in HPLFs. In conclusion, Wnt/β-catenin signaling may be an important signaling pathway that inhibits the osteogenic differentiation in HPLFs by the upregulation of Scx gene expression and downregulation of osteogenic differentiation-related genes.
Collapse
|
17
|
Bai L, Han Q, Meng Z, Chen B, Qu X, Xu M, Su Y, Qiu Z, Xue Y, He J, Zhang J, Yin Z. Bioprinted living tissue constructs with layer-specific, growth factor-loaded microspheres for improved enthesis healing of a rotator cuff. Acta Biomater 2022; 154:275-289. [PMID: 36328126 DOI: 10.1016/j.actbio.2022.10.058] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/13/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Substantial challenges remain in constructing the native tendon-to-bone interface for rotator cuff healing owing to the enthesis tissues' highly organized structural and compositional gradients. Herein, we propose to bioprint living tissue constructs with layer-specific growth factors (GFs) to promote enthesis regeneration by guiding the zonal differentiation of the loaded stem cells in situ. The sustained release of tenogenic, chondrogenic, and osteogenic GFs was achieved via microsphere-based delivery carriers embedded in the bioprinted constructs. Compared to the basal construct without GFs, the layer-specific tissue analogs realized region-specific differentiation of stem cells in vitro. More importantly, bioprinted living tissue constructs with layer-specific GFs rapidly enhanced the enthesis regeneration in a rabbit rotator cuff tear model in terms of biomechanical restoration, collagen deposition, and alignment, showing gradient interface of fibrocartilage structures with aligned collagen fibrils and an ultimate load failure of 154.3 ± 9.5 N resembling those of native enthesis tissues in 12 weeks. This exploration provides a feasible strategy to engineer living tissue constructions with region-specific differentiation potentials for the functional repair of gradient enthesis tissues. STATEMENT OF SIGNIFICANCE: Previous studies that employed acellular layer-specific scaffolds or stem cells for the reconstruction of the rotator cuff faced challenges due to their insufficient capability to rebuild the anisotropic compositional and structural gradients of native enthesis tissues. This manuscript proposed a living tissue construct with layer-specific, GFs-loaded µS, which can direct in situ and region-specific differentiation of the embedded stem cells to tenogenic, chondrogenic, and osteogenic lineages for functional regeneration of the enthesis tissues. This bioprinted living tissue construct with the unique capability to reduce fibrovascular scar tissue formation and simultaneously facilitate enthesis tissue remodeling might provide a promising strategy to repair complex and gradient tissues in the future.
Collapse
Affiliation(s)
- Lang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Qian Han
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zijie Meng
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Baojun Chen
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, 450003, China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Meiguang Xu
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanwen Su
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhennan Qiu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yuan Xue
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Jing Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| | - Zhanhai Yin
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
18
|
Mohawk impedes angiofibrosis by preventing the differentiation of tendon stem/progenitor cells into myofibroblasts. Sci Rep 2022; 12:20003. [PMID: 36411329 PMCID: PMC9678895 DOI: 10.1038/s41598-022-24195-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022] Open
Abstract
Adult tendons heal via fibrovascular scarring with inferior biomechanical properties. Mohawk (Mkx) emerged as a pivotal actor in tenolineage commitment. However, its precise function in tendinopathy remains poorly understood. This study investigates the cellular and molecular mechanisms underlying Mkx' role in fibrovascular healing. Human samples were collected to test fibrovascular markers. We then performed RNAseq on Mkx-/- mice compared to their wild type littermates to decipher Mkx regulome. We therefore sought to reproduce TSPCs transition to myofibroblasts in-vitro by over-expressing MyoD and followed by phenotypic and experimental cells' characterization using microscopy, qRT-PCR, flow cytometry sorting, presto-blue cell viability assay and immunofluorescence. Two different in vivo models were used to assess the effect of the MyoD-expressing myofibroblasts: transplantation in the dorsal area of immunodeficient mice and in an adult Achilles tendon injury model. To prevent angiofibrosis, we tested the molecule Xav939 and proceeded with histological stainings, q-RT PCR transcriptional quantification of angifibrotic markers, mechanical tests, and immunofluorescence. Tendinopathy samples showed fibrovascular healing with decreased tenolineage phenotype. Transcriptomic analysis of Mkx-/- tendons revealed myofibroblast-associated biological processes. Over-expression of MyoD in WT tendon stem progenitor cells (TSPCs) gave rise to myofibroblasts reprogramming in-vitro and fibrovascular scarring in-vivo. MKX directly binds to MyoD promoter and underlies global regulative processes related to angiogenesis and Wnt signaling pathway. Blocking Wnt signaling with the small molecule Xav393 resulted in higher histological and biomechanical properties. Taken together, our data provide the first in vivo and in-vitro evidence of tendon stem progenitor cells to myofibroblasts transition and show improved tendon healing via angiofibrosis modulation, thus opening potential therapeutic avenues to treat tendinopathy patients.
Collapse
|
19
|
Freedman BR, Adu-Berchie K, Barnum C, Fryhofer GW, Salka NS, Shetye S, Soslowsky LJ. Nonsurgical treatment reduces tendon inflammation and elevates tendon markers in early healing. J Orthop Res 2022; 40:2308-2319. [PMID: 34935170 PMCID: PMC9209559 DOI: 10.1002/jor.25251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/07/2021] [Accepted: 12/19/2021] [Indexed: 02/04/2023]
Abstract
Operative treatment is assumed to provide superior outcomes to nonoperative (conservative) treatment following Achilles tendon rupture, however, this remains controversial. This study explores the effect of surgical repair on Achilles tendon healing. Rat Achilles tendons (n = 101) were bluntly transected and were randomized into groups receiving repair or non-repair treatments. By 1 week after injury, repaired tendons had inferior mechanical properties, which continued to 3- and 6-week post-injury, evidenced by decreased dynamic modulus and failure stress. Transcriptomics analysis revealed >7000 differentially expressed genes between repaired and non-repaired tendons after 1-week post-injury. While repaired tendons showed enriched inflammatory gene signatures, non-repaired tendons showed increased tenogenic, myogenic, and mechanosensitive gene signatures, with >200-fold enrichment in Tnmd expression. Analysis of gastrocnemius muscle revealed elevated MMP activity in tendons receiving repair treatment, despite no differences in muscle fiber morphology. Transcriptional regulation analysis highlighted that the highest expressed transcription factors in repaired tendons were associated with inflammation (Nfκb, SpI1, RelA, and Stat1), whereas non-repaired tendons expressed markers associated with tissue development and mechano-activation (Smarca1, Bnc2, Znf521, Fbn1, and Gli3). Taken together, these data highlight distinct differences in healing mechanism occurring immediately following injury and provide insights for new therapies to further augment tendons receiving repaired and non-repaired treatments.
Collapse
Affiliation(s)
- Benjamin R Freedman
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Kwasi Adu-Berchie
- John A Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Carrie Barnum
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - George W Fryhofer
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nabeel S Salka
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Snehal Shetye
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Louis J Soslowsky
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Walia B, Li T, Crosio G, Montero A, Huang A. Axin2-lineage cells contribute to neonatal tendon regeneration. Connect Tissue Res 2022; 63:530-543. [PMID: 35180018 PMCID: PMC9491382 DOI: 10.1080/03008207.2022.2036732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 01/17/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Tendon injuries are a challenging clinical problem with few treatment options. Identifying the molecular regulators of tendon is required for the development of new therapies. While the Wnt pathway is critical for the maintenance and differentiation of many tissues, the role of Wnt signaling in tendon cell biology remains largely unexplored. METHODS The effects of Wnt activation were tested in vitro using neonatal tendon-derived cells cultured in 2D and 3D conditions. The inducible Axin2CreERT2 was then used to label Axin2+ cells in vivo and cells were traced during neonatal tendon regeneration. RESULTS We showed that activation of Wnt signaling results in proliferation of neonatal tendon cells. While tendon marker expression was inhibited by Wnt activation under 2D conditions, Scx expression was not affected under 3D uniaxial tension, suggesting that the microenvironment contextualizes tendon cell response to Wnt signaling. Using an in vivo model of neonatal tendon regeneration, we further showed that Wnt signaling cells comprise a subpopulation of tenocyte and epitenon cells that proliferate after injury and are recruited during regeneration. DISCUSSION Collectively, these studies suggest that Wnt signaling may play a role in tendon cell proliferation, differentiation, and regeneration.
