51
|
Cho Y, Kim HS, Kang D, Kim H, Lee N, Yun J, Kim YJ, Lee KM, Kim JH, Kim HR, Hwang YI, Jo CH, Kim JH. CTRP3 exacerbates tendinopathy by dysregulating tendon stem cell differentiation and altering extracellular matrix composition. SCIENCE ADVANCES 2021; 7:eabg6069. [PMID: 34797714 PMCID: PMC8604415 DOI: 10.1126/sciadv.abg6069] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 10/01/2021] [Indexed: 05/31/2023]
Abstract
Tendinopathy, the most common disorder affecting tendons, is characterized by chronic disorganization of the tendon matrix, which leads to tendon tear and rupture. The goal was to identify a rational molecular target whose blockade can serve as a potential therapeutic intervention for tendinopathy. We identified C1q/TNF-related protein-3 (CTRP3) as a markedly up-regulated cytokine in human and rodent tendinopathy. Overexpression of CTRP3 enhanced the progression of tendinopathy by accumulating cartilaginous proteoglycans and degenerating collagenous fibers in the mouse tendon, whereas CTRP3 knockdown suppressed the tendinopathy pathogenesis. Functional blockade of CTRP3 using a neutralizing antibody ameliorated overuse-induced tendinopathy of the Achilles and rotator cuff tendons. Mechanistically, CTRP3 elicited a transcriptomic pattern that stimulates abnormal differentiation of tendon stem/progenitor cells and ectopic chondrification as an effect linked to activation of Akt signaling. Collectively, we reveal an essential role for CTRP3 in tendinopathy and propose a potential therapeutic strategy for the treatment of tendinopathy.
Collapse
Affiliation(s)
- Yongsik Cho
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Hyeon-Seop Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Donghyun Kang
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Hyeonkyeong Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Narae Lee
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Jihye Yun
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
- School of Medicine, CHA University, 13496 Seongnam, South Korea
| | - Yi-Jun Kim
- Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, 07985 Seoul, South Korea
| | - Kyoung Min Lee
- Foot and Ankle Division, Department of Orthopedic Surgery, Seoul National University Bundang Hospital, 13620 Seongnam, South Korea
| | - Jin-Hee Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 03080 Seoul, South Korea
| | - Hang-Rae Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 03080 Seoul, South Korea
| | - Young-il Hwang
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 03080 Seoul, South Korea
| | - Chris Hyunchul Jo
- Department of Orthopedic Surgery, Seoul Metropolitan Government–Seoul National University (SMG-SNU) Boramae Medical Center, Seoul National University College of Medicine, 07061 Seoul, South Korea
| | - Jin-Hong Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, 08826 Seoul, South Korea
| |
Collapse
|
52
|
Liao G, Liao Y, Li D, Fu Z, Wu S, Cheng D, Ouyang Q, Tang Z, Zeng G, Liang X, Xu S, Hu J, Liu M. Human Platelet Lysate Maintains Stemness of Umbilical Cord-Derived Mesenchymal Stromal Cells and Promote Lung Repair in Rat Bronchopulmonary Dysplasia. Front Cell Dev Biol 2021; 9:722953. [PMID: 34858970 PMCID: PMC8631747 DOI: 10.3389/fcell.2021.722953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/05/2021] [Indexed: 11/23/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) show potential for treating preclinical models of newborn bronchopulmonary dysplasia (BPD), but studies of their therapeutic effectiveness have had mixed results, in part due to the use of different media supplements for MSCs expansion in vitro. The current study sought to identify an optimal culture supplement of umbilical cord-derived MSCs (UC-MSCs) for BPD therapy. In this study, we found that UC-MSCs cultured with human platelet lysate (hPL-UCMSCs) were maintained a small size from Passage 1 (P1) to P10, while UC-MSCs cultured with fetal bovine serum (FBS-UCMSCs) became wide and flat. Furthermore, hPL was associated with lower levels of senescence in UC-MSCs during in vitro expansion compared with FBS, as indicated by the results of β-galactosidase staining and measures of senescence-related genes (CDKN2A, CDKN1A, and mTOR). In addition, hPL enhanced the proliferation and cell viability of the UC-MSCs and reduced their doubling time in vitro. Compared with FBS-UCMSCs, hPL-UCMSCs have a greater potential to differentiate into osteocytes and chondrocytes. Moreover, using hPL resulted in greater expression of Nestin and specific paracrine factors (VEGF, TGF-β1, FGF2, IL-8, and IL-6) in UC-MSCs compared to using FBS. Critically, we also found that hPL-UCMSCs are more effective than FBS-UCMSCs for the treatment of BPD in a rat model, with hPL leading to improvements in survival rate, lung architecture and fibrosis, and lung capillary density. Finally, qPCR of rat lung mRNA demonstrated that hPL-UCMSCs had lower expression levels of inflammatory factors (TNF-α and IL-1β) and a key chemokine (MCP-1) at postnatal day 10, and there was significant reduction of CD68+ macrophages in lung tissue after hPL-UCMSCs transplantation. Altogether, our findings suggest that hPL is an optimal culture supplement for UC-MSCs expansion in vitro, and that hPL-UCMSCs promote lung repair in rat BPD disease.
Collapse
Affiliation(s)
- Guilian Liao
- Obstetrics and Gynecology, Maternal and Child Health Hospital of Longgang District, Shenzhen, China
| | - Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Duanduan Li
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Zeqin Fu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Shiduo Wu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Danling Cheng
- Obstetrics and Gynecology, Maternal and Child Health Hospital of Longgang District, Shenzhen, China
| | - Qiuxing Ouyang
- Neurological Rehabilitation for Children, Maternal and Child Health Hospital of Luohu District, Shenzhen, China
| | - Zan Tang
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Guifang Zeng
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Xiao Liang
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Shaokun Xu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Muyun Liu
- National-Local Associated Engineering Laboratory for Personalized Cell Therapy, Shenzhen, China
| |
Collapse
|
53
|
Taguchi T, Zhang N, Angibeau D, Spivey KP, Lopez MJ. Evaluation of canine adipose-derived multipotent stromal cell differentiation to ligamentoblasts on tensioned collagen type I templates in a custom bioreactor culture system. Am J Vet Res 2021; 82:924-934. [PMID: 34669492 DOI: 10.2460/ajvr.82.11.924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate differentiation of canine adipose-derived multipotent stromal cells (ASCs) into ligamentoblasts on tensioned collagen type I (Col1) templates in a perfusion culture system. SAMPLES Infrapatellar fat pad ASCs from healthy stifle joints of 6 female mixed-breed dogs. PROCEDURES Third-passage ASCs (6 × 106 cells/template) were loaded onto suture-augmented Col1 templates under 15% static strain in perfusion bioreactors. Forty-eight ASC-Col1 constructs were incubated with ligamentogenic (ligamentogenic constructs; n = 24) or stromal medium (stromal constructs; 24) for up to 21 days. Specimens were collected from each construct after 2 hours (day 0) and 7, 14, and 21 days of culture. Cell number, viability, distribution, and morphology; construct collagen content; culture medium procollagen-I-N-terminal peptide concentration; and gene expression were compared between ligamentogenic and stromal constructs. RESULTS ASCs adhered to collagen fibers. Cell numbers increased from days 0 to 7 and days 14 to 21 for both construct types. Relative to stromal constructs, cell morphology and extracellular matrix were more mature and collagen content on day 21 and procollagen-I-N-terminal peptide concentration on days 7 and 21 were greater for ligamentogenic constructs. Ligamentogenic constructs had increased expression of the genes biglycan on day 7, decorin throughout the culture period, and Col1, tenomodulin, fibronectin, and tenascin-c on day 21; expression of Col1, tenomodulin, and tenascin-c increased between days 7 and 21. CONCLUSIONS AND CLINICAL RELEVANCE Ligamentogenic medium was superior to stromal medium for differentiation of ASCs to ligamentoblasts on suture-augmented Col1 scaffolds. Customized ligament neotissue may augment treatment options for dogs with cranial cruciate ligament rupture.
