1
|
Hammerman M, Pierantoni M, Isaksson H, Eliasson P. Deprivation of loading during rat Achilles tendon healing affects extracellular matrix composition and structure, and reduces cell density and alignment. Sci Rep 2024; 14:23380. [PMID: 39379568 PMCID: PMC11461875 DOI: 10.1038/s41598-024-74783-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
Tendon healing involves mechanosensitive cells that adapt to mechanical stimuli through mechanotransduction, resulting in increased tissue strength. However, detailed insights into this process in response to different loads remain limited. We aimed to investigate how different loading regimes impact the spatial composition of elastin and collagens during Achilles tendon healing. Histological analysis was conducted on healing rat Achilles tendons exposed to (1) full loading, (2) reduced loading, or (3) minimal loading. Histological analysis included Hematoxylin & Eosin and immunohistochemical staining targeting elastin, Collagen 1, Collagen 3, and CD31. Our results showed that the impact of mechanical stimuli on healing tendons varied with the degree of loading. Unexpectedly, minimal loading led to higher staining intensity for collagens and elastin. However, tendons exposed to minimal loading appeared thinner and exhibited a less organized matrix structure, with fewer, less aligned, and more rounded cells. Additionally, our findings indicated an inverse correlation between angiogenesis and load level, with more blood vessels in tendons subjected to less loading. Tissue integrity improved by 12 weeks post-injury, but the healing process continued and did not regain the structure seen in intact tendons even after 20 weeks. This study reveals a load-dependent effect on matrix alignment, cell density, and cell alignment.
Collapse
Affiliation(s)
- Malin Hammerman
- Department of Biomedical Engineering, Lund University, Lund, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maria Pierantoni
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Hanna Isaksson
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Pernilla Eliasson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
- Sahlgrenska University Hospital, Department of Orthopaedics, Mölndal, 341 80, Sweden.
| |
Collapse
|
2
|
Liu X, He L, Wang N, Xie L, Wu B. Bioinformatics analysis and experimental validation of key genes associated with lumbar disc degeneration and biomechanics. Heliyon 2024; 10:e27016. [PMID: 38463775 PMCID: PMC10920361 DOI: 10.1016/j.heliyon.2024.e27016] [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: 09/07/2023] [Revised: 02/01/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024] Open
Abstract
Background Lumbar disc degeneration (LDD) is an important pathological basis for the development of degenerative diseases of the lumbar spine. Most clinical patients have low back pain as their main symptom. The deterioration of the biomechanical environment is an important cause of LDD. Although there is a large amount of basic research on LDD, there are fewer reports that correlate biomechanical mechanisms with basic research. Our research aims to identify 304 key genes involved in LDD due to biomechanical deterioration, using a bioinformatics approach. We focus on SMAD3, CAV1, SMAD7, TGFB1 as hub genes, and screen for 30 potential target drugs, offering novel insights into LDD pathology and treatment options. Methods The Gene Cards, GenCLip3, OMIM and Drugbank databases were explored to obtain genes associated with biomechanics and LDD, followed by making veen plots to obtain both co-expressed genes. GO enrichment analysis and KEGG pathway analysis of the co-expressed genes were obtained using the DAVID online platform and visualised via a free online website. Protein interaction networks (PPI) were obtained through the STRING platform and visualised through Cytoscape 3.9.0. These genes were predicted for downstream interaction networks using the STITCH platform. Then, the GSE56081 dataset was used to validate the key genes. RT-PCR was used to detect mRNA expression of core genes in the degenerated nucleus pulposus (NP) samples and western bolt was used for protein expression. Lastly, the obtained hub genes were searched in the drug database (DGIdb) to find relevant drug candidates. Results From the perspective of biomechanics-induced LDD, we obtained a total of 304 genes, the GO functional enrichment and KEGG pathway enrichment analysis showed that the functions of these genes are mostly related to inflammation and apoptosis. The PPI network was constructed and four Hub genes were obtained through the plug-in of Cytoscape software, namely SMAD3, CAV1, SMAD7 and TGFB1. The analysis of key genes revealed that biomechanical involvement in LDD may be related to the TGF-β signaling pathway. Validation of the GSE56081 dataset revealed that SMAD3 and TGFB1 were highly expressed in degenerating NP samples. RT-PCR results showed that the mRNA expression of SMAD3 and TGFB1 was significantly increased in the severe degeneration group; Western blot results also showed that the protein expression of TGFB1 and P-SMAD3 was significantly increased. In addition, we identified 30 potential drugs. Conclusion This study presented a new approach to investigate the correlation between biomechanical mechanisms and LDD. The deterioration of the biomechanical environment may cause LDD through the TGF-β signaling pathway. TGFB1 and SMAD3 are important core targets. The important genes, pathways and drugs obtained in this study provided a new basis and direction for the study, diagnosis and treatment of LDD.
Collapse
Affiliation(s)
- Xiyu Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Lipeng He
- Department of Spine Surgery, Wuxi Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Wuxi, 214100 China
| | - Nan Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Lin Xie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Bin Wu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
3
|
Little D, Amadio PC, Awad HA, Cone SG, Dyment NA, Fisher MB, Huang AH, Koch DW, Kuntz AF, Madi R, McGilvray K, Schnabel LV, Shetye SS, Thomopoulos S, Zhao C, Soslowsky LJ. Preclinical tendon and ligament models: Beyond the 3Rs (replacement, reduction, and refinement) to 5W1H (why, who, what, where, when, how). J Orthop Res 2023; 41:2133-2162. [PMID: 37573480 PMCID: PMC10561191 DOI: 10.1002/jor.25678] [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: 05/08/2023] [Revised: 07/21/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Several tendon and ligament animal models were presented at the 2022 Orthopaedic Research Society Tendon Section Conference held at the University of Pennsylvania, May 5 to 7, 2022. A key objective of the breakout sessions at this meeting was to develop guidelines for the field, including for preclinical tendon and ligament animal models. This review summarizes the perspectives of experts for eight surgical small and large animal models of rotator cuff tear, flexor tendon transection, anterior cruciate ligament tear, and Achilles tendon injury using the framework: "Why, Who, What, Where, When, and How" (5W1H). A notable conclusion is that the perfect tendon model does not exist; there is no single gold standard animal model that represents the totality of tendon and ligament disease. Each model has advantages and disadvantages and should be carefully considered in light of the specific research question. There are also circumstances when an animal model is not the best approach. The wide variety of tendon and ligament pathologies necessitates choices between small and large animal models, different anatomic sites, and a range of factors associated with each model during the planning phase. Attendees agreed on some guiding principles including: providing clear justification for the model selected, providing animal model details at publication, encouraging sharing of protocols and expertise, improving training of research personnel, and considering greater collaboration with veterinarians. A clear path for translating from animal models to clinical practice was also considered as a critical next step for accelerating progress in the tendon and ligament field.
Collapse
Affiliation(s)
- Dianne Little
- Department of Basic Medical Sciences, The Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Peter C Amadio
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Hani A Awad
- Department of Orthopaedics, Department of Biomedical Engineering, The Center for Musculoskeletal Research, University of Rochester, Rochester, New York, USA
| | - Stephanie G Cone
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew B Fisher
- Joint Department of Biomedical Engineering, College of Engineering, North Carolina State University-University of North Carolina at Chapel Hill, Raleigh, North Carolina, USA
| | - Alice H Huang
- Department of Orthopedic Surgery, Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Drew W Koch
- Department of Clinical Sciences, College of Veterinary Medicine, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Andrew F Kuntz
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rashad Madi
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kirk McGilvray
- Department of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Lauren V Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Snehal S Shetye
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Chunfeng Zhao
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Louis J Soslowsky
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Leong NL, Wu J, Greskovich KE, Li Y, Jiang J. Pdgfrβ + lineage cells transiently increase at the site of Achilles tendon healing. J Orthop Res 2023; 41:1882-1889. [PMID: 36922361 DOI: 10.1002/jor.25552] [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: 02/22/2022] [Revised: 03/01/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
The purpose of this study was to track platelet-derived growth factor receptor-β (Pdgfr-β) lineage cells at the site of Achilles tendon injury over time. Pdgfr-β-CreERT2 :Ai9 mice were generated to track Pdgfr-β lineage cells in adult mice. A surgical Achilles transection injury model was employed to examine the presence of Pdgfr-β lineage cells in the healing tendon over time, with five mice per time point at 3, 7, 14, 28, and 56 days postoperatively. Histology and immunohistochemistry for tdTomato (Pdgfr-β lineage cells), PCNA (proliferating cell nuclear antigen, cell proliferation), and α-SMA (α-smooth muscle actin, myofibroblasts) were performed. The percentage of cells at the healing tendon site staining positive for tdTomato and PCNA were quantified. Over 75% of cells at the injury site were Pdgfr-β lineage cells at Days 3, 7, and 14, and this percentage decreased significantly by Days 28 and 56 postinjury. Cell proliferation at the injury site peaked on Day 7 and decreased thereafter. Immunohistochemistry for α-SMA demonstrated minimal colocalization of myofibroblasts with Pdgfr-β lineage cells. This study demonstrates that in a mouse model of Achilles tendon injury, Pdgfr-β lineage cells' presence at the injury site is transient. Thus, we conclude that they are unlikely to be the cells that differentiate into myofibroblasts and directly contribute to tendon fibrous scar formation. Clinical Significance: This study provides some insight into the presence of Pdgfr-β lineage cells (including pericytes) following Achilles injury, furthering our understanding of tendon healing.