Collapse
Affiliation(s)
- B. Walia
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - T.M. Li
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - G. Crosio
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - A.M. Montero
- Department of Orthopedic Surgery, Columbia University, New York, NY
| | - A.H. Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY
| |
Collapse
|
21
|
Ciardulli MC, Scala P, Giudice V, Santoro A, Selleri C, Oliva F, Maffulli N, Porta GD. Stem Cells from Healthy and Tendinopathic Human Tendons: Morphology, Collagen and Cytokines Expression and Their Response to T3 Thyroid Hormone. Cells 2022; 11:2545. [PMID: 36010622 PMCID: PMC9406581 DOI: 10.3390/cells11162545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to investigate the effect of triiodothyronine (T3) on tendon specific markers and cytokines expression of stem cells extracted from human tendons. Indeed, thyroid hormones have been reported to be protective factors, maintaining tendons' homeostasis, whereas tendinopathy is believed to be related to a failed healing response. Healthy and tendinopathic human tendons were harvested to isolate tendon stem/progenitor cells (TSPCs). TSPCs obtained from pathological samples showed gene expression and morphological modifications at baseline in comparison with cells harvested from healthy tissues. When cells were maintained in a medium supplemented with T3 (10-6 M), only pathological populations showed a significant upregulation of tenogenic markers (DCN, TNC, COL1A1, COL3A1). Immunostaining revealed that healthy cells constantly released type I collagen, typical of tendon matrix, whereas pathological ones overexpressed and secreted type III collagen, typical of scarred and impaired tissue. Pathological cells also overexpressed pro- and anti-inflammatory cytokines, suggesting an impaired balance in the presence of T3, without STAT3 activation. Moreover, DKK-1 was significantly high in the culture medium of pathological cell cultures and was reversed by T3. This study opens perspectives on the complex biochemical alteration of cells from pathological tendons, which may lead to the chronic disease context with an impaired extracellular matrix.
Collapse
Affiliation(s)
- Maria Camilla Ciardulli
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Pasqualina Scala
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Valentina Giudice
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| | - Antonietta Santoro
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Carmine Selleri
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| | - Francesco Oliva
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Nicola Maffulli
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, 275 Bancroft Road, London E1 4DG, UK
| | - Giovanna Della Porta
- Laboratory of Translational Medicine, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Interdepartment Centre BIONAM, University of Salerno, Via Giovanni Paolo I, 84084 Fisciano, Italy
| |
Collapse
|
22
|
Rao Y, Zhu C, Suen HC, Huang S, Liao J, Ker DFE, Tuan RS, Wang D. Tenogenic induction of human adipose-derived stem cells by soluble tendon extracellular matrix: composition and transcriptomic analyses. Stem Cell Res Ther 2022; 13:380. [PMID: 35906661 PMCID: PMC9338462 DOI: 10.1186/s13287-022-03038-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 07/06/2022] [Indexed: 11/27/2022] Open
Abstract
Background Tendon healing is clinically challenging largely due to its inferior regenerative capacity. We have previously prepared a soluble, DNA-free, urea-extracted bovine tendon-derived extracellular matrix (tECM) that exhibits strong pro-tenogenic bioactivity on human adipose-derived stem cells (hASCs). In this study, we aimed to elucidate the mechanism of tECM bioactivity via characterization of tECM protein composition and comparison of transcriptomic profiles of hASC cultures treated with tECM versus collagen type I (Col1) as a control ECM component.
Methods The protein composition of tECM was characterized by SDS-PAGE, hydroxyproline assay, and proteomics analysis. To investigate tECM pro-tenogenic bioactivity and mechanism of action, differentiation of tECM-treated hASC cultures was compared to serum control medium or Col1-treated groups, as assessed via immunofluorescence for tenogenic markers and RNA Sequencing (RNA-Seq).
Results Urea-extracted tECM yielded consistent protein composition, including collagens (20% w/w) and at least 17 non-collagenous proteins (< 100 kDa) based on MS analysis. Compared to current literature, tECM included key tendon ECM components that are functionally involved in tendon regeneration, as well as those that are involved in similar principal Gene Ontology (GO) functions (ECM-receptor interaction and collagen formation) and signaling pathways (ECM-receptor interaction and focal adhesion). When used as a cell culture supplement, tECM enhanced hASC proliferation and tenogenic differentiation compared to the Col1 and FBS treatment groups based on immunostaining of tenogenesis-associated markers. Furthermore, RNA-Seq analysis revealed a total of 584 genes differentially expressed among the three culture groups. Specifically, Col1-treated hASCs predominantly exhibited expression of genes and pathways related to ECM-associated processes, while tECM-treated hASCs expressed a mixture of ECM- and cell activity-associated processes, which may explain in part the enhanced proliferation and tenogenic differentiation of tECM-treated hASCs. Conclusions Our findings showed that urea-extracted tECM contained 20% w/w collagens and is significantly enriched with other non-collagenous tendon ECM components. Compared to Col1 treatment, tECM supplementation enhanced hASC proliferation and tenogenic differentiation as well as induced distinct gene expression profiles. These findings provide insights into the potential mechanism of the pro-tenogenic bioactivity of tECM and support the development of future tECM-based approaches for tendon repair. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03038-0.
Collapse
Affiliation(s)
- Ying Rao
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Chenxian Zhu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Hoi Ching Suen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jinyue Liao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Ministry of Education Key Laboratory for Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China.
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Ministry of Education Key Laboratory for Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China.
| |
Collapse
|
23
|
Shojaee A, Ejeian F, Parham A, Nasr-Esfahani MH. Optimizing Tenogenic Differentiation of Equine Adipose-Derived Mesenchymal Stem Cells (eq-ASC) Using TGFB3 Along with BMP Antagonists. CELL JOURNAL 2022; 24:370-379. [PMID: 36043405 PMCID: PMC9428478 DOI: 10.22074/cellj.2022.7892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Tendon repair strategies usually are accompanied by pathological mineralization and scar tissue formation that increases the risk of re-injuries. This study aimed to establish an efficient tendon regeneration method simultaneously with a reduced risk of ectopic bone formation. MATERIALS AND METHODS In this experimental study, tenogenic differentiation was induced through transforming growth factor- β3 (TGFB3) treatment in combination with the inhibiting concentrations of bone morphogenetic proteins (BMP) antagonists, gremlin-2 (GREM2), and a Wnt inhibitor, namely sclerostin (SOST). The procedure's efficacy was evaluated using real-time polymerase chain reaction (qPCR) for expression analysis of tenogenic markers and osteochondrogenic marker genes. The expression level of two tenogenic markers, SCX and MKX, was also evaluated by immunocytochemistry. Sirius Red staining was performed to examine the amounts of collagen fibers. Moreover, to investigate the impact of the substrate on tenogenic differentiation, the nanofibrous scaffolds that highly resemble tendon extracellular matrix was employed. RESULTS Aggregated features formed in spontaneous normal culture conditions followed by up-regulation of tenogenic and osteogenic marker genes, including SCX, MKX, COL1A1, RUNX2, and CTNNB1. TGFB3 treatment exaggerated morphological changes and markedly amplified tenogenic differentiation in a shorter period of time. Along with TGFB3 treatment, inhibition of BMPs by GREM2 and SOST delayed migratory events to some extent and dramatically reduced osteo-chondrogenic markers synergistically. Nanofibrous scaffolds increased tenogenic markers while declining the expression of osteo-chondrogenic genes. CONCLUSION These findings revealed an appropriate in vitro potential of spontaneous tenogenic differentiation of eq- ASCs that can be improved by simultaneous activation of TGFB and inhibition of osteoinductive signaling pathways.