Collapse
Affiliation(s)
- Takashi Taguchi
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Nan Zhang
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Dominique Angibeau
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Kathryn P Spivey
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Mandi J Lopez
- From the Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| |
Collapse
|
54
|
Tendon and multiomics: advantages, advances, and opportunities. NPJ Regen Med 2021; 6:61. [PMID: 34599188 PMCID: PMC8486786 DOI: 10.1038/s41536-021-00168-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 09/01/2021] [Indexed: 02/08/2023] Open
Abstract
Tendons heal by fibrosis, which hinders function and increases re-injury risk. Yet the biology that leads to degeneration and regeneration of tendons is not completely understood. Improved understanding of the metabolic nuances that cause diverse outcomes in tendinopathies is required to solve these problems. 'Omics methods are increasingly used to characterize phenotypes in tissues. Multiomics integrates 'omic datasets to identify coherent relationships and provide insight into differences in molecular and metabolic pathways between anatomic locations, and disease stages. This work reviews the current literature pertaining to multiomics in tendon and the potential of these platforms to improve tendon regeneration. We assessed the literature and identified areas where 'omics platforms contribute to the field: (1) Tendon biology where their hierarchical complexity and demographic factors are studied. (2) Tendon degeneration and healing, where comparisons across tendon pathologies are analyzed. (3) The in vitro engineered tendon phenotype, where we compare the engineered phenotype to relevant native tissues. (4) Finally, we review regenerative and therapeutic approaches. We identified gaps in current knowledge and opportunities for future study: (1) The need to increase the diversity of human subjects and cell sources. (2) Opportunities to improve understanding of tendon heterogeneity. (3) The need to use these improvements to inform new engineered and regenerative therapeutic approaches. (4) The need to increase understanding of the development of tendon pathology. Together, the expanding use of various 'omics platforms and data analysis resulting from these platforms could substantially contribute to major advances in the tendon tissue engineering and regenerative medicine field.
Collapse
|
55
|
Marr N, Meeson R, Kelly EF, Fang Y, Peffers MJ, Pitsillides AA, Dudhia J, Thorpe CT. CD146 Delineates an Interfascicular Cell Sub-Population in Tendon That Is Recruited during Injury through Its Ligand Laminin-α4. Int J Mol Sci 2021; 22:9729. [PMID: 34575887 PMCID: PMC8472220 DOI: 10.3390/ijms22189729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/02/2021] [Accepted: 09/05/2021] [Indexed: 12/18/2022] Open
Abstract
The interfascicular matrix (IFM) binds tendon fascicles and contains a population of morphologically distinct cells. However, the role of IFM-localised cell populations in tendon repair remains to be determined. The basement membrane protein laminin-α4 also localises to the IFM. Laminin-α4 is a ligand for several cell surface receptors, including CD146, a marker of pericyte and progenitor cells. We used a needle injury model in the rat Achilles tendon to test the hypothesis that the IFM is a niche for CD146+ cells that are mobilised in response to tendon damage. We also aimed to establish how expression patterns of circulating non-coding RNAs alter with tendon injury and identify potential RNA-based markers of tendon disease. The results demonstrate the formation of a focal lesion at the injury site, which increased in size and cellularity for up to 21 days post injury. In healthy tendon, CD146+ cells localised to the IFM, compared with injury, where CD146+ cells migrated towards the lesion at days 4 and 7, and populated the lesion 21 days post injury. This was accompanied by increased laminin-α4, suggesting that laminin-α4 facilitates CD146+ cell recruitment at injury sites. We also identified a panel of circulating microRNAs that are dysregulated with tendon injury. We propose that the IFM cell niche mediates the intrinsic response to injury, whereby an injury stimulus induces CD146+ cell migration. Further work is required to fully characterise CD146+ subpopulations within the IFM and establish their precise roles during tendon healing.
Collapse
Affiliation(s)
- Neil Marr
- Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK; (N.M.); (A.A.P.)
| | - Richard Meeson
- Clinical Sciences and Services, Royal Veterinary College, Hawkshead Lane, Hatfield AL9 7TA, UK; (R.M.); (E.F.K.); (J.D.)
| | - Elizabeth F. Kelly
- Clinical Sciences and Services, Royal Veterinary College, Hawkshead Lane, Hatfield AL9 7TA, UK; (R.M.); (E.F.K.); (J.D.)
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK;
| | - Mandy J. Peffers
- Institute of Ageing and Chronic Disease, University of Liverpool, Apex Building, 6 West Derby Street, Liverpool L7 9TX, UK;
| | - Andrew A. Pitsillides
- Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK; (N.M.); (A.A.P.)
| | - Jayesh Dudhia
- Clinical Sciences and Services, Royal Veterinary College, Hawkshead Lane, Hatfield AL9 7TA, UK; (R.M.); (E.F.K.); (J.D.)
| | - Chavaunne T. Thorpe
- Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK; (N.M.); (A.A.P.)
| |
Collapse
|
56
|
Geng Y, Zhao X, Xu J, Zhang X, Hu G, Fu SC, Dai K, Chen X, Patrick YSH, Zhang X. Overexpression of mechanical sensitive miR-337-3p alleviates ectopic ossification in rat tendinopathy model via targeting IRS1 and Nox4 of tendon-derived stem cells. J Mol Cell Biol 2021; 12:305-317. [PMID: 31065679 PMCID: PMC7232128 DOI: 10.1093/jmcb/mjz030] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/24/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023] Open
Abstract
Tendinopathy, which is characterized by the ectopic ossification of tendon, is a common disease occurring in certain population, such as athletes that suffer from repetitive tendon strains. However, the molecular mechanism underlying the pathogenesis of tendinopathy caused by the overuse of tendon is still lacking. Here, we found that the mechanosensitive miRNA, miR-337-3p, had lower expression under uniaxial cyclical mechanical loading in tendon-derived stem cells (TDSCs) and negatively controlled chondro-osteogenic differentiation of TDSCs. Importantly, downregulation of miR-337-3p expression was also observed in both rat and human calcified tendons, and overexpressing miR-337-3p in patellar tendons of rat tendinopathy model displayed a robust therapeutic efficiency. Mechanistically, we found that the proinflammatory cytokine interleukin-1β was the upstream factor of miR-337-3p that bridges the mechanical loading with its downregulation. Furthermore, the target genes of miR-337-3p, NADPH oxidase 4, and insulin receptor substrate 1, activated chondro-osteogenic differentiation of TDSCs through JNK and ERK signaling, respectively. Thus, these findings not only provide novel insight into the molecular mechanisms underlying ectopic ossification in tendinopathy but also highlight the significance of miR-337-3p as a putative therapeutic target for clinic treatment of tendinopathy.
Collapse
Affiliation(s)
- Yiyun Geng
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.,Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518035, China.,The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoying Zhao
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Jiajia Xu
- The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xudong Zhang
- The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoli Hu
- The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sai-Chuen Fu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kerong Dai
- The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaodong Chen
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Yung Shu-Huang Patrick
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.,The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
57
|
Still C, Chang WT, Sherman SL, Sochacki KR, Dragoo JL, Qi LS. Single-cell transcriptomic profiling reveals distinct mechanical responses between normal and diseased tendon progenitor cells. Cell Rep Med 2021; 2:100343. [PMID: 34337559 PMCID: PMC8324492 DOI: 10.1016/j.xcrm.2021.100343] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 02/23/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022]
Abstract
Regenerative medicine approaches utilizing stem cells offer a promising strategy to address tendinopathy, a class of common tendon disorders associated with pain and impaired function. Tendon progenitor cells (TPCs) are important in healing and maintaining tendon tissues. Here we provide a comprehensive single cell transcriptomic profiling of TPCs from three normal and three clinically classified tendinopathy samples in response to mechanical stimuli. Analysis reveals seven distinct TPC subpopulations including subsets that are responsive to the mechanical stress, highly clonogenic, and specialized in cytokine or growth factor expression. The single cell transcriptomic profiling of TPCs and their subsets serves as a foundation for further investigation into the pathology and molecular hallmarks of tendinopathy in mechanical stimulation conditions.