Collapse
Affiliation(s)
- Natalie L Leong
- Baltimore VA Medical Center, VA Maryland Healthcare System, Baltimore, Maryland, USA
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jocelyn Wu
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kathryn E Greskovich
- Baltimore VA Medical Center, VA Maryland Healthcare System, Baltimore, Maryland, USA
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yang Li
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jie Jiang
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Wang C, Jiang Z, Pang R, Zhang H, Li H, Li Z. Global trends in research of achilles tendon injury/rupture: A bibliometric analysis, 2000–2021. Front Surg 2023; 10:1051429. [PMID: 37051567 PMCID: PMC10083236 DOI: 10.3389/fsurg.2023.1051429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/10/2023] [Indexed: 03/28/2023] Open
Abstract
BackgroundThe Achilles tendon is the strongest and most susceptible tendon in humans. Achilles tendon injuries and ruptures have gradually attracted research attention. However, a bibliometric analysis of global research in this field is lacking. This study involved a bibliometric analysis of the developmental trends and research hotspots in Achilles tendon injuries/ruptures from 2000 to 2021.MethodsArticles published between 2001 and 2021 were retrieved from an extended database of the Science Citation Index using Web of Science. VOSviewer and CiteSpace were used to analyze the relationships between publications, countries, institutions, journals, authors, references, and keywords.ResultsThis study included 3,505 studies of 73 countries, 3,274 institutions, and 12,298 authors and explored the cooperation between them and the relationships between citations. Over the past 22 years, the number of publications has significantly increased. Foot Ankle International has published the most papers on Achilles tendon injuries/ruptures, and British Journal of Sports Medicine is the most famous journal. Re-rupture, exosomes, acute Achilles tendon rupture, and tendon adhesions gradually become the research focus over the past few years.ConclusionAchilles tendon injury and rupture are important research topics. A vast number of newly published papers on this topic have demonstrated that clinicians and researchers are interested in their study. Over time, these recent studies will be widely cited; therefore, this bibliometric analysis should be constantly updated.
Collapse
Affiliation(s)
- Chenguang Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaohui Jiang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ran Pang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huafeng Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Chinese & Western Medicine Hospital, Tianjin, China
- Correspondence: Zhijun Li Hui Li
| | - Zhijun Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Correspondence: Zhijun Li Hui Li
| |
Collapse
|
6
|
Freedman BR, Kuttler A, Beckmann N, Nam S, Kent D, Schuleit M, Ramazani F, Accart N, Rock A, Li J, Kurz M, Fisch A, Ullrich T, Hast MW, Tinguely Y, Weber E, Mooney DJ. Enhanced tendon healing by a tough hydrogel with an adhesive side and high drug-loading capacity. Nat Biomed Eng 2022; 6:1167-1179. [PMID: 34980903 PMCID: PMC9250555 DOI: 10.1038/s41551-021-00810-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 09/13/2021] [Indexed: 12/14/2022]
Abstract
Hydrogels that provide mechanical support and sustainably release therapeutics have been used to treat tendon injuries. However, most hydrogels are insufficiently tough, release drugs in bursts, and require cell infiltration or suturing to integrate with surrounding tissue. Here we report that a hydrogel serving as a high-capacity drug depot and combining a dissipative tough matrix on one side and a chitosan adhesive surface on the other side supports tendon gliding and strong adhesion (larger than 1,000 J m-2) to tendon on opposite surfaces of the hydrogel, as we show with porcine and human tendon preparations during cyclic-friction loadings. The hydrogel is biocompatible, strongly adheres to patellar, supraspinatus and Achilles tendons of live rats, boosted healing and reduced scar formation in a rat model of Achilles-tendon rupture, and sustainably released the corticosteroid triamcinolone acetonide in a rat model of patellar tendon injury, reducing inflammation, modulating chemokine secretion, recruiting tendon stem and progenitor cells, and promoting macrophage polarization to the M2 phenotype. Hydrogels with 'Janus' surfaces and sustained-drug-release functionality could be designed for a range of biomedical applications.
Collapse
Affiliation(s)
- Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Andreas Kuttler
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Sungmin Nam
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Daniel Kent
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | | | | | - Nathalie Accart
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Anna Rock
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jianyu Li
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada
| | - Markus Kurz
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Andreas Fisch
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Thomas Ullrich
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Michael W Hast
- Biedermann Lab for Orthopaedic Research, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Yann Tinguely
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Eckhard Weber
- Novartis Institutes for Biomedical Research, Basel, Switzerland.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
7
|
Conrad B, Yang F. Hydroxyapatite-coated gelatin microribbon scaffolds induce rapid endogenous cranial bone regeneration in vivo. BIOMATERIALS ADVANCES 2022; 140:213050. [PMID: 35917686 DOI: 10.1016/j.bioadv.2022.213050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/25/2022] [Accepted: 07/24/2022] [Indexed: 06/15/2023]
Abstract
Hydroxyapatite (HA) has a composition similar to mineral bone and has been used for coating macroporous scaffolds to enhance bone formation. However, previous macroporous scaffolds did not support minimally invasive delivery. Our lab has reported on gelatin-based microribbon (μRB) shaped hydrogels, which combine injectability with macroporosity and support cranial bone formation in an immunocompromised mouse model. However, gelatin alone was not sufficient to support cranial bone formation in immunocompetent animals. To overcome this challenge, here we evaluated two methods to incorporate HA into gelatin μRB scaffolds using either modified simulated body fluid (mSBF) or commercially available HA nanoparticles (HAnp). HA incorporation and distribution were characterized using scanning electron microscopy and energy-dispersive X-ray spectroscopy. While both methods enhanced MSC osteogenesis and mineralization, the mSBF method led to undesirable reduction in mechanical properties. HAnp-coated μRB scaffolds were further evaluated in an immunocompetent mouse cranial defect model. Acellular HAnp-coated gelatin μRB scaffolds induced rapid and robust endogenous cranial bone regeneration as shown by MicroCT imaging and histology. Co-delivery with exogenous MSCs led to later bone resorption accompanied by increased osteoclast activity. In summary, our results demonstrate the promise of gelatin μRBs with HAnps as a promising therapy for cranial bone regeneration without the need for exogenous cells or growth factors.
Collapse
Affiliation(s)
- Bogdan Conrad
- Program of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 240 Pasteur Dr., Biomedical Innovation Building 1200, Palo Alto, CA 94304, United States of America.
| | - Fan Yang
- Departments of Orthopaedic Surgery and Bioengineering Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1200, Palo Alto, CA 94304, United States of America.
| |
Collapse
|
8
|
Izumi S, Oichi T, Shetye SS, Zhang K, Wilson K, Iwamoto M, Kuo CK, Akabudike N, Adachi N, Soslowsky LJ, Enomoto-Iwamoto M. Inhibition of glucose use improves structural recovery of injured Achilles tendon in mice. J Orthop Res 2022; 40:1409-1419. [PMID: 34460123 PMCID: PMC8882710 DOI: 10.1002/jor.25176] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/21/2021] [Accepted: 08/16/2021] [Indexed: 02/04/2023]
Abstract
Injured tendons do not regain their native structure except at fetal or very young ages. Healing tendons often show mucoid degeneration involving accumulation of sulfated glycosaminoglycans (GAGs), but its etiology and molecular base have not been studied substantially. We hypothesized that quality and quantity of gene expression involving the synthesis of proteoglycans having sulfated GAGs are altered in injured tendons and that a reduction in synthesis of sulfated GAGs improves structural and functional recovery of injured tendons. C57BL6/j mice were subjected to Achilles tendon tenotomy surgery. The injured tendons accumulated sulfate proteoglycans as early as 1-week postsurgery and continued so by 4-week postsurgery. Transcriptome analysis revealed upregulation of a wide range of proteoglycan genes that have sulfated GAGs in the injured tendons 1 and 3 weeks postsurgery. Genes critical for enzymatic reaction of initiation and elongation of chondroitin sulfate GAG chains were also upregulated. After the surgery, mice were treated with the 2-deoxy-d-glucose (2DG) that inhibits conversion of glucose to glucose-6-phosphate, an initial step of glucose metabolism as an energy source and precursors of monosaccharides of GAGs. The 2DG treatment reduced accumulation of sulfated proteoglycans, improved collagen fiber alignment, and reduced the cross-sectional area of the injured tendons. The modulus of the 2DG-treated groups was higher than that in the vehicle group, but not of statistical significance. Our findings suggest that mucoid degeneration in injured tendons may result from the upregulated expression of genes involved the synthesis of sulfate proteoglycans and can be inhibited by reduction of glucose utilization.