Collapse
Affiliation(s)
- Asiyeh Shojaee
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad,
Mashhad, Iran
| | - Fatemeh Ejeian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Abbas Parham
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad,
Mashhad, Iran,Stem Cell Biology and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad,
Mashhad, Iran,P.O.Box: 9177948974Division of PhysiologyDepartment of Basic SciencesFaculty of Veterinary MedicineFerdowsi
University of MashhadMashhadIranP.O.Box: 8159358686Department of Animal BiotechnologyReproductive Biomedicine Research CenterRoyan Institute for BiotechnologyACECRIsfahanIran
Emails:,
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran,P.O.Box: 9177948974Division of PhysiologyDepartment of Basic SciencesFaculty of Veterinary MedicineFerdowsi
University of MashhadMashhadIranP.O.Box: 8159358686Department of Animal BiotechnologyReproductive Biomedicine Research CenterRoyan Institute for BiotechnologyACECRIsfahanIran
Emails:,
| |
Collapse
|
24
|
Supokawej A, Korchunjit W, Wongtawan T. The combination of BMP12 and KY02111 enhances tendon differentiation in bone marrow-derived equine mesenchymal stromal cells (BM-eMSCs). J Equine Sci 2022; 33:19-26. [PMID: 35847484 PMCID: PMC9260033 DOI: 10.1294/jes.33.19] [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: 01/27/2022] [Accepted: 05/10/2022] [Indexed: 11/18/2022] Open
Abstract
The Wingless and Int-1 (WNT) and bone morphogenic protein/growth differentiation factor
(BMP/GDF) signalling pathways contribute significantly to the development of the
musculoskeletal system. The mechanism by which they contribute is as follows: BMP/GDF
signalling usually promotes tendon differentiation, whereas WNT signalling inhibits it. We
hypothesised that inhibiting WNT and subsequently stimulating BMP signalling may enhance
the tenogenic differentiation of stem cells. The objective of this study was to determine
whether a combination of WNT inhibitor (KY02111) and BMP12/GDF7 protein could enhance the
differentiation of bone marrow-derived equine mesenchymal stromal cells (BM-eMSCs) into
tenocytes. Cells were cultured in five treatments: control, BMP12, and three different
combinations of BMP12 and KY02111. The results indicated that a 1-day treatment with
KY02111 followed by a 13-day treatment with BMP12 resulted in the highest tenogenic
differentiation score in this experiment. The effect of KY02111 is dependent on the
incubation time, with 1 day being better than 3 or 5 days. This combination increased
tenogenic gene marker expression, including SCX, TNMD, DCN, and TNC, as well as COL1
protein expression. In conclusion, we propose that a combination of BMP12 and KY02111 can
enhance the in vitro tenogenic differentiation of BM-eMSCs more than BMP12 alone. The
findings of this study might be useful for improving tendon differentiation protocols for
stem cell transplantation and application to tendon regeneration.
Collapse
Affiliation(s)
- Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Wasamon Korchunjit
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand.,Laboratory of Cellular Biomedicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Tuempong Wongtawan
- Akkhararatchakumari Veterinary College, Walailak University, Nakhon Si Thammarat 80160, Thailand.,Centre for One Health, Walailak University, Nakhon Si Thammarat 80160, Thailand.,Laboratory of Cellular Biomedicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand
| |
Collapse
|
25
|
Sakaguchi T, Ohkawara B, Kishimoto Y, Miyamoto K, Ishizuka S, Hiraiwa H, Ishiguro N, Imagama S, Ohno K. Promethazine Downregulates Wnt/β-Catenin Signaling and Increases the Biomechanical Forces of the Injured Achilles Tendon in the Early Stage of Healing. Am J Sports Med 2022; 50:1317-1327. [PMID: 35234523 DOI: 10.1177/03635465221077116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Wnt/β-catenin signaling suppresses the differentiation of cultured tenocytes, but its roles in tendon repair remain mostly elusive. No chemical compounds are currently available to treat tendon injury. HYPOTHESIS We hypothesized that the inhibition of Wnt/β-catenin signaling would accelerate tendon healing. STUDY DESIGN Controlled laboratory study. METHODS Tendon-derived cells (TDCs) were isolated from rat Achilles tendons. The right Achilles tendon was injured via a dermal punch, while the left tendon was sham operated. A Wnt/β-catenin inhibitor, IWR-1, and an antihistamine agent, promethazine (PH), were locally and intramuscularly injected, respectively, for 2 weeks after surgery. The healing tendons were histologically and biomechanically evaluated. RESULTS The amount of β-catenin protein was increased in the injured tendons from postoperative weeks 0.5 to 2. Inhibition of Wnt/β-catenin signaling by IWR-1 in healing tendons improved the histological abnormalities and decreased β-catenin, but it compromised the biomechanical properties. As we previously reported that antihistamine agents suppressed Wnt/β-catenin signaling in human chondrosarcoma cells, we examined the effects of antihistamines on TDCs. We found that a first-generation antihistamine agent, PH, increased the expression of the tendon marker genes Mkx and Tnmd in TDCs. Intramuscular injection of PH did not improve histological abnormalities, but it decreased β-catenin in healing tendons and increased the peak force and stiffness of the healing tendons on postoperative week 2. On postoperative week 8, however, the biomechanical properties of vehicle-treated tendons became similar to those of PH-treated tendons. CONCLUSION IWR-1 and PH suppressed Wnt/β-catenin signaling and improved the histological abnormalities of healing tendons. IWR-1, however, compromised the biomechanical properties of healing tendons, whereas PH improved them. CLINICAL RELEVANCE PH is a candidate repositioned drug that potentially accelerates tendon repair.
Collapse
Affiliation(s)
- Takefumi Sakaguchi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yasuzumi Kishimoto
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kentaro Miyamoto
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Shinya Ishizuka
- Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hideki Hiraiwa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Naoki Ishiguro
- Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
26
|
Rajpar I, Tomlinson RE. Function of peripheral nerves in the development and healing of tendon and bone. Semin Cell Dev Biol 2022; 123:48-56. [PMID: 33994302 PMCID: PMC8589913 DOI: 10.1016/j.semcdb.2021.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 01/03/2023]
Abstract
Although the functions of the peripheral nervous system in whole body homeostasis and sensation have been understood for many years, recent investigation has uncovered new roles for innervation in the musculoskeletal system. This review centers on advances regarding the function of nerves in the development and repair of two connected tissues: tendon and bone. Innervation in healthy tendons is generally confined to the tendon sheaths, and tendon-bone attachment units are typically aneural. In contrast to tendon, bone is an innervated and vascularized structure. Historically, the function of abundant peripheral nerves in bone has been limited to pain and some non-painful sensory perception in disease and injury. Indeed, much of our understanding of peripheral nerves in tendons, bones, and entheses is limited to the source and type of innervation in healthy and injured tissues. However, more recent studies have made important observations regarding the appearance, type, and innervation patterns of nerves during embryonic and postnatal development and in response to injury, which suggest a more expansive role for peripheral nerves in the formation of musculoskeletal tissues. Indeed, tendons and bones develop in a close spatiotemporal relationship in the embryonic mesoderm. Models of limb denervation have shed light on the importance of sensory innervation in bone and to a lesser extent, tendon development, and more recent work has unraveled key nerve signaling pathways. Furthermore, loss of sensory innervation also impairs healing of bone fractures and may contribute to chronic tendinopathy. However, more study is required to translate our knowledge of peripheral nerves to therapeutic strategies to combat bone and tendon diseases.
Collapse
Affiliation(s)
- Ibtesam Rajpar
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryan E Tomlinson
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Song K, Jiang T, Pan P, Yao Y, Jiang Q. Exosomes from tendon derived stem cells promote tendon repair through miR-144-3p-regulated tenocyte proliferation and migration. Stem Cell Res Ther 2022; 13:80. [PMID: 35197108 PMCID: PMC8867681 DOI: 10.1186/s13287-022-02723-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tendon derived stem cells (TDSCs) have proven to be effective in tendon repair by secreting paracrine factors, which modulate the function of resident cells and inflammatory process. Exosomes, which are secreted from cells to mediate intercellular communication, may be used to treat tendon injuries. Here, we aimed to determine the effects of exosomes from TDSCs (TDSC-Exos) on tendon repair and to explore the underlying mechanism by investigating the role of microRNAs (miRNAs). METHODS TDSC-Exos were isolated from TDSC conditioned medium. In vitro studies were performed to investigate the effects of TDSC-Exos on the proliferation, migration, cytoprotection, collagen production and tendon-specific markers expression in tenocytes. In order to determine the therapeutic effects of TDSC-Exos in vivo, we used a scaffold of photopolymerizable hyaluronic acid (p-HA) loaded with TDSC-Exos (pHA-TDSC-Exos) to treat tendon defects in the rat model. Subsequently, RNA sequencing and bioinformatic analyses were used to screen for enriched miRNAs in TDSC-Exos and predict target genes. The miRNA-target transcript interaction was confirmed by a dual-luciferase reporter assay system. In order to determine the role of candidate miRNA and its target gene in TDSC-Exos-regulated tendon repair, miRNA mimic and inhibitor were transfected into tenocytes to evaluate cell proliferation and migration. RESULTS Treatment with TDSC-Exos promoted proliferation, migration, type I collagen production and tendon-specific markers expression in tenocytes, and also protected tenocytes from oxidative stress and serum deprivation. The scaffold of pHA-TDSC-Exos could sever as a sustained release system to treat the rat model of tendon defects. In vivo study showed that TDSC-Exos promoted early healing of injured tendons. Rats treated with TDSC-Exos had better fiber arrangement and histological scores at the injury site. Besides, the injured tendons treated with TDSC-Exos had better performance in the biomechanical testing. Therefore, the pHA-TDSC-Exos scaffold proved to facilitate tendon repair in the rat model. miR-144-3p was enriched in TDSC-Exos and promoted tenocyte proliferation and migration via targeting AT-rich interactive domain 1A (ARID1A). CONCLUSIONS TDSC-Exos enhanced tenon repair through miR-144-3p-regulated tenocyte proliferation and migration. These results suggest that TDSC-Exos can serve as a promising strategy to treat tendon injuries.