Collapse
Affiliation(s)
- Chris Still
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Wen-Teh Chang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Seth L. Sherman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Kyle R. Sochacki
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Jason L. Dragoo
- Deparment of Orthopaedic Surgery, University of Colorado, Denver, CO 80045, USA
| | - Lei S. Qi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Deparment of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
- ChEM-H, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
58
|
Madry H, Grässel S, Nöth U, Relja B, Bernstein A, Docheva D, Kauther MD, Katthagen JC, Bader R, van Griensven M, Wirtz DC, Raschke MJ, Huber-Lang M. The future of basic science in orthopaedics and traumatology: Cassandra or Prometheus? Eur J Med Res 2021; 26:56. [PMID: 34127057 PMCID: PMC8200553 DOI: 10.1186/s40001-021-00521-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/20/2021] [Indexed: 12/23/2022] Open
Abstract
Orthopaedic and trauma research is a gateway to better health and mobility, reflecting the ever-increasing and complex burden of musculoskeletal diseases and injuries in Germany, Europe and worldwide. Basic science in orthopaedics and traumatology addresses the complete organism down to the molecule among an entire life of musculoskeletal mobility. Reflecting the complex and intertwined underlying mechanisms, cooperative research in this field has discovered important mechanisms on the molecular, cellular and organ levels, which subsequently led to innovative diagnostic and therapeutic strategies that reduced individual suffering as well as the burden on the society. However, research efforts are considerably threatened by economical pressures on clinicians and scientists, growing obstacles for urgently needed translational animal research, and insufficient funding. Although sophisticated science is feasible and realized in ever more individual research groups, a main goal of the multidisciplinary members of the Basic Science Section of the German Society for Orthopaedics and Trauma Surgery is to generate overarching structures and networks to answer to the growing clinical needs. The future of basic science in orthopaedics and traumatology can only be managed by an even more intensified exchange between basic scientists and clinicians while fuelling enthusiasm of talented junior scientists and clinicians. Prioritized future projects will master a broad range of opportunities from artificial intelligence, gene- and nano-technologies to large-scale, multi-centre clinical studies. Like Prometheus in the ancient Greek myth, transferring the elucidating knowledge from basic science to the real (clinical) world will reduce the individual suffering from orthopaedic diseases and trauma as well as their socio-economic impact.
Collapse
Affiliation(s)
- Henning Madry
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg, Germany
| | - Susanne Grässel
- Experimental Orthopedics, Department of Orthopedic Surgery, University of Regensburg, Regensburg, Germany
| | - Ulrich Nöth
- Department of Orthopaedics and Trauma Surgery, Evangelisches Waldkrankenhaus Berlin Spandau, Berlin, Germany
| | - Borna Relja
- Experimental Radiology, University Clinic for Radiology and Nuclear Medicine, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Anke Bernstein
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Max Daniel Kauther
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Essen, Essen, Germany
| | - Jan Christoph Katthagen
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Rainer Bader
- Department of Orthopaedics, Research Lab for Biomechanics and Implant Technology, Rostock University Medical Center, Rostock, Germany
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN-Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Dieter C Wirtz
- Department of Orthopaedics and Trauma Surgery, University Hopsital Bonn, Bonn, Germany
| | - Michael J Raschke
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Helmholzstr. 8/1, Ulm, Germany.
| |
Collapse
|
59
|
Li Y, Wu T, Liu S. Identification and Distinction of Tenocytes and Tendon-Derived Stem Cells. Front Cell Dev Biol 2021; 9:629515. [PMID: 33937230 PMCID: PMC8085586 DOI: 10.3389/fcell.2021.629515] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/29/2021] [Indexed: 01/01/2023] Open
Abstract
Restoring the normal structure and function of injured tendons is one of the biggest challenges in orthopedics and sports medicine department. The discovery of tendon-derived stem cells (TDSCs) provides a novel perspective to treat tendon injuries, which is expected to be an ideal seed cell to promote tendon repair and regeneration. Because of the lack of specific markers, the identification of tenocytes and TDSCs has not been conclusive in the in vitro study of tendons. In addition, the morphology of tendon derived cells is similar, and the comparison and identification of tenocytes and TDSCs are insufficient, which causes some obstacles to the in vitro study of tendon. In this review, the characteristics of tenocytes and TDSCs are summarized and compared based on some existing research results (mainly in terms of biomarkers), and a potential marker selection for identification is suggested. It is of profound significance to further explore the mechanism of biomarkers in vivo and to find more specific markers.
Collapse
Affiliation(s)
- Yuange Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianyi Wu
- Department of Orthopaedics, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shen Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
60
|
Zha K, Li X, Yang Z, Tian G, Sun Z, Sui X, Dai Y, Liu S, Guo Q. Heterogeneity of mesenchymal stem cells in cartilage regeneration: from characterization to application. NPJ Regen Med 2021; 6:14. [PMID: 33741999 PMCID: PMC7979687 DOI: 10.1038/s41536-021-00122-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
Articular cartilage is susceptible to damage but hard to self-repair due to its avascular nature. Traditional treatment methods are not able to produce satisfactory effects. Mesenchymal stem cells (MSCs) have shown great promise in cartilage repair. However, the therapeutic effect of MSCs is often unstable partly due to their heterogeneity. Understanding the heterogeneity of MSCs and the potential of different types of MSCs for cartilage regeneration will facilitate the selection of superior MSCs for treating cartilage damage. This review provides an overview of the heterogeneity of MSCs at the donor, tissue source and cell immunophenotype levels, including their cytological properties, such as their ability for proliferation, chondrogenic differentiation and immunoregulation, as well as their current applications in cartilage regeneration. This information will improve the precision of MSC-based therapeutic strategies, thus maximizing the efficiency of articular cartilage repair.
Collapse
Affiliation(s)
- Kangkang Zha
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhen Yang
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Guangzhao Tian
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiqiang Sun
- Medical School of Chinese PLA, Beijing, China
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xiang Sui
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
| | - Yongjing Dai
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China
| | - Shuyun Liu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China.
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, China.
| |
Collapse
|
61
|
Tsai SL, Noedl MT, Galloway JL. Bringing tendon biology to heel: Leveraging mechanisms of tendon development, healing, and regeneration to advance therapeutic strategies. Dev Dyn 2021; 250:393-413. [PMID: 33169466 PMCID: PMC8486356 DOI: 10.1002/dvdy.269] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Tendons are specialized matrix-rich connective tissues that transmit forces from muscle to bone and are essential for movement. As tissues that frequently transfer large mechanical loads, tendons are commonly injured in patients of all ages. Following injury, mammalian tendons heal poorly through a slow process that forms disorganized fibrotic scar tissue with inferior biomechanical function. Current treatments are limited and patients can be left with a weaker tendon that is likely to rerupture and an increased chance of developing degenerative conditions. More effective, alternative treatments are needed. However, our current understanding of tendon biology remains limited. Here, we emphasize why expanding our knowledge of tendon development, healing, and regeneration is imperative for advancing tendon regenerative medicine. We provide a comprehensive review of the current mechanisms governing tendon development and healing and further highlight recent work in regenerative tendon models including the neonatal mouse and zebrafish. Importantly, we discuss how present and future discoveries can be applied to both augment current treatments and design novel strategies to treat tendon injuries.