Collapse
Affiliation(s)
- Soutarou Izumi
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health sciences, Hiroshima University, Japan
| | - Takeshi Oichi
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore
| | - Snehal S. Shetye
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia PA
| | - Kairui Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University
| | - Kimberly Wilson
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore
| | - Masahiro Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore
| | - Catherine K. Kuo
- Fischell Department of Bioengineering, University of Maryland College Park
| | - Ngozi Akabudike
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health sciences, Hiroshima University, Japan
| | - Louis J. Soslowsky
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia PA
| | | |
Collapse
|
9
|
Enthesis Healing Is Dependent on Scaffold Interphase Morphology—Results from a Rodent Patellar Model. Cells 2022; 11:cells11111752. [PMID: 35681447 PMCID: PMC9179925 DOI: 10.3390/cells11111752] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
The use of multiphasic scaffolds to treat injured tendon-to-bone entheses has shown promising results in vitro. Here, we used two versions of a biphasic silk fibroin scaffold to treat an enthesis defect created in a rat patellar model in vivo. One version presented a mixed transition between the bony and the tendon end of the construct (S-MT) while this transition was abrupt in the second version (S-AT). At 12 weeks after surgery, the S-MT scaffold promoted better healing of the injured enthesis, with minimal undesired ossification of the insertion area. The expression of tenogenic and chondrogenic markers was sustained for longer in the S-MT-treated group and the tangent modulus of the S-MT-treated samples was similar to the native tissue at 12 weeks while that of the S-AT-treated enthesis was lower. Our study highlights the important role of the transition zone of multiphasic scaffolds in the treatment of complex interphase tissues such as the tendon-to-bone enthesis.
Collapse
|
10
|
Han L, Liu H, Fu H, Hu Y, Fang W, Liu J. Exosome-delivered BMP-2 and polyaspartic acid promotes tendon bone healing in rotator cuff tear via Smad/RUNX2 signaling pathway. Bioengineered 2022; 13:1459-1475. [PMID: 35258414 PMCID: PMC8805918 DOI: 10.1080/21655979.2021.2019871] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/13/2021] [Indexed: 11/02/2022] Open
Abstract
Rotator cuff tear is the main form of shoulder joint injury, which seriously affects shoulder joint function. This study aimed to clarify the function and mechanism of exosomes containing polylactic acid (PLA), polylactic acid copolymer and BMP-2 in tendon bone healing of rotator cuff tear. First, CD44 expression in bone marrow mesenchymal stem cells (BMSCs) and CD90 and CD44 in exosomes were analyzed by flow cytometry. Then, stability and targeting identification of exosome-delivered bone morphogenetic protein (BMP)-2 and PLA microcapsules were measured by transmission electron microscopy (TEM), DiO/DiI staining. Finally, tendon-bone repair after acute rotator cuff rupture in rabbits was established, and the function of BMP-2 exosomes for tendon bone healing in rotator cuff tear was evaluated by micro-CT, biomechanical determination and histochemical staining methods. The results showed that the exosomes of polyaspartic acid-polylactic acid-glycolic acid copolymer (PASP-PLGA) microcapsules were successfully established which showed good stability and targeting. The bone mineral density (BMD), tissue mineral density (TMD) and bone volume fraction (BV/TV) were higher, while the stiffness and the ultimate load strength of the tendon interface were enhanced under treatment with exosomes of PASP-PLGA microcapsules. Histochemical staining showed that exosomes of PASP-PLGA microcapsules promoted tendon and bone interface healing after rotator cuff injury. The tendon regeneration- and cartilage differentiation-related protein expressions were significantly upregulated under treatment with exosomes of PASP-PLGA microcapsules. In conclusion, exosome-delivered BMP-2 and PLA promoted tendon bone healing in rotator cuff tear via Smad/RUNX2 pathway. Our findings may provide a new insight for promoting tendon healing.
Collapse
Affiliation(s)
- Lei Han
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Hong Liu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Huajun Fu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Yugen Hu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Weili Fang
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| | - Junsheng Liu
- Department of Orthopaedics Institute, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Fang F, Sup M, Luzzi A, Ferrer X, Thomopoulos S. Hedgehog signaling underlying tendon and enthesis development and pathology. Matrix Biol 2022; 105:87-103. [PMID: 34954379 PMCID: PMC8821161 DOI: 10.1016/j.matbio.2021.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 02/08/2023]
Abstract
Hedgehog (Hh) signaling has been widely acknowledged to play essential roles in many developmental processes, including endochondral ossification and growth plate maintenance. Furthermore, a rising number of studies have shown that Hh signaling is necessary for tendon enthesis development. Specifically, the well-tuned regulation of Hh signaling during development drives the formation of a mineral gradient across the tendon enthesis fibrocartilage. However, aberrant Hh signaling can also lead to pathologic heterotopic ossification in tendon or osteophyte formation at the enthesis. Therefore, the therapeutic potential of Hh signaling modulation for treating tendon and enthesis diseases remains uncertain. For example, increased Hh signaling may enhance tendon-to-bone healing by promoting the formation of mineralized fibrocartilage at the healing interface, but pathologic heterotopic ossification may also be triggered in the adjacent tendon. Further work is needed to elucidate the distinct functions of Hh signaling in the tendon and enthesis to support the development of therapies that target the pathway.
Collapse
Affiliation(s)
- Fei Fang
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States
| | - McKenzie Sup
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Andrew Luzzi
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States
| | - Xavier Ferrer
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States; Department of Biomedical Engineering, Columbia University, New York, NY, United States.
| |
Collapse
|
12
|
Liang Q, Lu Y, Yu L, Zhu Q, Xie W, Wang Y, Ye L, Li Q, Liu S, Liu Y, Zhu C. Disruption of the mouse Bmal1 locus promotes heterotopic ossification with aging via TGF-beta/BMP signaling. J Bone Miner Metab 2022; 40:40-55. [PMID: 34626248 DOI: 10.1007/s00774-021-01271-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/12/2021] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Heterotopic ossification of tendons and ligaments is a painful and debilitating disease with no effective treatment. Although aging has been reported to be correlated with the occurrence and development of this disease, the mechanism remains unknown. MATERIALS AND METHODS In the present study, we generated Bmal1-/- mice, which disrupted the circadian clock and displayed premature aging, as an aging model to explore the role of Bmal1 in TGF-beta (β)/BMP signaling in progressive heterotopic ossification of tendons and ligaments with aging. RESULTS We first confirmed that BMAL1 expression is downregulated in human fibroblasts from ossification of the posterior longitudinal ligament using online datasets. Bmal1 deficiency in mice caused significantly progressive heterotopic ossification with aging starting at week 6, notably in the Achilles tendons and posterior longitudinal ligaments. Ossification of the Achilles tendons was accompanied by progressive motor dysfunction of the ankle joint. Histology and immunostaining showed markedly increased endochondral ossification in the posterior longitudinal ligaments and Achilles tendons of Bmal1-/- mice. Ligament-derived Bmal1-/- fibroblasts showed an osteoblast-like phenotype, upregulated osteogenic and chondrogenic markers, and activated TGFβ/BMP signaling, which was enhanced by TGFβ1 stimulation. Furthermore, Bmal1-/- mouse embryonic fibroblasts had a stronger potential for osteogenic differentiation with activation of TGFβ/BMP signaling. CONCLUSIONS These findings demonstrated that Bmal1 negatively regulates endochondral ossification in heterotopic ossification of tendons and ligaments with aging via TGFβ/BMP signaling, thereby identifying a new regulatory mechanism in age-related heterotopic ossification of tendons and ligaments.