Collapse
Affiliation(s)
- Kai Song
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China.,Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China
| | - Tao Jiang
- Department of Orthopedic Surgery, The Affiliated Yixing Hospital of Jiangsu University, Wuxi, 214200, Jiangsu, People's Republic of China
| | - Pin Pan
- Department of Orthopedic Surgery, The Second People's Hospital of Hefei, Hefei, 230011, Anhui, People's Republic of China
| | - Yao Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China.,Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China. .,Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China.
| |
Collapse
|
28
|
Wang Y, Zhang J, Lin Y, Cheng S, Wang D, Rao M, Jiang Y, Huang X, Chen R, Xie Y, Yin P, Cheng B. A Global Phosphorylation Atlas of Proteins Within Pathological Site of Rotator Cuff Tendinopathy. Front Mol Biosci 2022; 8:787008. [PMID: 35242811 PMCID: PMC8886731 DOI: 10.3389/fmolb.2021.787008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Rotator cuff tendinopathy (RCT) is the most common cause of shoulder pain, therefore posing an important clinical problem. Understanding the mechanism and biochemical changes of RCT would be of crucial importance and pave the path to targeting novel and effective therapeutic strategies in translational perspectives and clinical practices. Phosphorylation, as one of the most important and well-studied post-translational modifications, is tightly associated with protein activity and protein functional regulation. Here in this study, we generated a global protein phosphorylation atlas within the pathological site of human RCT patients. By using Tandem Mass Tag (TMT) labeling combined with mass spectrometry, an average of 7,741 phosphorylation sites (p-sites) and 3,026 proteins were identified. Compared with their normal counterparts, 1,668 p-sites in 706 proteins were identified as upregulated, while 73 p-sites in 57 proteins were downregulated. GO enrichment analyses have shown that majority of proteins with upregulated p-sites functioned in neutrophil-mediated immunity whereas downregulated p-sites are mainly involved in muscle development. Furthermore, pathway analysis identified NF-κB–related TNF signaling pathway and protein kinase C alpha type (PKCα)–related Wnt signaling pathway were associated with RCT pathology. At last, a weighted kinase-site phosphorylation network was built to identify potentially core kinase, from which serine/threonine-protein kinase 39 (STLK3) and mammalian STE20-like protein kinase 1 (MST1) were proposed to be positively correlated with the activation of Wnt pathway.
Collapse
Affiliation(s)
- Yezhou Wang
- School of Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Jiawei Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China
- China Rehabilitation Research Center, Beijing, China
| | - Yuan Lin
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shi Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Duanyang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Man Rao
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Yuheng Jiang
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Xiang Huang
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Ruijing Chen
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Yong Xie
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
- *Correspondence: Pengbin Yin, ; Biao Cheng,
| | - Biao Cheng
- School of Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Pengbin Yin, ; Biao Cheng,
| |
Collapse
|
29
|
He P, Ruan D, Huang Z, Wang C, Xu Y, Cai H, Liu H, Fei Y, Heng BC, Chen W, Shen W. Comparison of Tendon Development Versus Tendon Healing and Regeneration. Front Cell Dev Biol 2022; 10:821667. [PMID: 35141224 PMCID: PMC8819183 DOI: 10.3389/fcell.2022.821667] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/07/2022] [Indexed: 12/27/2022] Open
Abstract
Tendon is a vital connective tissue in human skeletal muscle system, and tendon injury is very common and intractable in clinic. Tendon development and repair are two closely related but still not fully understood processes. Tendon development involves multiple germ layer, as well as the regulation of diversity transcription factors (Scx et al.), proteins (Tnmd et al.) and signaling pathways (TGFβ et al.). The nature process of tendon repair is roughly divided in three stages, which are dominated by various cells and cell factors. This review will describe the whole process of tendon development and compare it with the process of tendon repair, focusing on the understanding and recent advances in the regulation of tendon development and repair. The study and comparison of tendon development and repair process can thus provide references and guidelines for treatment of tendon injuries.
Collapse
Affiliation(s)
- Peiwen He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Zizhan Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yiwen Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Honglu Cai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Hengzhi Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School of Stomatology, Bejing, China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| |
Collapse
|
30
|
Delgado Caceres M, Angerpointner K, Galler M, Lin D, Michel PA, Brochhausen C, Lu X, Varadarajan AR, Warfsmann J, Stange R, Alt V, Pfeifer CG, Docheva D. Tenomodulin knockout mice exhibit worse late healing outcomes with augmented trauma-induced heterotopic ossification of Achilles tendon. Cell Death Dis 2021; 12:1049. [PMID: 34741033 PMCID: PMC8571417 DOI: 10.1038/s41419-021-04298-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/23/2022]
Abstract
Heterotopic ossification (HO) represents a common problem after tendon injury with no effective treatment yet being developed. Tenomodulin (Tnmd), the best-known mature marker for tendon lineage cells, has important effects in tendon tissue aging and function. We have reported that loss of Tnmd leads to inferior early tendon repair characterized by fibrovascular scaring and therefore hypothesized that its lack will persistently cause deficient repair during later stages. Tnmd knockout (Tnmd-/-) and wild-type (WT) animals were subjected to complete Achilles tendon surgical transection followed by end-to-end suture. Lineage tracing revealed a reduction in tendon-lineage cells marked by ScleraxisGFP, but an increase in alpha smooth muscle actin myofibroblasts in Tnmd-/- tendon scars. At the proliferative stage, more pro-inflammatory M1 macrophages and larger collagen II cartilaginous template were detected in this group. At the remodeling stage, histological scoring revealed lower repair quality in the injured Tnmd-/- tendons, which was coupled with higher HO quantified by micro-CT. Tendon biomechanical properties were compromised in both groups upon injury, however we identified an abnormal stiffening of non-injured Tnmd-/- tendons, which possessed higher static and dynamic E-moduli. Pathologically thicker and abnormally shaped collagen fibrils were observed by TEM in Tnmd-/- tendons and this, together with augmented HO, resulted in diminished running capacity of Tnmd-/- mice. These novel findings demonstrate that Tnmd plays a protecting role against trauma-induced endochondral HO and can inspire the generation of novel therapeutics to accelerate repair.
Collapse
Affiliation(s)
- Manuel Delgado Caceres
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Katharina Angerpointner
- Hand, Elbow and Plastic Surgery Department, Schön Klinik München Harlaching, Munich, Germany
| | - Michael Galler
- Department of Trauma Surgery, Caritas Hospital St. Josef, Regensburg, Germany
| | - Dasheng Lin
- Orthopaedic Center of People's Liberation Army, The Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Philipp A Michel
- Department of Trauma-, Hand-, and Reconstructive Surgery, University Hospital Münster, Münster, Germany
| | | | - Xin Lu
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Adithi R Varadarajan
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Jens Warfsmann
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute for Musculoskeletal Medicine, University Hospital Münster, Westfälische Wilhelms-University, Münster, Germany
| | - Volker Alt
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Clinic and Policlinic for Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Christian G Pfeifer
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Clinic and Policlinic for Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
31
|
Moreno SE, Massee M, Bara H, Koob TJ. Dehydrated human amniotic membrane modulates canonical Wnt signaling in multiple cell types in vitro. Eur J Cell Biol 2021; 100:151168. [PMID: 34246182 DOI: 10.1016/j.ejcb.2021.151168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 10/21/2022] Open
Abstract
Canonical Wnt signaling is a major pathway known to regulate diverse physiological processes in multicellular organisms. Signaling is tightly regulated by feedback mechanisms; however, persistent dysregulation of this pathway is implicated in the progression of multiple disease states. In this study, proteomic analysis identified endogenous Wnt antagonists in micronized dehydrated human amnion/chorion membrane (μdHACM); thereby, prompting a study to further characterize the intrinsic properties of μdHACM as it relates to Wnt activity, in vitro. A TCF/LEF reporter cell line demonstrated the general ability of μdHACM to inhibit β-catenin induced transcription activity. Furthermore, in vitro systems, modeling elevated Wnt signaling, were developed in relevant cell types including tenocytes, synoviocytes, and human dermal fibroblasts (HDFs). Stimulation of these cells with Wnt3A resulted in translocation of β-catenin to the nucleus and increased expression of Wnt related genes. The subsequent addition of μdHACM, in the continued presence of Wnt-stimulus, mitigated the downstream effects of Wnt3A in tenocytes, synoviocytes, and HDFs. Nuclear localization of β-catenin was abated with corresponding reduction of Wnt related gene expression. These data demonstrate the in vitro regulation of canonical Wnt signaling as an inherent property of μdHACM and a novel mechanism of action.