Collapse
Affiliation(s)
- Stephanie L. Tsai
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Marie-Therese Noedl
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Jenna L. Galloway
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
62
|
Wang Y, Jin S, Luo D, He D, Shi C, Zhu L, Guan B, Li Z, Zhang T, Zhou Y, Wang CY, Liu Y. Functional regeneration and repair of tendons using biomimetic scaffolds loaded with recombinant periostin. Nat Commun 2021; 12:1293. [PMID: 33637721 PMCID: PMC7910464 DOI: 10.1038/s41467-021-21545-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 02/02/2021] [Indexed: 12/21/2022] Open
Abstract
Tendon injuries disrupt the balance between stability and mobility, causing compromised functions and disabilities. The regeneration of mature, functional tendons remains a clinical challenge. Here, we perform transcriptional profiling of tendon developmental processes to show that the extracellular matrix-associated protein periostin (Postn) contributes to the maintenance of tendon stem/progenitor cell (TSPC) functions and promotes tendon regeneration. We show that recombinant periostin (rPOSTN) promotes the proliferation and stemness of TSPCs, and maintains the tenogenic potentials of TSPCs in vitro. We also find that rPOSTN protects TSPCs against functional impairment during long-term passage in vitro. For in vivo tendon formation, we construct a biomimetic parallel-aligned collagen scaffold to facilitate TSPC tenogenesis. Using a rat full-cut Achilles tendon defect model, we demonstrate that scaffolds loaded with rPOSTN promote endogenous TSPC recruitment, tendon regeneration and repair with native-like hierarchically organized collagen fibers. Moreover, newly regenerated tendons show recovery of mechanical properties and locomotion functions. The regeneration of functional tendons remains a clinical challenge. Here the authors develop a biomimetic scaffold loaded with recombinant periostin and demonstrate its functionality in promoting tendon stem/progenitor cell recruitment and tenogenic differentiation, and tendon regeneration in a rat full-cut Achilles tendon defect model.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Shanshan Jin
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Dan Luo
- State Key Laboratory of Heavy Oil Processing, College of New Energy and Materials, Beijing Key Laboratory of Biogas Upgrading Utilization, China University of Petroleum (Beijing), Beijing, China
| | - Danqing He
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Chunyan Shi
- Department of Radiology, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung & Vascular Diseases, Capital Medical University, Beijing, China
| | - Lisha Zhu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Bo Guan
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Zixin Li
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Ting Zhang
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yanheng Zhou
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Cun-Yu Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry and Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, United States
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| |
Collapse
|
63
|
Huang Z, Yin Z, Xu J, Fei Y, Heng BC, Jiang X, Chen W, Shen W. Tendon Stem/Progenitor Cell Subpopulations and Their Implications in Tendon Biology. Front Cell Dev Biol 2021; 9:631272. [PMID: 33681210 PMCID: PMC7930382 DOI: 10.3389/fcell.2021.631272] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/27/2021] [Indexed: 12/28/2022] Open
Abstract
Tendon harbors a cell population that possesses stem cell characteristics such as clonogenicity, multipotency and self-renewal capacity, commonly referred to as tendon stem/progenitor cells (TSPCs). Various techniques have been employed to study how TSPCs are implicated in tendon development, homeostasis and healing. Recent advances in single-cell analysis have enabled much progress in identifying and characterizing distinct subpopulations of TSPCs, which provides a more comprehensive view of TSPCs function in tendon biology. Understanding the mechanisms of physiological and pathological processes regulated by TSPCs, especially a particular subpopulation, would greatly benefit treatment of diseased tendons. Here, we summarize the current scientific literature on the various subpopulations of TSPCs, and discuss how TSPCs can contribute to tissue homeostasis and pathogenesis, as well as examine the key modulatory signaling pathways that determine stem/progenitor cell state. A better understanding of the roles that TSPCs play in tendon biology may facilitate the development of novel treatment strategies for tendon diseases.
Collapse
Affiliation(s)
- Zizhan Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China.,Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Zi Yin
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China.,Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China.,China Orthopedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Jialu Xu
- Department of Infectious Diseases, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China.,Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing, China
| | - Xuesheng Jiang
- Department of Orthopedic Surgery, Huzhou Hospital, Zhejiang University, Huzhou, China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China.,Institute of Sports Medicine, Zhejiang University, Hangzhou, China.,Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China.,China Orthopedic Regenerative Medicine (CORMed), Hangzhou, China
| |
Collapse
|
64
|
Characterization of the structure, vascularity, and stem/progenitor cell populations in porcine Achilles tendon (PAT). Cell Tissue Res 2021; 384:367-387. [PMID: 33496880 DOI: 10.1007/s00441-020-03379-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/03/2020] [Indexed: 01/26/2023]
Abstract
This study aimed to characterize porcine Achilles tendon (PAT) in terms of its structural components, vascularity, and resident tendon cells. We found that PAT is composed of a paratenon sheath, a core of fascicles, and an endotenon/interfascicular matrix (IFM) that encases the fascicle bundles. We analyzed each of these three tendon components structurally using tissue sections and by isolating cells from each component and analyzing in vitro. Many blood vessel-like tissues were present in the paratenon and IFM but not in fascicles, and the vessels in the paratenon and IFM appeared to be inter-connected. Cells isolated from the paratenon and IFM displayed characteristics of vascular stem/progenitor cells expressing the markers CD105, CD31, with α-smooth muscle actin (α-SMA) localized surrounding blood vessels. The isolated cells from paratenon and IFM also harbored abundant stem/progenitor cells as evidenced by their ability to form colonies and express stem cell markers including CD73 and CD146. Furthermore, we demonstrate that both paratenon and IFM-isolated cells were capable of undergoing multi-differentiation. In addition, both paratenon and IFM cells expressed elastin, osteocalcin, tubulin polymerization promoting protein (TPPP), and collagen IV, whereas fascicle cells expressed none of these markers, except collagen I. The neurotransmitter substance P (SP) was also found in the paratenon and IFM-localized surrounding blood vessels. The findings of this study will help us to better understand the vascular and cellular mechanisms of tendon homeostasis, injury, healing, and regeneration.
Collapse
|
65
|
Millar NL, Silbernagel KG, Thorborg K, Kirwan PD, Galatz LM, Abrams GD, Murrell GAC, McInnes IB, Rodeo SA. Tendinopathy. Nat Rev Dis Primers 2021; 7:1. [PMID: 33414454 DOI: 10.1038/s41572-020-00234-1] [Citation(s) in RCA: 398] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 12/14/2022]
Abstract
Tendinopathy describes a complex multifaceted pathology of the tendon, characterized by pain, decline in function and reduced exercise tolerance. The most common overuse tendinopathies involve the rotator cuff tendon, medial and lateral elbow epicondyles, patellar tendon, gluteal tendons and the Achilles tendon. The prominent histological and molecular features of tendinopathy include disorganization of collagen fibres, an increase in the microvasculature and sensory nerve innervation, dysregulated extracellular matrix homeostasis, increased immune cells and inflammatory mediators, and enhanced cellular apoptosis. Although diagnosis is mostly achieved based on clinical symptoms, in some cases, additional pain-provoking tests and imaging might be necessary. Management consists of different exercise and loading programmes, therapeutic modalities and surgical interventions; however, their effectiveness remains ambiguous. Future research should focus on elucidating the key functional pathways implicated in clinical disease and on improved rehabilitation protocols.