Collapse
Affiliation(s)
- Qian Liang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yingsi Lu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Lu Yu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Qingqing Zhu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wenlin Xie
- Department of Pathology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yun Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Liping Ye
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Qiji Li
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Shaoyu Liu
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yan Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Chengming Zhu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
13
|
Ali OJ, Ehrle A, Comerford EJ, Canty-Laird EG, Mead A, Clegg PD, Maddox TW. Intrafascicular chondroid-like bodies in the ageing equine superficial digital flexor tendon comprise glycosaminoglycans and type II collagen. J Orthop Res 2021; 39:2755-2766. [PMID: 33580534 DOI: 10.1002/jor.25002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/24/2020] [Accepted: 01/29/2021] [Indexed: 02/04/2023]
Abstract
The superficial digital flexor tendon (SDFT) is considered functionally equivalent to the human Achilles tendon. Circular chondroid depositions scattered amongst the fascicles of the equine SDFT are rarely reported. The purpose of this study was the detailed characterization of intrafascicular chondroid-like bodies (ICBs) in the equine SDFT, and the assessment of the effect of ageing on the presence and distribution of these structures. Ultrahigh field magnetic resonance imaging (9.4T) series of SDFT samples of young (1-9 years) and aged (17-25 years) horses were obtained, and three-dimensional reconstruction of ICBs was performed. Morphological evaluation of the ICBs included histology, immunohistochemistry and transmission electron microscopy. The number, size, and position of ICBs was determined and compared between age groups. There was a significant difference (p = .008) in the ICB count between young and old horses with ICBs present in varying number (13-467; median = 47, mean = 132.6), size and distribution in the SDFT of aged horses only. There were significantly more ICBs in the tendon periphery when compared with the tendon core region (p = .010). Histological characterization identified distinctive cells associated with increased glycosaminoglycan and type II collagen extracellular matrix content. Ageing and repetitive strain frequently cause tendon micro-damage before the development of clinical tendinopathy. Documentation of the presence and distribution of ICBs is a first step towards improving our understanding of the impact of these structures on the viscoelastic properties, and ultimately their effect on the risk of age-related tendinopathy in energy-storing tendons.
Collapse
Affiliation(s)
- Othman J Ali
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Department of Surgery and Theriogenology, College of Veterinary Medicine, University of Sulaimani, Sulaymaniyah, Sulaymaniyah, Iraq.,Department of Medical Laboratory Science, Komar University of Science and Technology, Sulaymaniyah, Kurdistan Region, Iraq
| | - Anna Ehrle
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Eithne J Comerford
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK.,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Elizabeth G Canty-Laird
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Ashleigh Mead
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
| | - Peter D Clegg
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK.,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Thomas W Maddox
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
| |
Collapse
|
14
|
Extensive Ossification of the Achilles Tendon with and without Acute Fracture: A Scoping Review. J Clin Med 2021; 10:jcm10163480. [PMID: 34441776 PMCID: PMC8396855 DOI: 10.3390/jcm10163480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022] Open
Abstract
Extensive Ossification of the Achilles Tendon (EOAT) is an uncommon condition characterized by the presence of heterotopic ossification within the substance of the Achilles Tendon and is distinct from other tendinopathies associated with tendon mineralization. The purpose of this scoping review of the literature on EOAT is to describe the pathogenesis, patient population, presentation, management, and outcomes of this rare condition. Fifty-four articles were included in the scoping review after screening and selection. According to the literature, EOAT often presents with pain and swelling around the Achilles Tendon and is frequently associated with acute trauma. EOAT is more common in men, and although the exact mechanisms of the pathology are not fully understood, EOAT may demonstrate specific molecular signaling patterns. The lack of knowledge regarding the molecular mechanism may be a significant hindrance to the management of the condition. Even though a standard treatment regimen for EOAT does not exist, conservative management for six months in patients without complications is recommended. Those who have an acute fracture of the ossification should be managed more aggressively and will often require surgical repair with autograft, although there is no standardized procedure at this time. Clinicians should be aware of the typical presentation, risk factors, and management options of patients with EOAT. Additionally, they should be cautious when selecting treatment strategies and conduct a thorough evaluation of long-term outcomes with various treatment modalities, which this review provides. Most important, this review highlights the need for further research to determine the best course of clinical treatment of EOAT injuries, in order to establish a standard treatment regimen.
Collapse
|
15
|
Howell KL, Kaji DA, Li TM, Montero A, Yeoh K, Nasser P, Huang AH. Macrophage depletion impairs neonatal tendon regeneration. FASEB J 2021; 35:e21618. [PMID: 33982337 DOI: 10.1096/fj.202100049r] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022]
Abstract
Tendons are dense connective tissues that transmit muscle forces to the skeleton. After adult injury, healing potential is generally poor and dominated by scar formation. Although the immune response is a key feature of healing, the specific immune cells and signals that drive tendon healing have not been fully defined. In particular, the immune regulators underlying tendon regeneration are almost completely unknown due to a paucity of tendon regeneration models. Using a mouse model of neonatal tendon regeneration, we screened for immune-related markers and identified upregulation of several genes associated with inflammation, macrophage chemotaxis, and TGFβ signaling after injury. Depletion of macrophages using AP20187 treatment of MaFIA mice resulted in impaired functional healing, reduced cell proliferation, reduced ScxGFP+ neo-tendon formation, and altered tendon gene expression. Collectively, these results show that inflammation is a key component of neonatal tendon regeneration and demonstrate a requirement for macrophages in effective functional healing.
Collapse
Affiliation(s)
- Kristen L Howell
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepak A Kaji
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas M Li
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angela Montero
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenji Yeoh
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip Nasser
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
16
|
Yang Y, Wu Y, Zhou K, Wu D, Yao X, Heng BC, Zhou J, Liu H, Ouyang H. Interplay of Forces and the Immune Response for Functional Tendon Regeneration. Front Cell Dev Biol 2021; 9:657621. [PMID: 34150755 PMCID: PMC8213345 DOI: 10.3389/fcell.2021.657621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/26/2021] [Indexed: 01/11/2023] Open
Abstract
Tendon injury commonly occurs during sports activity, which may cause interruption or rapid decline in athletic career. Tensile strength, as one aspect of tendon biomechanical properties, is the main parameter of tendon function. Tendon injury will induce an immune response and cause the loss of tensile strength. Regulation of mechanical forces during tendon healing also changes immune response to improve regeneration. Here, the effects of internal/external forces and immune response on tendon regeneration are reviewed. The interaction between immune response and internal/external forces during tendon regeneration is critically examined and compared, in relation to other tissues. In conclusion, it is essential to maintain a fine balance between internal/external forces and immune response, to optimize tendon functional regeneration.
Collapse
Affiliation(s)
- Yuwei Yang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yicong Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongmei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xudong Yao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Boon Chin Heng
- Central Laboratories, School of Stomatology, Peking University, Beijing, China
| | - Jing Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| |
Collapse
|
17
|
Kusano T, Nakatani M, Ishiguro N, Ohno K, Yamamoto N, Morita M, Yamada H, Uezumi A, Tsuchida K. Desloratadine inhibits heterotopic ossification by suppression of BMP2-Smad1/5/8 signaling. J Orthop Res 2021; 39:1297-1304. [PMID: 32043642 DOI: 10.1002/jor.24625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 01/30/2020] [Indexed: 02/04/2023]
Abstract
Heterotopic ossification (HO) is a pathological condition in which ectopic bone forms within soft tissues such as skeletal muscle. Human platelet-derived growth factor receptor α positive (PDGFRα+) cells, which were proved to be the original cells of HO were incubated in osteogenic differentiation medium with Food and Drug Administration-approved compounds. Alkaline phosphatase activity was measured as a screening to inhibit osteogenic differentiation. For the compounds which inhibited osteogenic differentiation of PDGFRα+ cells, we examined dose dependency of its effect using alizarin red S staining and its cell toxicity using WST-8. In addition, regulation of bone morphogenetic proteins (BMP)-Smad signaling which is the major signal of osteogenic differentiation was investigated by Western blotting to elucidate the mechanism of osteogenesis inhibitory effect by the compound. In vivo experiment, complete transverse incision of Achilles tendons in mice was made and mice were fed the compound by mixing with drinking water after operation. Ten weeks after operation, we assessed and quantified HO by micro-computed tomography scan. Intriguingly, we discovered desloratadine inhibited osteogenic differentiation of PDGFRα+ cells using the drug repositioning method. Desloratadine inhibited osteogenic differentiation of the cells dose dependently without cell toxicity. Desloratadine suppressed phosphorylation of Smad1/5/8 induced by BMP2 in PDGFRα+ cells. In Achilles tenotomy mice model, desloratadine treatment significantly inhibited ectopic bone formation compared with control. In conclusion, we discovered desloratadine inhibited osteogenic differentiation using human PDGFRα+ cells and proved its efficacy using Achilles tenotomy ectopic bone formation model in vivo. Our study paved the way to inhibit HO in early clinical use because of its guaranteed safety.