Collapse
Affiliation(s)
- Sarah E Moreno
- MiMedx Group, Inc. 1775 West Oak Commons Court NE, Marietta, GA 30062, USA
| | - Michelle Massee
- MiMedx Group, Inc. 1775 West Oak Commons Court NE, Marietta, GA 30062, USA.
| | - Heather Bara
- MiMedx Group, Inc. 1775 West Oak Commons Court NE, Marietta, GA 30062, USA
| | - Thomas J Koob
- MiMedx Group, Inc. 1775 West Oak Commons Court NE, Marietta, GA 30062, USA
| |
Collapse
|
32
|
Abdulmalik S, Ramos D, Rudraiah S, Banasavadi-Siddegowda YK, Kumbar SG. The glucagon-like peptide 1 receptor agonist Exendin-4 induces tenogenesis in human mesenchymal stem cells. Differentiation 2021; 120:1-9. [PMID: 34062407 DOI: 10.1016/j.diff.2021.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/21/2021] [Accepted: 05/16/2021] [Indexed: 11/26/2022]
Abstract
Tendon injuries are common and account for up to 50% of musculoskeletal injuries in the United States. The poor healing nature of the tendon is attributed to poor vascularization and cellular composition. In the absence of FDA-approved growth factors for tendon repair, engineering strategies using bioactive factors, donor cells, and delivery matrices to promote tendon repair and regeneration are being explored. Growth factor alternatives in the form of small molecules, donor cells, and progenitors offer several advantages and enhance the tendon healing response. Small drug molecules and peptides offer stability over growth factors that are known to suffer from relatively short biological half-lives. The primary focus of this study was to assess the ability of the exendin-4 (Ex-4) peptide, a glucagon-like peptide 1 (GLP-1) receptor agonist, to induce tenocyte differentiation in bone marrow-derived human mesenchymal stem cells (hMSCs). We treated hMSCs with varied doses of Ex-4 in culture media to evaluate proliferation and tendonogenic differentiation. A 20 nM Ex-4 concentration was optimal for promoting cell proliferation and tendonogenic differentiation. Tendonogenic differentiation of hMSCs was evaluated via gene expression profile, immunofluorescence, and biochemical analyses. Collectively, the levels of tendon-related transcription factors (Mkx and Scx) and extracellular matrix (Col-I, Dcn, Bgn, and Tnc) genes and proteins were elevated compared to media without Ex-4 and other controls including insulin and IGF-1 treatments. The tendonogenic factor Ex-4 in conjunction with hMSCs appear to enhance tendon regeneration.
Collapse
Affiliation(s)
- Sama Abdulmalik
- University of Connecticut Health Center, Department of Orthopedic Surgery, Farmington, CT, USA; University of Connecticut, Biomedical Engineering, Storrs, CT, USA
| | - Daisy Ramos
- University of Connecticut Health Center, Department of Orthopedic Surgery, Farmington, CT, USA; University of Connecticut, Materials Science and Engineering, Storrs, CT, USA
| | - Swetha Rudraiah
- University of Connecticut Health Center, Department of Orthopedic Surgery, Farmington, CT, USA; University of St. Joseph, Department of Pharmaceutical Sciences, Hartford, CT, USA
| | | | - Sangamesh G Kumbar
- University of Connecticut Health Center, Department of Orthopedic Surgery, Farmington, CT, USA; University of Connecticut, Biomedical Engineering, Storrs, CT, USA; University of Connecticut, Materials Science and Engineering, Storrs, CT, USA.
| |
Collapse
|
33
|
Theodossiou SK, Murray JB, Hold LA, Courtright JM, Carper AM, Schiele NR. Akt signaling is activated by TGFβ2 and impacts tenogenic induction of mesenchymal stem cells. Stem Cell Res Ther 2021; 12:88. [PMID: 33499914 PMCID: PMC7836508 DOI: 10.1186/s13287-021-02167-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Background Tissue engineered and regenerative approaches for treating tendon injuries are challenged by the limited information on the cellular signaling pathways driving tenogenic differentiation of stem cells. Members of the transforming growth factor (TGF) β family, particularly TGFβ2, play a role in tenogenesis, which may proceed via Smad-mediated signaling. However, recent evidence suggests some aspects of tenogenesis may be independent of Smad signaling, and other pathways potentially involved in tenogenesis are understudied. Here, we examined the role of Akt/mTORC1/P70S6K signaling in early TGFβ2-induced tenogenesis of mesenchymal stem cells (MSCs) and evaluated TGFβ2-induced tenogenic differentiation when Smad3 is inhibited. Methods Mouse MSCs were treated with TGFβ2 to induce tenogenesis, and Akt or Smad3 signaling was chemically inhibited using the Akt inhibitor, MK-2206, or the Smad3 inhibitor, SIS3. Effects of TGFβ2 alone and in combination with these inhibitors on the activation of Akt signaling and its downstream targets mTOR and P70S6K were quantified using western blot analysis, and cell morphology was assessed using confocal microscopy. Levels of the tendon marker protein, tenomodulin, were also assessed. Results TGFβ2 alone activated Akt signaling during early tenogenic induction. Chemically inhibiting Akt prevented increases in tenomodulin and attenuated tenogenic morphology of the MSCs in response to TGFβ2. Chemically inhibiting Smad3 did not prevent tenogenesis, but appeared to accelerate it. MSCs treated with both TGFβ2 and SIS3 produced significantly higher levels of tenomodulin at 7 days and morphology appeared tenogenic, with localized cell alignment and elongation. Finally, inhibiting Smad3 did not appear to impact Akt signaling, suggesting that Akt may allow TGFβ2-induced tenogenesis to proceed during disruption of Smad3 signaling. Conclusions These findings show that Akt signaling plays a role in TGFβ2-induced tenogenesis and that tenogenesis of MSCs can be initiated by TGFβ2 during disruption of Smad3 signaling. These findings provide new insights into the signaling pathways that regulate tenogenic induction in stem cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02167-2.
Collapse
Affiliation(s)
- Sophia K Theodossiou
- Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, ID, 83844, Moscow, USA
| | - Jett B Murray
- Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, ID, 83844, Moscow, USA
| | - LeeAnn A Hold
- Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, ID, 83844, Moscow, USA
| | - Jeff M Courtright
- Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, ID, 83844, Moscow, USA
| | - Anne M Carper
- Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, ID, 83844, Moscow, USA
| | - Nathan R Schiele
- Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, ID, 83844, Moscow, USA.
| |
Collapse
|
34
|
Comparative Analysis of Tenogenic Gene Expression in Tenocyte-Derived Induced Pluripotent Stem Cells and Bone Marrow-Derived Mesenchymal Stem Cells in Response to Biochemical and Biomechanical Stimuli. Stem Cells Int 2021; 2021:8835576. [PMID: 33510795 PMCID: PMC7825360 DOI: 10.1155/2021/8835576] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/19/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The tendon is highly prone to injury, overuse, or age-related degeneration in both humans and horses. Natural healing of injured tendon is poor, and cell-based therapeutic treatment is still a significant clinical challenge. In this study, we extensively investigated the expression of tenogenic genes in equine bone marrow mesenchymal stem cells (BMSCs) and tenocyte-derived induced pluripotent stem cells (teno-iPSCs) stimulated by growth factors (TGF-β3 and BMP12) combined with ectopic expression of tenogenic transcription factor MKX or cyclic uniaxial mechanical stretch. Western blotting revealed that TGF-β3 and BMP12 increased the expression of transcription factors SCX and MKX in both cells, but the tenocyte marker tenomodulin (TNMD) was detected only in BMSCs and upregulated by either inducer. On the other hand, quantitative real-time PCR showed that TGF-β3 increased the expression of EGR1, COL1A2, FMOD, and TNC in BMSCs and SCX, COL1A2, DCN, FMOD, and TNC in teno-iPSCs. BMP12 treatment elevated SCX, MKX, DCN, FMOD, and TNC in teno-iPSCs. Overexpression of MKX increased SCX, DCN, FMOD, and TNC in BMSCs and EGR1, COL1A2, DCN, FMOD, and TNC in teno-iPSCs; TGF-β3 further enhanced TNC in BMSCs. Moreover, mechanical stretch increased SCX, EGR1, DCN, ELN, and TNC in BMSCs and SCX, MKX, EGR1, COL1A2, DCN, FMOD, and TNC in teno-iPSCs; TGF-β3 tended to further elevate SCX, ELN, and TNC in BMSCs and SCX, MKX, COL1A2, DCN, and TNC in teno-iPSCs, while BMP12 further uptrended the expression of SCX and DCN in BMSCs and DCN in teno-iPSCs. Additionally, the aforementioned tenogenic inducers also affected the expression of signaling regulators SMAD7, ETV4, and SIRT1 in BMSCs and teno-iPSCs. Taken together, our data demonstrate that, in respect to the tenocyte-lineage-specific gene expression, BMSCs and teno-iPSCs respond differently to the tenogenic stimuli, which may affect the outcome of their application in tendon repair or regeneration.