Collapse
Affiliation(s)
- Neal L Millar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | | | - Kristian Thorborg
- Institute of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Paul D Kirwan
- School of Physiotherapy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Leesa M Galatz
- Department of Orthopaedic Surgery, Icahn School of Medicine, Mount Sinai Health System, New York, NY, USA
| | - Geoffrey D Abrams
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
66
|
Siadat SM, Zamboulis DE, Thorpe CT, Ruberti JW, Connizzo BK. Tendon Extracellular Matrix Assembly, Maintenance and Dysregulation Throughout Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:45-103. [PMID: 34807415 DOI: 10.1007/978-3-030-80614-9_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In his Lissner Award medal lecture in 2000, Stephen Cowin asked the question: "How is a tissue built?" It is not a new question, but it remains as relevant today as it did when it was asked 20 years ago. In fact, research on the organization and development of tissue structure has been a primary focus of tendon and ligament research for over two centuries. The tendon extracellular matrix (ECM) is critical to overall tissue function; it gives the tissue its unique mechanical properties, exhibiting complex non-linear responses, viscoelasticity and flow mechanisms, excellent energy storage and fatigue resistance. This matrix also creates a unique microenvironment for resident cells, allowing cells to maintain their phenotype and translate mechanical and chemical signals into biological responses. Importantly, this architecture is constantly remodeled by local cell populations in response to changing biochemical (systemic and local disease or injury) and mechanical (exercise, disuse, and overuse) stimuli. Here, we review the current understanding of matrix remodeling throughout life, focusing on formation and assembly during the postnatal period, maintenance and homeostasis during adulthood, and changes to homeostasis in natural aging. We also discuss advances in model systems and novel tools for studying collagen and non-collagenous matrix remodeling throughout life, and finally conclude by identifying key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Danae E Zamboulis
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Chavaunne T Thorpe
- Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Brianne K Connizzo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
67
|
He X, Li Y, Guo J, Xu J, Zu H, Huang L, Tim-Yun Ong M, Shu-Hang Yung P, Qin L. Biomaterials developed for facilitating healing outcome after anterior cruciate ligament reconstruction: Efficacy, surgical protocols, and assessments using preclinical animal models. Biomaterials 2020; 269:120625. [PMID: 33395579 DOI: 10.1016/j.biomaterials.2020.120625] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022]
Abstract
Anterior cruciate ligament (ACL) reconstruction is the recommended treatment for ACL tear in the American Academy of Orthopaedic Surgeons (AAOS) guideline. However, not a small number of cases failed because of the tunnel bone resorption, unsatisfactory bone-tendon integration, and graft degeneration. The biomaterials developed and designed for improving ACL reconstruction have been investigated for decades. According to the Food and Drug Administration (FDA) and the International Organization for Standardization (ISO) regulations, animal studies should be performed to prove the safety and bioeffect of materials before clinical trials. In this review, we first evaluated available biomaterials that can enhance the healing outcome after ACL reconstruction in animals and then discussed the animal models and assessments for testing applied materials. Furthermore, we identified the relevance and knowledge gaps between animal experimental studies and clinical expectations. Critical analyses and suggestions for future research were also provided to design the animal study connecting basic research and requirements for future clinical translation.
Collapse
Affiliation(s)
- Xuan He
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ye Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Haiyue Zu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Le Huang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Michael Tim-Yun Ong
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Patrick Shu-Hang Yung
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
68
|
Giduthuri AT, Theodossiou SK, Schiele NR, Srivastava SK. Dielectrophoresis as a tool for electrophysiological characterization of stem cells. BIOPHYSICS REVIEWS 2020; 1:011304. [PMID: 38505626 PMCID: PMC10903368 DOI: 10.1063/5.0025056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/20/2020] [Indexed: 03/21/2024]
Abstract
Dielectrophoresis (DEP), a nonlinear electrokinetic technique caused by Maxwell-Wagner interfacial polarization of neutral particles in an electrolyte solution, is a powerful cell manipulation method used widely for various applications such as enrichment, trapping, and sorting of heterogeneous cell populations. While conventional cell characterization and sorting methods require tagging or labeling of cells, DEP has the potential to manipulate cells in a label-free way. Due to its unique ability to characterize and sort cells without the need of labeling, there is renewed interest in using DEP for stem cell research and regenerative medicine. Stem cells have the potential to differentiate into various lineages, but achieving homogeneous cell phenotypes from an initially heterogeneous cell population is a challenge. Using DEP to efficiently and affordably identify, sort, and enrich either undifferentiated or differentiated stem cell populations in a label-free way would advance their potential uses for applications in tissue engineering and regenerative medicine. This review summarizes recent, significant research findings regarding the electrophysiological characterization of stem cells, with a focus on cellular dielectric properties, i.e., permittivity and conductivity, and on studies that have obtained these measurements using techniques that preserve cell viability, such as crossover frequency. Potential applications for DEP in regenerative medicine are also discussed. Overall, DEP is a promising technique and, when used to characterize, sort, and enrich stem cells, will advance stem cell-based regenerative therapies.
Collapse
Affiliation(s)
- Anthony T. Giduthuri
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Sophia K. Theodossiou
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Nathan R. Schiele
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Soumya K. Srivastava
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| |
Collapse
|
69
|
Dyment NA, Barrett JG, Awad H, Bautista CA, Banes A, Butler DL. A brief history of tendon and ligament bioreactors: Impact and future prospects. J Orthop Res 2020; 38:2318-2330. [PMID: 32579266 PMCID: PMC7722018 DOI: 10.1002/jor.24784] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/28/2020] [Accepted: 06/12/2020] [Indexed: 02/04/2023]
Abstract
Bioreactors are powerful tools with the potential to model tissue development and disease in vitro. For nearly four decades, bioreactors have been used to create tendon and ligament tissue-engineered constructs in order to define basic mechanisms of cell function, extracellular matrix deposition, tissue organization, injury, and tissue remodeling. This review provides a historical perspective of tendon and ligament bioreactors and their contributions to this advancing field. First, we demonstrate the need for bioreactors to improve understanding of tendon and ligament function and dysfunction. Next, we detail the history and evolution of bioreactor development and design from simple stretching of explants to fabrication and stimulation of two- and three-dimensional constructs. Then, we demonstrate how research using tendon and ligament bioreactors has led to pivotal basic science and tissue-engineering discoveries. Finally, we provide guidance for new basic, applied, and clinical research utilizing these valuable systems, recognizing that fundamental knowledge of cell-cell and cell-matrix interactions combined with appropriate mechanical and chemical stimulation of constructs could ultimately lead to functional tendon and ligament repairs in the coming decades.
Collapse
Affiliation(s)
- Nathaniel A. Dyment
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, PA
| | - Jennifer G. Barrett
- Department of Large Animal Clinical Sciences, Marion duPont Scott Equine Medical Center, Virginia Tech, Leesburg, VA
| | - Hani Awad
- Department of Biomedical Engineering, The Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14627
| | | | - Albert Banes
- Flexcell International Corp., 2730 Tucker St., Suite 200, Burlington, 27215, NC
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC
| | - David L. Butler
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221
| |
Collapse
|
70
|
Kendal AR, Layton T, Al-Mossawi H, Appleton L, Dakin S, Brown R, Loizou C, Rogers M, Sharp R, Carr A. Multi-omic single cell analysis resolves novel stromal cell populations in healthy and diseased human tendon. Sci Rep 2020; 10:13939. [PMID: 32883960 PMCID: PMC7471282 DOI: 10.1038/s41598-020-70786-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022] Open
Abstract
Tendinopathy accounts for over 30% of primary care consultations and represents a growing healthcare challenge in an active and increasingly ageing population. Recognising critical cells involved in tendinopathy is essential in developing therapeutics to meet this challenge. Tendon cells are heterogenous and sparsely distributed in a dense collagen matrix; limiting previous methods to investigate cell characteristics ex vivo. We applied next generation CITE-sequencing; combining surface proteomics with in-depth, unbiased gene expression analysis of > 6400 single cells ex vivo from 11 chronically tendinopathic and 8 healthy human tendons. Immunohistochemistry validated the single cell findings. For the first time we show that human tendon harbours at least five distinct COL1A1/2 expressing tenocyte populations in addition to endothelial cells, T-cells, and monocytes. These consist of KRT7/SCX+ cells expressing microfibril associated genes, PTX3+ cells co-expressing high levels of pro-inflammatory markers, APOD+ fibro–adipogenic progenitors, TPPP3/PRG4+ chondrogenic cells, and ITGA7+ smooth muscle-mesenchymal cells. Surface proteomic analysis identified markers by which these sub-classes could be isolated and targeted in future. Chronic tendinopathy was associated with increased expression of pro-inflammatory markers PTX3, CXCL1, CXCL6, CXCL8, and PDPN by microfibril associated tenocytes. Diseased endothelium had increased expression of chemokine and alarmin genes including IL33.