Collapse
Affiliation(s)
- Taiki Kusano
- Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan.,Division of Neurological Diseases and Cancer, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Masashi Nakatani
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Naoki Ishiguro
- Department of Orthopaedic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurological Diseases and Cancer, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Naoki Yamamoto
- Center for Joint Research Facilities Support, Fujita Health University, Research Promotion and Support Headquarters, Toyoake, Japan
| | - Mitsuhiro Morita
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake, Japan
| | - Harumoto Yamada
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake, Japan
| | - Akiyoshi Uezumi
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| |
Collapse
|
18
|
Michel PA, Kronenberg D, Neu G, Stolberg-Stolberg J, Frank A, Pap T, Langer M, Fehr M, Raschke MJ, Stange R. Microsurgical reconstruction affects the outcome in a translational mouse model for Achilles tendon healing. J Orthop Translat 2020; 24:1-11. [PMID: 32489862 PMCID: PMC7260609 DOI: 10.1016/j.jot.2020.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/17/2020] [Accepted: 04/08/2020] [Indexed: 01/02/2023] Open
Abstract
Background Animal models are one of the first steps in translation of basic science findings to clinical practice. For tendon healing research, transgenic mouse models are important to advance therapeutic strategies. However, the small size of the structures complicates surgical approaches, histological assessment, and biomechanical testing. In addition, available models are not standardized and difficult to compare. How surgery itself affects the healing outcome has not been investigated yet. The focus of the study was to develop a procedure that includes a transection and microsurgical reconstruction of the Achilles tendon but, unlike other models, preserves the sciatic nerve. We wanted to examine how distinct parts of the technique influenced healing. Methods For this animal model study, we used 96 wild-type male C57BL/6 mice aged 8–12 weeks. We evaluated different suture techniques and macroscopically confirmed the optimal combination of suture material and technique to minimize tendon gap formation. A key element is the detailed, step-by-step illustration of the surgery. In addition, we assessed histological (Herovici and Alcian blue staining) outcome parameters at 1–16 weeks postoperatively. Microcomputed tomography (micro-CT) was performed to measure the bone volume of heterotopic ossifications (HOs). Biomechanical analyses were carried out using a viscoelastic protocol on the biomechanical testing machine LM1. Results A modified 4-strand suture combined with a cerclage for immobilization without transection of the sciatic nerve reliably eliminated gap formation. The maximal dorsal extension of the hindlimb at the upper ankle joint from the equinus position (limited by the immobilization cerclage) increased over time postoperatively (operation: 28.8 ± 2.2°; 1 week: 54 ± 36°; 6 weeks: 80 ± 11.7°; 16 weeks: 96 ± 15.8°, p > 0.05). Histological staining revealed a maturation of collagen fibres within 6 weeks, whereas masses of cartilage were visible throughout the healing period. Micro-CT scans detected the development of HOs starting at 4 weeks and further progression at 6 and 16 weeks (bone volume, 4 weeks: 0.07604 ± 0.05286 mm3; 6 weeks: 0.50682 ± 0.68841 mm3; 16 weeks: 2.36027 ± 0.85202 mm3, p > 0.001). In-depth micro-CT analysis of the different surgical elements revealed that an injury of the tendon is a key factor for the development of HOs. Immobilization alone does not trigger HOs. Biomechanical properties of repaired tendons were greatly altered and remained inferior 6 weeks after surgery. Conclusion With this study, we demonstrated that the microsurgical technique greatly influences the short- and longer-term healing outcome. When the sciatic nerve is preserved, the best surgical reconstruction of the tendon defect is achieved by a 4-strand core suture in combination with a tibiofibular cerclage for postoperative immobilization. The cerclage promotes a gradual increase in the range of motion of the upper ankle joint, comparable with an early mobilization rehabilitation protocol. HO, as a key mechanism for poor tendon healing, is progressive and can be monitored early in the model. The translational potential of this article The study enhances the understanding of model dependent factors of healing. The described reconstruction technique provides a reproducible and translational rodent model for future Achilles tendon healing research. In combination with transgenic strains, it can be facilitated to advance therapeutic strategies to improve the clinical results of tendon injuries.
Collapse
Affiliation(s)
- Philipp A Michel
- Department of Trauma, Hand- and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Daniel Kronenberg
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, Westfaelische Wilhelms University Muenster, Muenster, Germany
| | - Gertje Neu
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Josef Stolberg-Stolberg
- Department of Trauma, Hand- and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Andre Frank
- Department of Trauma, Hand- and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Thomas Pap
- Institute of Musculoskeletal Medicine, Westfaelische Wilhelms University Muenster, Muenster, Germany
| | - Martin Langer
- Department of Trauma, Hand- and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Michael Fehr
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Michael J Raschke
- Department of Trauma, Hand- and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, Westfaelische Wilhelms University Muenster, Muenster, Germany
| |
Collapse
|
19
|
Kang X, Tian B, Zhang L, Ge Z, Zhao Y, Zhang Y. Relationship of common variants in MPP7, TIMP2 and CASP8 genes with the risk of chronic achilles tendinopathy. Sci Rep 2019; 9:17627. [PMID: 31772230 PMCID: PMC6879592 DOI: 10.1038/s41598-019-54097-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/08/2019] [Indexed: 11/08/2022] Open
Abstract
Previous etiologic studies have indicated that both environmental and genetic factors play important roles in the occurrence and development of chronic Achilles tendinopathy (AT). A recent study documented the results of the largest genome-wide association study for chronic AT to date, indicating that MPP7, TIMP2 and CASP8 may be involved in the occurrence and development of chronic AT. In this study, we aimed to investigate whether MPP7, TIMP2 and CASP8 were associated with susceptibility to chronic AP in a Han Chinese population. A total of 3,680 study subjects comprised 1,288 chronic AT cases, and 2,392 healthy controls were recruited. Forty-four tag SNPs (7 from CASP8, 20 from MPP7, and 17 from TIMP2) were genotyped in the study. Genetic association analyses were performed at both single marker and haplotype levels. Functional consequences of significant SNPs were examined in the RegulomeDB and GTEx databases. Two SNPs, SNP rs1937810 (OR [95%CI] = 1.20 [1.09-1.32], χ2 = 13.50, P = 0.0002) in MPP7 and rs4789932 (OR [95%CI] = 1.24 [1.12-1.37], χ2 = 17.98, P = 2.23 × 10-5) in TIMP2, were significantly associated with chronic AT. Significant eQTL signals for SNP rs4789932 on TIMP2 were identified in human heart and artery tissues. Our results provide further supportive evidence for the association of the TIMP2 and MPP7 genes with chronic AT, which supports important roles for TIMP2 and MPP7 in the etiology of chronic AT, adding to the current understanding of the susceptibility of chronic AT.
Collapse
Affiliation(s)
- Xin Kang
- Department of Orthopedics, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bin Tian
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Liang Zhang
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhaogang Ge
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yang Zhao
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yingang Zhang
- Department of Orthopedics, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China.
| |
Collapse
|
20
|
de Girolamo L, Morlin Ambra LF, Perucca Orfei C, McQuilling JP, Kimmerling KA, Mowry KC, Johnson KA, Phan AT, Whited JL, Gomoll AH. Treatment with Human Amniotic Suspension Allograft Improves Tendon Healing in a Rat Model of Collagenase-Induced Tendinopathy. Cells 2019; 8:E1411. [PMID: 31717431 PMCID: PMC6912389 DOI: 10.3390/cells8111411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022] Open
Abstract
Treatment of tendon injuries is challenging, with neither conservative nor surgical approaches providing full recovery. Placental-derived tissues represent a promising tool for the treatment of tendon injuries. In this study, human amniotic suspension allograft (ASA) was investigated in a pre-clinical model of Achilles tendinopathy. Collagenase type I was injected in the right hind limb of Sprague Dawley rats to induce disease. Contralateral tendons were either left untreated or injected with saline as controls. Seven days following induction, tendons were injected with saline, ASA, or left untreated. Rats were sacrificed 14 and 28 days post-treatment. Histological and biomechanical analysis of tendons was completed. Fourteen days after ASA injection, improved fiber alignment and reduced cell density demonstrated improvement in degenerated tendons. Twenty-eight days post-treatment, tendons in all treatment groups showed fewer signs of degeneration, which is consistent with normal tendon healing. No statistically significant differences in histological or biomechanical analyses were observed between treatment groups at 28 days independent of the treatment they received. In this study, ASA treatment was safe, well-tolerated, and resulted in a widespread improvement of the tissue. The results of this study provide preliminary insights regarding the potential use of ASA for the treatment of Achilles tendinopathy.