Collapse
|
35
|
Wang HN, Huang YC, Ni GX. Mechanotransduction of stem cells for tendon repair. World J Stem Cells 2020; 12:952-965. [PMID: 33033557 PMCID: PMC7524696 DOI: 10.4252/wjsc.v12.i9.952] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/06/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tendon is a mechanosensitive tissue that transmits force from muscle to bone. Physiological loading contributes to maintaining the homeostasis and adaptation of tendon, but aberrant loading may lead to injury or failed repair. It is shown that stem cells respond to mechanical loading and play an essential role in both acute and chronic injuries, as well as in tendon repair. In the process of mechanotransduction, mechanical loading is detected by mechanosensors that regulate cell differentiation and proliferation via several signaling pathways. In order to better understand the stem-cell response to mechanical stimulation and the potential mechanism of the tendon repair process, in this review, we summarize the source and role of endogenous and exogenous stem cells active in tendon repair, describe the mechanical response of stem cells, and finally, highlight the mechanotransduction process and underlying signaling pathways.
Collapse
Affiliation(s)
- Hao-Nan Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Yong-Can Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
- National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Guo-Xin Ni
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| |
Collapse
|
36
|
Carroll CC, Patel SH, Simmons J, Gordon BD, Olson JF, Chemelewski K, Saw S, Hale TM, Howden R, Sabbaghi A. The Impact of Genistein Supplementation on Tendon Functional Properties and Gene Expression in Estrogen-Deficient Rats. J Med Food 2020; 23:1266-1274. [PMID: 32345111 DOI: 10.1089/jmf.2019.0293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tendinopathy risk increases with menopause. The phytoestrogen genistein prevents collagen loss during estrogen deficiency (ovariectomy [OVX]). The influence of genistein on tendon function and extracellular matrix (ECM) regulation is not well known. We determined the impact of genistein on tendon function and the expression of several genes important for the regulation of tendon ECM. Eight-week-old rats (n = 42) were divided into three groups: intact, OVX, or OVX-genistein (6 mg/kg/day) for 6 weeks. Tail fascicles were assessed with a Deben tensile stage. Achilles tendon mRNA expression was determined with digital droplet polymerase chain reaction. Compared to intact, fascicle stress tended to be lower in untreated OVX rats (P = .022). Furthermore, fascicle modulus and energy density were greater in genistein-treated rats (P < .05) compared to intact. Neither OVX nor genistein altered expression of Col1a1, Col3a1, Casp3, Casp8, Mmp1a, Mmp2, or Mmp9 (P > .05). Compared to intact, Tnmd and Esr1 expression were greater and Pcna and Timp1 expression were lower in OVX rats (P < .05). Genistein treatment returned Tnmd, Pcna, and Timp1 to levels of intact-vehicle (P < .05), but did not alter Scx or Esr1 (P > .05). Several β-catenin/Wnt signaling-related molecules were not altered by OVX or genistein (P > .05). Our findings demonstrate that genistein improves tendon function in estrogen-deficient rats. The effect of genistein in vivo was predominately on genes related to cell proliferation rather than collagen remodeling.
Collapse
Affiliation(s)
- Chad C Carroll
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana, USA.,Department of Physiology, Midwestern University, Glendale, Arizona, USA
| | - Shivam H Patel
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana, USA
| | - Jessica Simmons
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana, USA
| | - Ben Dh Gordon
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Jay F Olson
- Department of Physiology, Midwestern University, Glendale, Arizona, USA
| | - Kali Chemelewski
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana, USA
| | - Shannon Saw
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana, USA
| | - Taben M Hale
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Reuben Howden
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Arman Sabbaghi
- Department of Statistics, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
37
|
Rajpar I, Barrett JG. Multi-differentiation potential is necessary for optimal tenogenesis of tendon stem cells. Stem Cell Res Ther 2020; 11:152. [PMID: 32272975 PMCID: PMC7146987 DOI: 10.1186/s13287-020-01640-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 02/21/2020] [Accepted: 03/10/2020] [Indexed: 01/07/2023] Open
Abstract
Background Tendon injury is a significant clinical problem due to poor healing and a high reinjury rate; successful treatment is limited by our poor understanding of endogenous tendon stem cells. Recent evidence suggests that adult stem cells are phenotypically diverse, even when comparing stem cells isolated from the same tissue from the same individual, and may in fact exist on a spectrum of proliferation and differentiation capacities. Additionally, the relationships between and clinical relevance of this phenotypic variation are poorly understood. In particular, tenogenic capacity has not been studied in comparison to tenogenic differentiation and cell proliferation. Toward this end, we performed a comprehensive assessment of cell proliferation and differentiation capacity toward four connective tissue lineages (tendon, cartilage, bone, and adipose) using tendon stem cell lines derived from single cells released directly from tendon tissue to (1) evaluate the differences, if any, in tenogenic potential, and (2) identify the relationships between differentiation phenotypes and proliferation capacity. Methods Tendon stem cells were derived from the endotenon of superficial digital flexor tendon from 3 horses. The cell suspension from each horse was separately plated simultaneously (1) at moderate density to generate a heterogenous population of cells—parent tendon cell line—and (2) at low density to separate single cells from each other to allow isolation of colonies that derive from single mother cells—clonal tendon stem cell lines. Thirty clonal tendon stem cell lines—10 from each horse—and each parent tendon cell line were assessed for tenogenesis, tri-lineage differentiation, and cell proliferation. Differentiation was confirmed by lineage-specific cell staining and quantified by the relative gene expression of lineage-specific markers. Statistical significance was determined using analysis of variance and post hoc Tukey’s tests. Results Three distinct differentiation phenotypes—differentiation potency toward all 4 tissue lineages and two tri-lineage differentiation potencies—were identified in tendon clonal stem cell lines. These phenotypes were differentiation toward (1) tendon, cartilage, bone, and adipose (TCOA); (2) tendon, cartilage, and bone (TCO); and (3) tendon, cartilage, and adipose (TCA). Further, clonal cell lines that differentiated toward all four lineages had the highest expression of scleraxis and mohawk upon tenogenesis. Moreover, cell proliferation was significantly different between phenotypic groups, as evidenced by increased numbers of cumulative cell population doublings in clonal cell lines that did not differentiate toward adipose. Conclusions Our study provides evidence of the heterogenous character of adult stem cells and identifies key differences in tendon stem cell differentiation and proliferative potentials from the same individual and from the same tendon. Isolation of tendon stem cell lines with the capacity to differentiate into all four connective tissue lineages may yield improved therapeutic benefits in clinical models of repair and promote a native, regenerative phenotype in engineered tendons. Future studies may be targeted to understanding the functional contributions of each tendon stem cell phenotype in vivo and identifying additional cell phenotypes.
Collapse
Affiliation(s)
- Ibtesam Rajpar
- Department of Large Animal Clinical Sciences, Marion duPont Scott Equine Medical Center, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, 17690 Old Waterford Road, Leesburg, VA, 20176, USA
| | - Jennifer G Barrett
- Department of Large Animal Clinical Sciences, Marion duPont Scott Equine Medical Center, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, 17690 Old Waterford Road, Leesburg, VA, 20176, USA.
| |
Collapse
|
38
|
Chen M, Li Y, Xiao L, Dai G, Lu P, Wang Y, Rui Y. AQP1 modulates tendon stem/progenitor cells senescence during tendon aging. Cell Death Dis 2020; 11:193. [PMID: 32188840 PMCID: PMC7080760 DOI: 10.1038/s41419-020-2386-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
The link between tendon stem/progenitor cells (TSPCs) senescence and tendon aging has been well recognized. However, the cellular and molecular mechanisms of TSPCs senescence are still not fully understood. In present study, we investigated the role of Aquaporin 1 (AQP1) in TSPCs senescence. We showed that AQP1 expression declines with age during tendon aging. In aged TSPCs, overexpression of AQP1 significantly attenuated TSPCs senescence. In addition, AQP1 overexpression also restored the age-related dysfunction of self-renewal, migration and tenogenic differentiation. Furthermore, we demonstrated that the JAK-STAT signaling pathway is activated in aged TSPCs, and AQP1 overexpression inhibited the JAK-STAT signaling pathway activation which indicated that AQP1 attenuates senescence and age-related dysfunction of TSPCs through the repression of JAK−STAT signaling pathway. Taken together, our findings demonstrated the critical role of AQP1 in the regulation of TSPCs senescence and provided a novel target for antagonizing tendon aging.