Collapse
Affiliation(s)
- Adrian R Kendal
- The Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK. .,Nuffield Orthopaedic Centre, Windmill Road, Oxford, OX3 7LD, UK.
| | - Thomas Layton
- The Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Hussein Al-Mossawi
- The Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Louise Appleton
- The Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Stephanie Dakin
- The Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Rick Brown
- Nuffield Orthopaedic Centre, Windmill Road, Oxford, OX3 7LD, UK
| | | | - Mark Rogers
- Nuffield Orthopaedic Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Robert Sharp
- Nuffield Orthopaedic Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Andrew Carr
- The Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| |
Collapse
|
71
|
Paterson YZ, Cribbs A, Espenel M, Smith EJ, Henson FMD, Guest DJ. Genome-wide transcriptome analysis reveals equine embryonic stem cell-derived tenocytes resemble fetal, not adult tenocytes. Stem Cell Res Ther 2020; 11:184. [PMID: 32430075 PMCID: PMC7238619 DOI: 10.1186/s13287-020-01692-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Tendon injuries occur frequently in human and equine athletes. Treatment options are limited, and the prognosis is often poor with functionally deficient scar tissue resulting. Fetal tendon injuries in contrast are capable of healing without forming scar tissue. Embryonic stem cells (ESCs) may provide a potential cellular therapeutic to improve adult tendon regeneration; however, whether they can mimic the properties of fetal tenocytes is unknown. To this end, understanding the unique expression profile of normal adult and fetal tenocytes is crucial to allow validation of ESC-derived tenocytes as a cellular therapeutic. METHODS Equine adult, fetal and ESC-derived tenocytes were cultured in a three-dimensional environment, with histological, morphological and transcriptomic differences compared. Additionally, the effects on gene expression of culturing adult and fetal tenocytes in either conventional two-dimensional monolayer culture or three-dimensional culture were compared using RNA sequencing. RESULTS No qualitative differences in three-dimensional tendon constructs generated from adult, fetal and ESCs were found using histological and morphological analysis. However, genome-wide transcriptomic analysis using RNA sequencing revealed that ESC-derived tenocytes' transcriptomic profile more closely resembled fetal tenocytes as opposed to adult tenocytes. Furthermore, this study adds to the growing evidence that monolayer cultured cells' gene expression profiles converge, with adult and fetal tenocytes having only 10 significantly different genes when cultured in this manner. In contrast, when adult and fetal tenocytes were cultured in 3D, large distinctions in gene expression between these two developmental stages were found, with 542 genes being differentially expressed. CONCLUSION The information provided in this study makes a significant contribution to the investigation into the differences between adult reparative and fetal regenerative cells and supports the concept of using ESC-derived tenocytes as a cellular therapy. Comparing two- and three-dimensional culture also indicates three-dimensional culture as being a more physiologically relevant culture system for determining transcriptomic difference between the same cell types from different developmental stages.
Collapse
Affiliation(s)
- Y. Z. Paterson
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES UK
- Centre for Preventive Medicine, Animal Health Trust, Lanwades Park, Kentford, Newmarket, Suffolk, CB8 7UU UK
| | - A. Cribbs
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD UK
| | - M. Espenel
- Centre for Preventive Medicine, Animal Health Trust, Lanwades Park, Kentford, Newmarket, Suffolk, CB8 7UU UK
| | - E. J. Smith
- Centre for Preventive Medicine, Animal Health Trust, Lanwades Park, Kentford, Newmarket, Suffolk, CB8 7UU UK
| | - F. M. D. Henson
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES UK
- Centre for Preventive Medicine, Animal Health Trust, Lanwades Park, Kentford, Newmarket, Suffolk, CB8 7UU UK
| | - D. J. Guest
- Centre for Preventive Medicine, Animal Health Trust, Lanwades Park, Kentford, Newmarket, Suffolk, CB8 7UU UK
| |
Collapse
|
72
|
Lin J, Chen L, Dou D. Progress of orthopaedic research in China over the last decade. J Orthop Translat 2020; 24:131-137. [PMID: 32913711 PMCID: PMC7452214 DOI: 10.1016/j.jot.2020.04.010] [Citation(s) in RCA: 4] [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: 03/19/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
Abstract
Objective To summarize the representative scientific achievements in the past decade, and discuss the future challenges and directions for orthopaedic research in China. Methods In this review, we used the data provided by National Natural Science Foundation of China (NSFC) for analysis. Results Over the last decade, NSFC has initiated various research programs with a total funding of over 1149 million RMB to support orthopaedic exploration. Under the strong support of NSFC, great progresses have been made in basic research, talent training, platform construction and the clinical translation in the field of orthopaedics in China. Conclusion In general, since the establishment of the Department of Health Sciences of NSFC 10 years ago, both the amount of funding and the scale of researchers in the field of orthopaedic research have increased substantially. Despite of several shortcomings in orthopaedic research, with continuous support from NSFC both in funding and in policy, we believe that the orthopaedic research in China will surely make steady and significant progress. The translational potential of this article This article summarizes the representative scientific achievements in the past decade and puts forward the future challenges and directions for orthopaedic research in China.