Collapse
Affiliation(s)
- Laura de Girolamo
- IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Luiz Felipe Morlin Ambra
- University Hospital São Paulo, Av. Prof. Lineu Prestes, 2565-Butantã, São Paulo, SP 05508-000, Brazil;
| | | | - John P. McQuilling
- Organogenesis, 2641 Rocky Ridge Lane, Birmingham, AL 35216, USA; (J.P.M.); (K.A.K.); (K.C.M.)
| | - Kelly A. Kimmerling
- Organogenesis, 2641 Rocky Ridge Lane, Birmingham, AL 35216, USA; (J.P.M.); (K.A.K.); (K.C.M.)
| | - Katie C. Mowry
- Organogenesis, 2641 Rocky Ridge Lane, Birmingham, AL 35216, USA; (J.P.M.); (K.A.K.); (K.C.M.)
| | - Kimberly A. Johnson
- Harvard Medical School, the Harvard Stem Cell Institute, and Department of Orthopedic Surgery, Brigham and Women’s Hospital, 7 Divinity Avenue, Cambridge, MA 02138, USA; (K.A.J.); (A.T.P.); (J.L.W.)
| | - Amy T. Phan
- Harvard Medical School, the Harvard Stem Cell Institute, and Department of Orthopedic Surgery, Brigham and Women’s Hospital, 7 Divinity Avenue, Cambridge, MA 02138, USA; (K.A.J.); (A.T.P.); (J.L.W.)
| | - Jessica L. Whited
- Harvard Medical School, the Harvard Stem Cell Institute, and Department of Orthopedic Surgery, Brigham and Women’s Hospital, 7 Divinity Avenue, Cambridge, MA 02138, USA; (K.A.J.); (A.T.P.); (J.L.W.)
| | - Andreas H. Gomoll
- Harvard Medical School, the Harvard Stem Cell Institute, and Department of Orthopedic Surgery, Brigham and Women’s Hospital, 7 Divinity Avenue, Cambridge, MA 02138, USA; (K.A.J.); (A.T.P.); (J.L.W.)
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| |
Collapse
|
21
|
Kurtaliaj I, Golman M, Abraham AC, Thomopoulos S. Biomechanical Testing of Murine Tendons. J Vis Exp 2019:10.3791/60280. [PMID: 31680671 PMCID: PMC7217614 DOI: 10.3791/60280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tendon disorders are common, affect people of all ages, and are often debilitating. Standard treatments, such as anti-inflammatory drugs, rehabilitation, and surgical repair, often fail. In order to define tendon function and demonstrate efficacy of new treatments, the mechanical properties of tendons from animal models must be accurately determined. Murine animal models are now widely used to study tendon disorders and evaluate novel treatments for tendinopathies; however, determining the mechanical properties of mouse tendons has been challenging. In this study, a new system was developed for tendon mechanical testing that includes 3D-printed fixtures that exactly match the anatomies of the humerus and calcaneus to mechanically test supraspinatus tendons and Achilles tendons, respectively. These fixtures were developed using 3D reconstructions of native bone anatomy, solid modeling, and additive manufacturing. The new approach eliminated artifactual gripping failures (e.g., failure at the growth plate failure rather than in the tendon), decreased overall testing time, and increased reproducibility. Furthermore, this new method is readily adaptable for testing other murine tendons and tendons from other animals.
Collapse
Affiliation(s)
- Iden Kurtaliaj
- Department of Orthopedic Surgery, Columbia University; Department of Biomedical Engineering, Columbia University
| | - Mikhail Golman
- Department of Orthopedic Surgery, Columbia University; Department of Biomedical Engineering, Columbia University
| | | | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University; Department of Biomedical Engineering, Columbia University;
| |
Collapse
|
22
|
Freedman BR, Rodriguez AB, Hillin CD, Weiss SN, Han B, Han L, Soslowsky LJ. Tendon healing affects the multiscale mechanical, structural and compositional response of tendon to quasi-static tensile loading. J R Soc Interface 2019; 15:rsif.2017.0880. [PMID: 29467258 DOI: 10.1098/rsif.2017.0880] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/29/2018] [Indexed: 12/20/2022] Open
Abstract
Tendon experiences a variety of multiscale changes to its extracellular matrix during mechanical loading at the fascicle, fibre and fibril levels. For example, tensile loading of tendon increases its stiffness, with organization of collagen fibres, and increases cell strain in the direction of loading. Although applied macroscale strains correlate to cell and nuclear strains in uninjured tendon, the multiscale response during tendon healing remains unknown and may affect cell mechanosensing and response. Therefore, this study evaluated multiscale structure-function mechanisms in response to quasi-static tensile loading in uninjured and healing tendons. We found that tendon healing affected the macroscale mechanical and structural response to mechanical loading, evidenced by decreases in strain stiffening and collagen fibre realignment. At the micro- and nanoscales, healing resulted in increased collagen fibre disorganization, nuclear disorganization, decreased change in nuclear aspect ratio with loading, and decreased indentation modulus compared to uninjured tendons. Taken together, this work supports a new concept of nuclear strain transfer attenuation during tendon healing and identifies several multiscale properties that may contribute. Our work also provides benchmarks for the biomechanical microenvironments that tendon cells may experience following cell delivery therapies.
Collapse
Affiliation(s)
- Benjamin R Freedman
- McKay Orthopedic Research Laboratory, University of Pennsylvania, 110 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081, USA
| | - Ashley B Rodriguez
- McKay Orthopedic Research Laboratory, University of Pennsylvania, 110 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081, USA
| | - Cody D Hillin
- McKay Orthopedic Research Laboratory, University of Pennsylvania, 110 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081, USA
| | - Stephanie N Weiss
- McKay Orthopedic Research Laboratory, University of Pennsylvania, 110 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081, USA
| | - Biao Han
- Department of Biomedical Engineering, Drexel University, Philadelphia, PA, USA
| | - Lin Han
- Department of Biomedical Engineering, Drexel University, Philadelphia, PA, USA
| | - Louis J Soslowsky
- McKay Orthopedic Research Laboratory, University of Pennsylvania, 110 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081, USA
| |
Collapse
|
23
|
Wu J, Ren B, Shi F, Hua P, Lin H. BMP and mTOR signaling in heterotopic ossification: Does their crosstalk provide therapeutic opportunities? J Cell Biochem 2019; 120:12108-12122. [PMID: 30989716 DOI: 10.1002/jcb.28710] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/17/2019] [Accepted: 03/22/2019] [Indexed: 12/23/2022]
Abstract
Heterotopic ossification (HO) refers to the pathological formation of ectopic bone in soft tissues, it occurs following severe trauma or in patients with a rare genetic disorder known as fibrodysplasia ossificans progressiva. The pathological process of HO formation is a two-step mechanism: inflammation and destruction of connective tissues, followed by bone formation. The latter is further subdivided into three stages: fibroproliferation/angiogenesis, chondrogenesis, and osteogenesis. Currently, therapeutic options for HO are limited. New potential therapeutics will most likely arise from a more detailed understanding of the signaling pathways implicated in each stage of ectopic bone formation and molecular targets that may be effective at both the early and late stages of HO. Bone morphogenetic protein (BMP) signaling is believed to play a key role in the overall HO process. Recently, the mammalian target of rapamycin (mTOR) signaling pathway has received attention as a critical pathway for chondrogenesis, osteogenesis, and HO. Inhibition of mTOR signaling has been shown to block trauma-induced and genetic HO. Intriguingly, recent studies have revealed crosstalk between mTOR and BMP signaling. Moreover, mTOR has emerged as a factor involved in the early hypoxic and inflammatory stages of HO. We will summarize the current knowledge of the roles of mTOR and BMP signaling in HO, with a particular focus on the crosstalk between mTOR and BMP signaling. We also discuss the activation of AMP activated protein kinase (AMPK) by the most widely used drug for type 2 diabetes, metformin, which exerts a dual negative regulatory effect on mTOR and BMP signaling, suggesting that metformin is a promising drug treatment for HO. The discovery of an mTOR-BMP signaling network may be a potential molecular mechanism of HO and may represent a novel therapeutic target for the pharmacological control of HO.