Collapse
Affiliation(s)
- Minhao Chen
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Yingjuan Li
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China.,Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Longfei Xiao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Guangchun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China. .,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China. .,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China. .,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
39
|
Abstract
Tendons connect muscles to bones to transfer the forces necessary for movement. Cell-cell junction proteins, cadherins and connexins, may play a role in tendon development and injury. In this review, we begin by highlighting current understanding of how cell-cell junctions may regulate embryonic tendon development and differentiation. We then examine cell-cell junctions in postnatal tendon, before summarizing the role of cadherins and connexins in adult tendons. More information exists regarding the role of cell-cell junctions in the formation and homeostasis of other musculoskeletal tissues, namely cartilage and bone. Therefore, to inform future tendon studies, we include a brief survey of cadherins and connexins in chondrogenesis and osteogenesis, and summarize how cell-cell junctions are involved in some musculoskeletal tissue pathologies. An enhanced understanding of how cell-cell junctions participate in tendon development, maintenance, and disease will benefit future regenerative strategies.
Collapse
Affiliation(s)
| | - Jett B Murray
- Biological Engineering, University of Idaho, Moscow, ID
| | | |
Collapse
|
40
|
Pan T, Tong X, Ye L, Ji M, Jiao J. EFFECTS OF CONTUSION AND EXHAUSTIVE EXERCISE ON MG53, PTRF IN SKELETAL MUSCLE OF RATS. REV BRAS MED ESPORTE 2019. [DOI: 10.1590/1517-869220192506197718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Objectives To study the effects of contusion and exhaustive exercise on gene expression of MG53, PTRF, Pax7 and β-catenin in skeletal muscle of rats, and reveal the repair mechanism of skeletal muscle injury. Methods Forty-two male Wistar rats were randomly divided into 7 groups, with 6 rats in each group. All groups were euthanized at different time points after exhaustive exercise and contusion, respectively, while the control group was euthanized in resting state. The right gastrocnemius muscles were measured for mRNAs of MG53, PTRF, Pax7 and β-catenin by real time PCR. Results MG53 mRNA and PTRF mRNA of skeletal muscle in groups immediately after exhaustive exercise and after contusion increased significantly (p<0.05), while the two indices decreased constantly at 24 and 48 hours after injury with a similar change trend. Compared with the control group, Pax7 mRNA of skeletal muscle as a marker showed no significant difference in exhaustive exercise groups, but decreased at 48 hours after contusion (p<0.05). β-catenin mRNA of skeletal muscle down-regulated significantly over 24 hours after injury, then activated with an increased value at 48 hours after contusion (p<0.05). As a whole, the variations in the above indices in the contusion groups covered a wider range than in the exhaustive exercise groups. Conclusion The cytomembrane repair mechanism of MG53 and PTRF began immediately after the end of exhaustive exercise and contusion. Activation of Pax7 as the satellite cell marker took longer, and Wnt/β-catenin pathway showed first a decrease and then an increase resulting from the time-dependent gene expression during the repair of skeletal muscle injury. Level of evidence III, Therapeutic studies investigating the results of treatment.
Collapse
Affiliation(s)
| | | | - Leilei Ye
- Nanjing Institute of Physical Education and Sports, China
| | | | | |
Collapse
|
41
|
A Tppp3 +Pdgfra + tendon stem cell population contributes to regeneration and reveals a shared role for PDGF signalling in regeneration and fibrosis. Nat Cell Biol 2019; 21:1490-1503. [PMID: 31768046 PMCID: PMC6895435 DOI: 10.1038/s41556-019-0417-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 09/27/2019] [Indexed: 12/27/2022]
Abstract
Tendon injuries cause prolonged disability and never recover completely. Current mechanistic understanding of tendon regeneration is limited. Here we use single cell transcriptomics to identify a tubulin polymerization-promoting protein family member 3-expressing (Tppp3+) cell population as potential tendon stem cells. Through inducible lineage tracing, we demonstrated that these cells can generate new tenocytes and self-renew upon injury. A fraction of Tppp3+ cells expresses platelet-derived growth factor receptor alpha (Pdfgra). Ectopic platelet-derived growth factor-AA (PDGF-AA) protein induces new tenocyte production while inactivation of Pdgfra in Tppp3+ cells blocks tendon regeneration. These results support Tppp3+Pdgfra+ cells as tendon stem cells. Unexpectedly, Tppp3−Pdgfra+ fibro-adipogenic progenitors coexist in tendon stem cell niche and give rise to fibrotic cells, revealing a clandestine origin of fibrotic scars in healing tendons. Our results explain why fibrosis occurs in injured tendons and present clinical challenges to enhance tendon regeneration without a concurrent increase in fibrosis by PDGF application.
Collapse
|
42
|
Dissecting cell diversity and connectivity in skeletal muscle for myogenesis. Cell Death Dis 2019; 10:427. [PMID: 31160550 PMCID: PMC6546706 DOI: 10.1038/s41419-019-1647-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/25/2019] [Accepted: 05/06/2019] [Indexed: 12/28/2022]
Abstract
Characterized by their slow adhering property, skeletal muscle myogenic progenitor cells (MPCs) have been widely utilized in skeletal muscle tissue engineering for muscle regeneration, but with limited efficacy. Skeletal muscle regeneration is regulated by various cell types, including a large number of rapidly adhering cells (RACs) where their functions and mechanisms are still unclear. In this study, we explored the function of RACs by co-culturing them with MPCs in a biomimetic skeletal muscle organoid system. Results showed that RACs promoted the myogenic potential of MPCs in the organoid. Single-cell RNA-Seq was also performed, classifying RACs into 7 cell subtypes, including one newly described cell subtype: teno-muscular cells (TMCs). Connectivity map of RACs and MPCs subpopulations revealed potential growth factors (VEGFA and HBEGF) and extracellular matrix (ECM) proteins involvement in the promotion of myogenesis of MPCs during muscle organoid formation. Finally, trans-well experiments and small molecular inhibitors blocking experiments confirmed the role of RACs in the promotion of myogenic differentiation of MPCs. The RACs reported here revealed complex cell diversity and connectivity with MPCs in the biomimetic skeletal muscle organoid system, which not only offers an attractive alternative for disease modeling and in vitro drug screening but also provides clues for in vivo muscle regeneration.
Collapse
|
43
|
Shi L, Li YJ, Dai GC, Lin YC, Li G, Wang C, Chen H, Rui YF. Impaired function of tendon-derived stem cells in experimental diabetes mellitus rat tendons: implications for cellular mechanism of diabetic tendon disorder. Stem Cell Res Ther 2019; 10:27. [PMID: 30646947 PMCID: PMC6332703 DOI: 10.1186/s13287-018-1108-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 12/20/2022] Open
Abstract
Background Patients with diabetes mellitus (DM) often suffered with many musculoskeletal disorders, such as tendon rupture and tendinopathy. However, the understanding of the pathogenesis of these alternations is limited. This study was designed to investigate the role of tendon-derived stem cells (TDSCs) in histopathological alterations of DM tendons. Methods Forty-two SD rats were randomly and equally divided into a diabetes group (DG) and control group (CG). DM was induced by streptozotocin (65 mg/kg). The patellar tendons were isolated at weeks 1, 2, and 4 for histological analysis. TDSCs were isolated at week 2 for osteo-chondrogenic differentiation analysis. Mann-Whitney U test was used with SPSS. p < 0.050 was statistically significant. Results Micro-tears of collagen fibers and altered appearance of tendon cells were observed in DG tendons. DG tendons exhibited significantly higher expression of OPN, OCN, SOX9, and Col II and decreased expression of Col I and tenomodulin (TNMD) at week 2. Diabetic TDSCs (dTDSCs) demonstrated significantly decreased proliferation ability and increased osteogenic and chondrogenic differentiation ability. Osteo-chondrogenic markers BMP2, ALP, OPN, OCN, Col II, and SOX9 were also significantly increased while tenogenic markers Col I and TNMD were decreased in dTDSCs. Conclusion These results suggested the erroneous differentiation of dTDSCs might account for the structural and non-tenogenic alternations in DM tendons, which provided new cues for the pathogenesis of tendon disorders in DM. Electronic supplementary material The online version of this article (10.1186/s13287-018-1108-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liu Shi
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Comprehensive Management, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Ying-Juan Li
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Comprehensive Management, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, 87 Ding Jia Qiao, Nanjing, 210009, People's Republic of China
| | - Guang-Chun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Comprehensive Management, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yu-Cheng Lin
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Chen Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Comprehensive Management, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Department of Orthopaedics, Xishan People's Hospital, 588 Guang Rui Road, Wuxi, 214011, Jiangsu, People's Republic of China
| | - Hui Chen
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Comprehensive Management, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China.,School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yun-Feng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China. .,Orthopaedic Trauma Institute, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China. .,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China. .,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Comprehensive Management, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China. .,School of Medicine, Southeast University, No. 87 Ding Jia Qiao, Nanjing, 210009, Jiangsu, People's Republic of China. .,Department of Orthopaedics, Xishan People's Hospital, 588 Guang Rui Road, Wuxi, 214011, Jiangsu, People's Republic of China. .,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, People's Republic of China.