Collapse
Affiliation(s)
- Jun Lin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dou Dou
- Department of Health Sciences, National Natural Science Foundation of China, Beijing, China
| |
Collapse
|
73
|
Wang D, Pun CCM, Huang S, Tang TCM, Ho KKW, Rothrauff BB, Yung PSH, Blocki AM, Ker EDF, Tuan RS. Tendon-derived extracellular matrix induces mesenchymal stem cell tenogenesis via an integrin/transforming growth factor-β crosstalk-mediated mechanism. FASEB J 2020; 34:8172-8186. [PMID: 32301551 DOI: 10.1096/fj.201902377rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/26/2022]
Abstract
Treatment of tendon injuries is challenging. To develop means to augment tendon regeneration, we have previously prepared a soluble, low immunogenic (DNA-free), tendon extracellular matrix fraction (tECM) by urea extraction of juvenile bovine tendons, which is capable of enhancing transforming growth factor-β (TGF-β) mediated tenogenesis in human adipose-derived stem cells (hASCs). Here, we aimed to elucidate the mechanism of tECM-driven hASC tenogenic differentiation in vitro, focusing on the integrin and TGF-β/SMAD pathways. Our results showed that tECM promoted hASC proliferation and tenogenic differentiation in vitro based on tenogenesis-associated markers. tECM also induced higher expression of several integrin subunits and TGF-β receptors, and nuclear translocation of p-SMAD2 in hASCs. Pharmacological inhibition of integrin-ECM binding, focal adhesion kinase (FAK) signaling, or TGF-β signaling independently led to compromised pro-tenogenic effects of tECM and actin fiber polymerization. Additionally, integrin blockade inhibited tECM-driven TGFBR2 expression, while inhibiting TGF-β signaling decreased tECM-mediated expression of integrin α1, α2, and β1 in hASCs. Together, these findings suggest that the strong pro-tenogenic bioactivity of tECM is regulated via integrin/TGF-β signaling crosstalk. Understanding how integrins interact with signaling by TGF-β and/or other growth factors (GFs) within the tendon ECM microenvironment will provide a rational basis for an ECM-based approach for tendon repair.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Charmaine C M Pun
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Thomas C M Tang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kevin K W Ho
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Benjamin B Rothrauff
- Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick S H Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Anna M Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Elmer D F Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
74
|
No YJ, Tarafder S, Reischl B, Ramaswamy Y, Dunstan C, Friedrich O, Lee CH, Zreiqat H. High-Strength Fiber-Reinforced Composite Hydrogel Scaffolds as Biosynthetic Tendon Graft Material. ACS Biomater Sci Eng 2020; 6:1887-1898. [PMID: 33455306 DOI: 10.1021/acsbiomaterials.9b01716] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The development of suitable synthetic scaffolds for use as human tendon grafts to repair tendon ruptures remains a significant engineering challenge. Previous synthetic tendon grafts have demonstrated suboptimal tissue ingrowth and synovitis due to wear particles from fiber-to-fiber abrasion. In this study, we present a novel fiber-reinforced hydrogel (FRH) that mimics the hierarchical structure of the native human tendon for synthetic tendon graft material. Ultrahigh molecular weight polyethylene (UHMWPE) fibers were impregnated with either biosynthetic polyvinyl alcohol/gelatin hydrogel (FRH-PG) or with polyvinyl alcohol/gelatin + strontium-hardystonite (Sr-Ca2ZnSi2O7, Sr-HT) composite hydrogel (FRH-PGS). The scaffolds were fabricated and assessed to evaluate their suitability for tendon graft applications. The microstructure of both FRH-PG and FRH-PGS showed successful impregnation of the hydrogel component, and the tendon scaffolds exhibited equilibrium water content of ∼70 wt %, similar to the values reported for native human tendon, compared to ∼50 wt % water content retained in unmodified UHMWPE fibers. The tensile strength of FRH-PG and FRH-PGS (77.0-81.8 MPa) matched the range of human Achilles' tendon tensile strengths reported in the literature. In vitro culture of rat tendon stem cells showed cell and tissue infiltration into both FRH-PG and FRH-PGS after 2 weeks, and the presence of Sr-HT ceramic particles influenced the expression of tenogenic markers. On the other hand, FRH-PG supported the proliferation of murine C2C12 myoblasts, whereas FRH-PGS seemingly did not support it under static culture conditions. In vivo implantation of FRH-PG and FRH-PGS scaffolds into full-thickness rat patellar tendon defects showed good collagenous tissue ingrowth into these scaffolds after 6 weeks. This study demonstrates the potential viability for our FRH-PG and FRH-PGS scaffolds to be used for off-the-shelf biosynthetic tendon graft material.
Collapse
Affiliation(s)
- Young Jung No
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney 2006, Australia.,Australian Research Council Training Centre for Innovative BioEngineering, Sydney 2006, Australia
| | - Solaiman Tarafder
- Regenerative Engineering Laboratory, Columbia University, New York 10032, New York, United States
| | - Barbara Reischl
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen 91052, Germany
| | - Yogambha Ramaswamy
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney 2006, Australia.,Australian Research Council Training Centre for Innovative BioEngineering, Sydney 2006, Australia
| | - Colin Dunstan
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney 2006, Australia.,Australian Research Council Training Centre for Innovative BioEngineering, Sydney 2006, Australia
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen 91052, Germany
| | - Chang Hun Lee
- Regenerative Engineering Laboratory, Columbia University, New York 10032, New York, United States
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney 2006, Australia.,Australian Research Council Training Centre for Innovative BioEngineering, Sydney 2006, Australia
| |
Collapse
|
75
|
Chen M, Li Y, Xiao L, Dai G, Lu P, Wang Y, Rui Y. AQP1 modulates tendon stem/progenitor cells senescence during tendon aging. Cell Death Dis 2020; 11:193. [PMID: 32188840 PMCID: PMC7080760 DOI: 10.1038/s41419-020-2386-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
The link between tendon stem/progenitor cells (TSPCs) senescence and tendon aging has been well recognized. However, the cellular and molecular mechanisms of TSPCs senescence are still not fully understood. In present study, we investigated the role of Aquaporin 1 (AQP1) in TSPCs senescence. We showed that AQP1 expression declines with age during tendon aging. In aged TSPCs, overexpression of AQP1 significantly attenuated TSPCs senescence. In addition, AQP1 overexpression also restored the age-related dysfunction of self-renewal, migration and tenogenic differentiation. Furthermore, we demonstrated that the JAK-STAT signaling pathway is activated in aged TSPCs, and AQP1 overexpression inhibited the JAK-STAT signaling pathway activation which indicated that AQP1 attenuates senescence and age-related dysfunction of TSPCs through the repression of JAK-STAT signaling pathway. Taken together, our findings demonstrated the critical role of AQP1 in the regulation of TSPCs senescence and provided a novel target for antagonizing tendon aging.
Collapse
Affiliation(s)
- Minhao Chen
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Yingjuan Li
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
- Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Longfei Xiao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Guangchun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
76
|
Komura S, Satake T, Goto A, Aoki H, Shibata H, Ito K, Hirakawa A, Yamada Y, Akiyama H. Induced pluripotent stem cell-derived tenocyte-like cells promote the regeneration of injured tendons in mice. Sci Rep 2020; 10:3992. [PMID: 32132649 PMCID: PMC7055210 DOI: 10.1038/s41598-020-61063-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/20/2020] [Indexed: 12/17/2022] Open
Abstract
Tendons are dense fibrous structures that attach muscles to bones. Healing of tendon injuries is a clinical challenge owing to poor regenerative potential and scarring. Here, we created reporter mice that express EGFP, driven by the promoter of the tendon-specific Scleraxis (Scx) transcription-factor gene; we then generated induced pluripotent stem cells (iPSCs) from these mice. Utilising these fluorescently labelled iPSCs, we developed a tenogenic differentiation protocol. The iPSC-derived EGFP-positive cells exhibited elevated expression of tendon-specific genes, including Scx, Mohawk, Tenomodulin, and Fibromodulin, indicating that they have tenocyte-like properties. Finally, we demonstrated that these cells promoted tendon regeneration in mice after transplantation into injured tendons reducing scar formation via paracrine effect. Our data demonstrate that the tenogenic differentiation protocol successfully provided functional cells from iPSCs. We propose that pluripotent stem cell-based therapy using this protocol will provide an effective therapeutic approach for tendon injuries.
Collapse
Affiliation(s)
- Shingo Komura
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan. .,Laboratory of Stem Cell Oncology, Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.
| | - Takashi Satake
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Atsushi Goto
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Hitomi Aoki
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Hirofumi Shibata
- Laboratory of Stem Cell Oncology, Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Kenji Ito
- Laboratory of Stem Cell Oncology, Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.,Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Akihiro Hirakawa
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Yasuhiro Yamada
- Laboratory of Stem Cell Oncology, Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.,Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Haruhiko Akiyama
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| |
Collapse
|
77
|
Qi F, Deng Z, Ma Y, Wang S, Liu C, Lyu F, Wang T, Zheng Q. From the perspective of embryonic tendon development: various cells applied to tendon tissue engineering. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:131. [PMID: 32175424 DOI: 10.21037/atm.2019.12.78] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is a high risk of injury from damage to the force-bearing tissue of the tendon. Due to its poor self-healing ability, clinical interventions for tendon injuries are limited and yield unsatisfying results. Tissue engineering might supply an alternative to this obstacle. As one of the key elements of tissue engineering, various cell sources have been used for tendon engineering, but there is no consensue concerning a single optimal source. In this review, we summarized the development of tendon tissue from the embryonic stage and categorized the used cell sources in tendon engineering. By comparing various cell sources as the candidates for tendon regeneration, each cell type was found to have its advantages and limitations; therefore, it is difficult to define the best cell source for tendon engineering. The microenvironment cells located is also crucial for cell growth and differentiation; so, the optimal cells are unlikely to be the same for each patient. In the future, the clinical application of tendon engineering might be more precise and customized in contrast to the current use of a standardized/generic one-size-fits-all procedure. The best cell source for tendon engineering will require a case-based assessment.