Collapse
Affiliation(s)
- Jianhui Wu
- Jiangxi Medical School, Nanchang University, Nanchang, Jiangx, China.,Nanchang Joint Programme, Queen Mary University of London, London, UK
| | - Bowen Ren
- Jiangxi Medical School, Nanchang University, Nanchang, Jiangx, China.,Nanchang Joint Programme, Queen Mary University of London, London, UK
| | - Fuli Shi
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Ping Hua
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
24
|
Qing Q, Zhang YJ, Yang JL, Ning LJ, Zhang YJ, Jiang YL, Zhang Y, Luo JC, Qin TW. Effects of hydrogen peroxide on biological characteristics and osteoinductivity of decellularized and demineralized bone matrices. J Biomed Mater Res A 2019; 107:1476-1490. [PMID: 30786151 DOI: 10.1002/jbm.a.36662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 02/05/2023]
Abstract
Due to the similar collagen composition and closely physiological relationship with soft connective tissues, demineralized bone matrices (DBMs) were used to repair the injured tendon or ligament. However, the osteoinductivity of DBMs would be a huge barrier of these applications. Hydrogen peroxide (H2 O2 ) has been proved to reduce the osteoinductivity of DBMs. Nevertheless, the biological properties of H2 O2 -treated DBMs have not been evaluated completely, while the potential mechanism of H2 O2 compromising osteoinductivity is also unclear. Hence, the purpose of this study was to characterize the biological properties of H2 O2 -treated DBMs and search for the proof that H2 O2 could compromise osteoinductivity of DBMs. Decellularized and demineralized bone matrices (DCDBMs) were washed by 3% H2 O2 for 12 h to fabricate the H2 O2 -treated DCDBMs (HPTBMs). Similar biological properties including collagen, biomechanics, and biocompatibility were observed between DCDBMs and HPTBMs. The immunohistochemistry staining of bone morphogenetic protein 2 (BMP-2) was negative in HPTBMs. Furthermore, HPTBMs exhibited significantly reduced osteoinductivity both in vitro and in vivo. Taken together, these findings suggest that the BMP-2 in DCDBMs could be the target of H2 O2 . HPTBMs could be expected to be used as a promising scaffold for tissue engineering. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A, 2019.
Collapse
Affiliation(s)
- Quan Qing
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.,Faculty of Basic Medicine, Sichuan College of Traditional Chinese Medicine, Mianyang 621000, China
| | - Yan-Jing Zhang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie-Liang Yang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang-Ju Ning
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ya-Jing Zhang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan-Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing-Cong Luo
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting-Wu Qin
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
25
|
Abstract
Tendons connect muscle to bone and play an integral role in bone and joint alignment and loading. Tendons act as pulleys that provide anchorage of muscle forces for joint motion and stability, as well as for fracture reduction and realignment. Patients that experience complex fractures also have concomitant soft tissue injuries, such as tendon damage or rupture. Tendon injuries that occur at the time of bone fracture have long-term ramifications on musculoskeletal health, yet these injuries are often disregarded in clinical treatment and diagnosis for patients with bone fractures as well as in basic science approaches for understanding bone repair processes. Delayed assessment of soft tissue injuries during evaluation of trauma can lead to chronic pain, dysfunction, and delayed bone healing even following successful fracture repair, highlighting the importance of identifying and treating damaged tendons early. Treatment strategies for bone repair, such as mechanical stabilization and biological therapeutics, can impact tendon healing and function. Because poor tendon healing following complex fracture can significantly impact the function of tendon during bone fracture healing, a need exists to understand the healing process of complex fractures more broadly, beyond the healing of bone. In this review, we explored the mechanical and biological interaction of bone and tendon in the context of complex fracture, as well as the relevance and potential ramifications of tendon damage following bone fracture, which has particular impact on patients that experience complex fractures, such as from combat, automobile accidents, and other trauma.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware 19716
| | - Megan L. Killian
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716
| |
Collapse
|
26
|
Zhang K, Hast MW, Izumi S, Usami Y, Shetye S, Akabudike N, Philp NJ, Iwamoto M, Nissim I, Soslowsky LJ, Enomoto-Iwamoto M. Modulating Glucose Metabolism and Lactate Synthesis in Injured Mouse Tendons: Treatment With Dichloroacetate, a Lactate Synthesis Inhibitor, Improves Tendon Healing. Am J Sports Med 2018; 46:2222-2231. [PMID: 29927623 PMCID: PMC6510478 DOI: 10.1177/0363546518778789] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tendon injuries are common problems among athletes. Complete recovery of the mechanical structure and function of ruptured tendons is challenging. It has been demonstrated that upregulation of glycolysis and lactate production occurs in wounds, inflammation sites, and cancerous tumors, and these metabolic changes also control growth and differentiation of stem and progenitor cells. Similar metabolic changes have been reported in human healing tendons. In addition, lactate production has increased in progenitors isolated from injured tendons after treatment with IL-1β. It is thought that the metabolic changes play a role in tendon healing after injury. HYPOTHESIS Glucose metabolism is altered during tendon injury and healing, and modulation of this altered metabolism improves tendon repair. STUDY DESIGN Controlled laboratory study. METHODS The authors used the tendon injury model involving a complete incision of the Achilles tendon in C57BL/6J female mice and studied alterations of glucose metabolism in injured tendons with [U-13C]glucose and metabolomics analysis 1 and 4 weeks after surgery. They also examined the effects of dichloroacetate (DCA; an indirect lactate synthesis inhibitor) treatment on the recovery of structure and mechanical properties of injured tendons 4 weeks after surgery in the same mouse model. RESULTS Significant changes in glucose metabolism in tendons after injury surgery were detected. 13C enrichment of metabolites and intermediates, flux through glycolysis, and lactate synthesis, as well as tricarboxylic acid cycle activity, were acutely increased 1 week after injury. Increased glycolysis and lactate generation were also found 4 weeks after injury. DCA-treated injured tendons showed decreased cross-sectional area and higher values of modulus, maximum stress, and maximum force when compared with vehicle-treated injured tendons. Improved alignment of the collagen fibers was also observed in the DCA group. Furthermore, DCA treatment reduced mucoid accumulation and ectopic calcification in injured tendons. CONCLUSION The findings indicate that injured tendons acutely increase glycolysis and lactate synthesis after injury and that the inhibition of lactate synthesis by DCA is beneficial for tendon healing. CLINICAL RELEVANCE Changing metabolism in injured tendons may be a therapeutic target for tendon repair.
Collapse
Affiliation(s)
- Kairui Zhang
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michael W Hast
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Soutarou Izumi
- Department of Orthopaedics, University of Maryland, Baltimore, Maryland, USA
| | - Yu Usami
- Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Oral Pathology, Osaka University, Osaka, Japan
| | - Snehal Shetye
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ngozi Akabudike
- Department of Orthopaedics, University of Maryland, Baltimore, Maryland, USA
| | - Nancy J Philp
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Masahiro Iwamoto
- Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Orthopaedics, University of Maryland, Baltimore, Maryland, USA
| | - Itzhak Nissim
- Division of Metabolism and Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Louis J Soslowsky
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Motomi Enomoto-Iwamoto
- Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Orthopaedics, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Sakabe T, Sakai K, Maeda T, Sunaga A, Furuta N, Schweitzer R, Sasaki T, Sakai T. Transcription factor scleraxis vitally contributes to progenitor lineage direction in wound healing of adult tendon in mice. J Biol Chem 2018; 293:5766-5780. [PMID: 29507095 PMCID: PMC5912447 DOI: 10.1074/jbc.ra118.001987] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/16/2018] [Indexed: 01/02/2023] Open
Abstract
Tendon is a dense connective tissue that transmits high mechanical forces from skeletal muscle to bone. The transcription factor scleraxis (Scx) is a highly specific marker of both precursor and mature tendon cells (tenocytes). Mice lacking scx exhibit a specific and virtually complete loss of tendons during development. However, the functional contribution of Scx to wound healing in adult tendon has not yet been fully characterized. Here, using ScxGFP-tracking and loss-of-function systems, we show in an adult mouse model of Achilles tendon injury that paratenon cells, representing a stem cell antigen-1 (Sca-1)-positive and Scx-negative progenitor subpopulation, display Scx induction, migrate to the wound site, and produce extracellular matrix (ECM) to bridge the defect, whereas resident tenocytes exhibit a delayed response. Scx induction in the progenitors is initiated by transforming growth factor β (TGF-β) signaling. scx-deficient mice had migration of Sca-1-positive progenitor cell to the lesion site but impaired ECM assembly to bridge the defect. Mechanistically, scx-null progenitors displayed higher chondrogenic potential with up-regulation of SRY-box 9 (Sox9) coactivator PPAR-γ coactivator-1α (PGC-1α) in vitro, and knock-in analysis revealed that forced expression of full-length scx significantly inhibited Sox9 expression. Accordingly, scx-null wounds formed cartilage-like tissues that developed ectopic ossification. Our findings indicate a critical role of Scx in a progenitor-cell lineage in wound healing of adult mouse tendon. These progenitor cells could represent targets in strategies to facilitate tendon repair. We propose that this lineage-regulatory mechanism in tissue progenitors could apply to a broader set of tissues or biological systems in the body.