| |
Collapse
|
44
|
Wu L, He S, He Y, Wang X, Lu L. IC-2 Suppresses Proliferation and Induces Apoptosis of Bladder Cancer Cells via the Wnt/β-Catenin Pathway. Med Sci Monit 2018; 24:8074-8080. [PMID: 30415269 PMCID: PMC6240849 DOI: 10.12659/msm.910742] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background The Wnt/β-catenin signaling pathway participates in many important tumorigeneses processes, including bladder cancer. The inhibition of abnormal activation of Wnt pathways might provide a new approach to tumor treatment. In the present study, we investigated the role of IC-2, a novel Wnt pathways small molecular inhibitor, in bladder cancer tumorigenesis. Material/Methods Bladder cancer cells were treated with various concentrations of IC-2 (0–5 μM) in vitro. The proliferation ability was measured using colony formation assay and apoptosis was measured using flow cytometry analysis. The protein expression was detected using Western blot analysis. Xenograft in vivo assay was performed to assess tumor growth. Results IC-2 suppressed the proliferation and aggravated the apoptosis of bladder cancer cells in dose-dependent and time-dependent manners in vitro. Moreover, high concentrations of IC-2 inhibited the Wnt pathway-related protein expression levels, including β-catenin, Cyclin D1, and TCF4. In vivo, administration of IC-2 in xenograft mice decreased the β-catenin expression and reduced the tumor volume. Conclusions Our results validate the tumor-inhibition effect of IC-2 on bladder cancer in vivo and in vitro, providing a novel therapeutic strategy for bladder cancer.
Collapse
Affiliation(s)
- Lingfeng Wu
- Department of Urology, The Frist Hospital of Jiaxing, Frist Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China (mainland)
| | - Shunliang He
- Department of Urology, The Frist Hospital of Jiaxing, Frist Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China (mainland)
| | - Yi He
- Department of Urology, The Frist Hospital of Jiaxing, Frist Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China (mainland)
| | - Xueping Wang
- Department of Urology, The Frist Hospital of Jiaxing, Frist Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China (mainland)
| | - Linfeng Lu
- Department of Urology, The Frist Hospital of Jiaxing, Frist Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China (mainland)
| |
Collapse
|
45
|
Shu CC, Smith MM, Appleyard RC, Little CB, Melrose J. Achilles and tail tendons of perlecan exon 3 null heparan sulphate deficient mice display surprising improvement in tendon tensile properties and altered collagen fibril organisation compared to C57BL/6 wild type mice. PeerJ 2018; 6:e5120. [PMID: 30042881 PMCID: PMC6056265 DOI: 10.7717/peerj.5120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/07/2018] [Indexed: 02/02/2023] Open
Abstract
The aim of this study was to determine the role of the perlecan (Hspg2) heparan sulphate (HS) side chains on cell and matrix homeostasis in tail and Achilles tendons in 3 and 12 week old Hspg2 exon 3 null HS deficient (Hspg2Δ3 − ∕Δ3 −) and C57 BL/6 Wild Type (WT) mice. Perlecan has important cell regulatory and matrix organizational properties through HS mediated interactions with a range of growth factors and morphogens and with structural extracellular matrix glycoproteins which define tissue function and allow the resident cells to regulate tissue homeostasis. It was expected that ablation of the HS chains on perlecan would severely disrupt normal tendon organization and functional properties and it was envisaged that this study would better define the role of HS in normal tendon function and in tendon repair processes. Tail and Achilles tendons from each genotype were biomechanically tested (ultimate tensile stress (UTS), tensile modulus (TM)) and glycosaminoglycan (GAG) and collagen (hydroxyproline) compositional analyses were undertaken. Tenocytes were isolated from tail tendons from each mouse genotype and grown in monolayer culture. These cultures were undertaken in the presence of FGF-2 to assess the cell signaling properties of each genotype. Total RNA was isolated from 3–12 week old tail and Achilles tendons and qRT-PCR was undertaken to assess the expression of the following genes Vcan, Bgn, Dcn, Lum, Hspg2, Ltbp1, Ltbp2, Eln and Fbn1. Type VI collagen and perlecan were immunolocalised in tail tendon and collagen fibrils were imaged using transmission electron microscopy (TEM). FGF-2 stimulated tenocyte monolayers displayed elevated Adamts4, Mmp2, 3, 13 mRNA levels compared to WT mice. Non-stimulated tendon Col1A1, Vcan, Bgn, Dcn, Lum, Hspg2, Ltbp1, Ltbp2, Eln and Fbn1 mRNA levels showed no major differences between the two genotypes other than a decline with ageing while LTBP2 expression increased. Eln expression also declined to a greater extent in the perlecan exon 3 null mice (P < 0.05). Type VI collagen and perlecan were immunolocalised in tail tendon and collagen fibrils imaged using transmission electron microscopy (TEM). This indicated a more compact form of collagen localization in the perlecan exon 3 null mice. Collagen fibrils were also smaller by TEM, which may facilitate a more condensed fibril packing accounting for the superior UTS displayed by the perlecan exon 3 null mice. The amplified catabolic phenotype of Hspg2Δ3 − ∕Δ3 − mice may account for the age-dependent decline in GAG observed in tail tendon over 3 to 12 weeks. After Achilles tenotomy Hspg2Δ3 − ∕Δ3 − and WT mice had similar rates of recovery of UTS and TM over 12 weeks post operatively indicating that a deficiency of HS was not detrimental to tendon repair.
Collapse
Affiliation(s)
- Cindy C Shu
- Raymond Purves Bone and Joint Laboratory, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Margaret M Smith
- Raymond Purves Bone and Joint Laboratory, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Richard C Appleyard
- Murray Maxwell Biomechanics Laboratory, Royal North Shore Hospital, University of Sydney, St. Leonards, New South Wales, Australia.,Surgical Skills Laboratory, Australian School of Advanced Medicine, Macquarie University, Sydney, New South Wales, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Laboratory, Kolling Institute of Medical Research, University of Sydney, Australia.,Sydney Medical School, Northern, University of Sydney, Sydney, Australia
| | - James Melrose
- Raymond Purves Bone and Joint Laboratory, Kolling Institute of Medical Research, University of Sydney, Australia.,Sydney Medical School, Northern, University of Sydney, Sydney, Australia.,Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
46
|
Zhang YJ, Chen X, Li G, Chan KM, Heng BC, Yin Z, Ouyang HW. Concise Review: Stem Cell Fate Guided By Bioactive Molecules for Tendon Regeneration. Stem Cells Transl Med 2018; 7:404-414. [PMID: 29573225 PMCID: PMC5905226 DOI: 10.1002/sctm.17-0206] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022] Open
Abstract
Tendon disorders, which are commonly presented in the clinical setting, disrupt the patients' normal work and life routines, and they damage the careers of athletes. However, there is still no effective treatment for tendon disorders. In the field of tissue engineering, the potential of the therapeutic application of exogenous stem cells to treat tendon pathology has been demonstrated to be promising. With the development of stem cell biology and chemical biology, strategies that use inductive tenogenic factors to program stem cell fate in situ are the most easily and readily translatable to clinical applications. In this review, we focus on bioactive molecules that can potentially induce tenogenesis in adult stem cells, and we summarize the various differentiation factors found in comparative studies. Moreover, we discuss the molecular regulatory mechanisms of tenogenesis, and we examine the various challenges in developing standardized protocols for achieving efficient and reproducible tenogenesis. Finally, we discuss and predict future directions for tendon regeneration. Stem Cells Translational Medicine 2018;7:404-414.
Collapse
Affiliation(s)
- Yan-Jie Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| | - Gang Li
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Kai-Ming Chan
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Boon Chin Heng
- Faculty of Dentistry, Department of Endodontology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Hong-Wei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| |
Collapse
|