Collapse
Affiliation(s)
- Tomoya Sakabe
- From the Medical Research Council Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, United Kingdom
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Keiko Sakai
- From the Medical Research Council Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, United Kingdom
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Toru Maeda
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Ataru Sunaga
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Nao Furuta
- From the Medical Research Council Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, Oregon 97239, and
| | - Takako Sasaki
- Department of Biochemistry, Faculty of Medicine, Oita University, Oita 879-5593, Japan
| | - Takao Sakai
- From the Medical Research Council Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, United Kingdom,
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
28
|
Mead TJ, McCulloch DR, Ho JC, Du Y, Adams SM, Birk DE, Apte SS. The metalloproteinase-proteoglycans ADAMTS7 and ADAMTS12 provide an innate, tendon-specific protective mechanism against heterotopic ossification. JCI Insight 2018; 3:92941. [PMID: 29618652 DOI: 10.1172/jci.insight.92941] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/28/2018] [Indexed: 12/22/2022] Open
Abstract
Heterotopic ossification (HO) is a significant clinical problem with incompletely resolved mechanisms. Here, the secreted metalloproteinases ADAMTS7 and ADAMTS12 are shown to comprise a unique proteoglycan class that protects against a tendency toward HO in mouse hindlimb tendons, menisci, and ligaments. Adamts7 and Adamts12 mRNAs were sparsely expressed in murine forelimbs but strongly coexpressed in hindlimb tendons, skeletal muscle, ligaments, and meniscal fibrocartilage. Adamts7-/- Adamts12-/- mice, but not corresponding single-gene mutants, which demonstrated compensatory upregulation of the intact homolog mRNA, developed progressive HO in these tissues after 4 months of age. Adamts7-/- Adamts12-/- tendons had abnormal collagen fibrils, accompanied by reduced levels of the small leucine-rich proteoglycans (SLRPs) biglycan, fibromodulin, and decorin, which regulate collagen fibrillogenesis. Bgn-/0 Fmod-/- mice are known to have a strikingly similar hindlimb HO to that of Adamts7-/- Adamts12-/- mice, implicating fibromodulin and biglycan reduction as a likely mechanism underlying HO in Adamts7-/- Adamts12-/- mice. Interestingly, degenerated human biceps tendons had reduced ADAMTS7 mRNA compared with healthy biceps tendons, which expressed both ADAMTS7 and ADAMTS12. These results suggest that ADAMTS7 and ADAMTS12 drive an innate pathway protective against hindlimb HO in mice and may be essential for human tendon health.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Daniel R McCulloch
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Jason C Ho
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA.,Department of Orthopaedic Surgery and the Orthopaedic and Rheumatology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yaoyao Du
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Sheila M Adams
- Departments of Molecular Pharmacology and Physiology and Orthopaedics and Sports Medicine, University of South Florida, Morsani College of Medicine, Tampa, Florida, USA
| | - David E Birk
- Departments of Molecular Pharmacology and Physiology and Orthopaedics and Sports Medicine, University of South Florida, Morsani College of Medicine, Tampa, Florida, USA
| | - Suneel S Apte
- Department of Biomedical Engineering and the Orthopaedic and Rheumatologic Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| |
Collapse
|
29
|
Strategic Targeting of Multiple BMP Receptors Prevents Trauma-Induced Heterotopic Ossification. Mol Ther 2017; 25:1974-1987. [PMID: 28716575 DOI: 10.1016/j.ymthe.2017.01.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/09/2017] [Accepted: 01/11/2017] [Indexed: 01/08/2023] Open
Abstract
Trauma-induced heterotopic ossification (tHO) is a condition of pathologic wound healing, defined by the progressive formation of ectopic bone in soft tissue following severe burns or trauma. Because previous studies have shown that genetic variants of HO, such as fibrodysplasia ossificans progressiva (FOP), are caused by hyperactivating mutations of the type I bone morphogenetic protein receptor (T1-BMPR) ACVR1/ALK2, studies evaluating therapies for HO have been directed primarily toward drugs for this specific receptor. However, patients with tHO do not carry known T1-BMPR mutations. Here we show that, although BMP signaling is required for tHO, no single T1-BMPR (ACVR1/ALK2, BMPR1a/ALK3, or BMPR1b/ALK6) alone is necessary for this disease, suggesting that these receptors have functional redundancy in the setting of tHO. By utilizing two different classes of BMP signaling inhibitors, we developed a translational approach to treatment, integrating treatment choice with existing diagnostic options. Our treatment paradigm balances either immediate therapy with reduced risk for adverse effects (Alk3-Fc) or delayed therapy with improved patient selection but greater risk for adverse effects (LDN-212854).
Collapse
|
30
|
Morita W, Snelling SJB, Dakin SG, Carr AJ. Profibrotic mediators in tendon disease: a systematic review. Arthritis Res Ther 2016; 18:269. [PMID: 27863509 PMCID: PMC5116130 DOI: 10.1186/s13075-016-1165-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022] Open
Abstract
Background Tendon disease is characterized by the development of fibrosis. Transforming growth factor beta (TGF-β), bone morphogenic proteins (BMPs) and connective tissue growth factor (CTGF) are key mediators in the pathogenesis of fibrotic disorders. The aim of this systematic review was to investigate the evidence for the expression of TGF-β, BMPs and CTGF along tendon disease progression and the response of tendon cells to these growth factors accordingly. Method We conducted a systematic screen of the scientific literature using the Medline database. The search terms used were “tendon AND TGF-β,” “tendon AND BMP” or “tendon AND CTGF.” Studies of human samples, animal tendon injury and overuse models were included. Results Thirty-three studies were included. In eight studies the expression of TGF-β, BMPs or CTGF was dysregulated in chronic tendinopathy and tendon tear patient tissues in comparison with healthy control tissues. The expression of TGF-β, BMPs and CTGF was increased and showed temporal changes in expression in tendon tissues from animal injury or overuse models compared with the healthy control (23 studies), but the pattern of upregulation was inconsistent between growth factors and also the type of animal model. No study investigated the differences in the effect of TGF-β, BMPs or CTGF treatment between patient-derived cells from healthy and diseased tendon tissues. Tendon cells derived from animal models of tendon injury showed increased expression of extracellular matrix protein genes and increased cell signaling response to TGF-β and BMP treatments compared with the control cells (two studies). Conclusion The expression of TGF-β, BMPs and CTGF in tendon tissues is altered temporally during healing in animal models of tendon injury or overuse, but the transition during the development of human tendon disease is currently unknown. Findings from this systematic review suggest a potential and compelling role for TGF-β, BMPs and CTGF in tendon disease; however, there is a paucity of studies analyzing their expression and stimulated cellular response in well-phenotyped human samples. Future work should investigate the dynamic expression of these fibrotic growth factors and their interaction with tendon cells using patient samples at different stages of human tendon disease. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1165-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wataru Morita
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford, OX3 7LD, UK. .,NIHR Oxford Biomedical Research Unit, Botnar Research Centre, University of Oxford, Windmill Road, Headington, Oxford, OX3 7LD, UK.
| | - Sarah Jane Bothwell Snelling
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford, OX3 7LD, UK.,NIHR Oxford Biomedical Research Unit, Botnar Research Centre, University of Oxford, Windmill Road, Headington, Oxford, OX3 7LD, UK
| | - Stephanie Georgina Dakin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford, OX3 7LD, UK.,NIHR Oxford Biomedical Research Unit, Botnar Research Centre, University of Oxford, Windmill Road, Headington, Oxford, OX3 7LD, UK
| | - Andrew Jonathan Carr
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford, OX3 7LD, UK.,NIHR Oxford Biomedical Research Unit, Botnar Research Centre, University of Oxford, Windmill Road, Headington, Oxford, OX3 7LD, UK
| |
Collapse
|