Collapse
Affiliation(s)
- Fangjie Qi
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Chang Liu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Fengjuan Lyu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Tao Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China.,Centre for Orthopaedic Translational Research, School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China.,Centre for Orthopaedic Translational Research, School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
78
|
Zhou Y, Zhou J, Sun B, Xu W, Zhong M, Li Y, He C, Chen Y, Wang X, Jones PM, Sun Z. Vitamin A deficiency causes islet dysfunction by inducing islet stellate cell activation via cellular retinol binding protein 1. Int J Biol Sci 2020; 16:947-956. [PMID: 32140064 PMCID: PMC7053333 DOI: 10.7150/ijbs.37861] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Vitamin A (VA) plays an essential role in pancreatic homeostasis. Islet stellate cells (ISCs) are VA-storing cells in pancreatic islets. Herein, we have investigated the effect of VA on glucose homeostasis trough regulation of ISCs function in dietary VA deficiency model mice. Methods: Male C57BL/6 mice were randomly fed a VA-sufficient, a VA-deficient (VAD) or a VAD-rescued diet. Glucose metabolism was assessed by glucose tolerance tests and immunohistochemistry. ISCs activation degree was evaluated by immunofluorescence, quantitative PCR and western blotting in both, retinol-treated cultured ISCs and model mice. Changes in ISCs phenotype and their effect on islets were assessed by lentiviral transduction and enzyme-linked immunosorbent assays in a co-culture system. Results: VAD mice showed irregular shaped islet, glucose intolerance, islet size distribution excursions, and upregulated expression of α-smooth muscle actin (α-SMA, marker of ISCs activation). Reintroduction of dietary VA restored pancreatic VA levels, endocrine hormone profiles, and inhibited ISCs activation. Incubation with retinol increased the expression of VA signaling factors in ISCs, including cellular retinol binding protein 1 (CRBP1). The knockdown of CRBP1 maintained the quiescent ISCs phenotype and reduced the damage of activated ISCs on islet function. Conclusions: VA deficiency reduced islet function by activating ISCs in VAD mice. Restoring ISCs quiescence via CRBP1 inhibition could reverse the impairment of islet function caused by activated ISCs exposure.
Collapse
Affiliation(s)
- Yunting Zhou
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Junming Zhou
- Department of Gastroenterology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bo Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Wei Xu
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London, UK
| | - Ming Zhong
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
| | - Yumin Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Cong He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yang Chen
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Xiaohang Wang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Peter M Jones
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London, UK
| | - Zilin Sun
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
79
|
Steinmann S, Pfeifer CG, Brochhausen C, Docheva D. Spectrum of Tendon Pathologies: Triggers, Trails and End-State. Int J Mol Sci 2020; 21:ijms21030844. [PMID: 32013018 PMCID: PMC7037288 DOI: 10.3390/ijms21030844] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/18/2020] [Accepted: 01/24/2020] [Indexed: 12/31/2022] Open
Abstract
The biggest compartment of the musculoskeletal system is the tendons and ligaments. In particular, tendons are dense tissues connecting muscle to bone that are critical for the integrity, function and locomotion of this system. Due to the increasing age of our society and the overall rise in engagement in extreme and overuse sports, there is a growing prevalence of tendinopathies. Despite the recent advances in tendon research and due to difficult early diagnosis, a multitude of risk factors and vague understanding of the underlying biological mechanisms involved in the progression of tendon injuries, the toolbox of treatment strategies remains limited and non-satisfactory. This review is designed to summarize the current knowledge of triggers, trails and end state of tendinopathies.
Collapse
Affiliation(s)
- Sara Steinmann
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Am Biopark 9, 93053 Regensburg, Germany; (S.S.); (C.G.P.)
| | - Christian G. Pfeifer
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Am Biopark 9, 93053 Regensburg, Germany; (S.S.); (C.G.P.)
- Department of Trauma Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany;
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Am Biopark 9, 93053 Regensburg, Germany; (S.S.); (C.G.P.)
- Department of Medical Biology, Medical University-Plovdiv, 15A Vassil Aprilov Blvd., 4002 Plovdiv, Bulgaria
- Correspondence: ; Tel.: +49 941 943-1605
| |
Collapse
|
80
|
Tan GK, Pryce BA, Stabio A, Brigande JV, Wang C, Xia Z, Tufa SF, Keene DR, Schweitzer R. Tgfβ signaling is critical for maintenance of the tendon cell fate. eLife 2020; 9:52695. [PMID: 31961320 PMCID: PMC7025861 DOI: 10.7554/elife.52695] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
Studies of cell fate focus on specification, but little is known about maintenance of the differentiated state. In this study, we find that the mouse tendon cell fate requires continuous maintenance in vivo and identify an essential role for TGFβ signaling in maintenance of the tendon cell fate. To examine the role of TGFβ signaling in tenocyte function the TGFβ type II receptor (Tgfbr2) was targeted in the Scleraxis-expressing cell lineage using the ScxCre deletor. Tendon development was not disrupted in mutant embryos, but shortly after birth tenocytes lost differentiation markers and reverted to a more stem/progenitor state. Viral reintroduction of Tgfbr2 to mutants prevented and even rescued tenocyte dedifferentiation suggesting a continuous and cell autonomous role for TGFβ signaling in cell fate maintenance. These results uncover the critical importance of molecular pathways that maintain the differentiated cell fate and a key role for TGFβ signaling in these processes.
Collapse
Affiliation(s)
- Guak-Kim Tan
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Brian A Pryce
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Anna Stabio
- Research Division, Shriners Hospital for Children, Portland, United States
| | - John V Brigande
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, United States
| | - ChaoJie Wang
- Computational Biology Program, Oregon Health & Science University, Portland, United States
| | - Zheng Xia
- Computational Biology Program, Oregon Health & Science University, Portland, United States
| | - Sara F Tufa
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Douglas R Keene
- Research Division, Shriners Hospital for Children, Portland, United States
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, United States.,Department of Orthopedics, Oregon Health & Science University, Portland, United States
| |
Collapse
|
81
|
Liao Y, Li G, Zhang X, Huang W, Xie D, Dai G, Zhu S, Lu D, Zhang Z, Lin J, Wu B, Lin W, Chen Y, Chen Z, Peng C, Wang M, Chen X, Jiang MH, Xiang AP. Cardiac Nestin + Mesenchymal Stromal Cells Enhance Healing of Ischemic Heart through Periostin-Mediated M2 Macrophage Polarization. Mol Ther 2020; 28:855-873. [PMID: 31991111 DOI: 10.1016/j.ymthe.2020.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) show potential for treating cardiovascular diseases, but their therapeutic efficacy exhibits significant heterogeneity depending on the tissue of origin. This study sought to identify an optimal source of MSCs for cardiovascular disease therapy. We demonstrated that Nestin was a suitable marker for cardiac MSCs (Nes+cMSCs), which were identified by their self-renewal ability, tri-lineage differentiation potential, and expression of MSC markers. Furthermore, compared with bone marrow-derived MSCs (Nes+bmMSCs) or saline-treated myocardial infarction (MI) controls, intramyocardial injection of Nes+cMSCs significantly improved cardiac function and decreased infarct size after acute MI (AMI) through paracrine actions, rather than transdifferentiation into cardiac cells in infarcted heart. We further revealed that Nes+cMSC treatment notably reduced pan-macrophage infiltration while inducing macrophages toward an anti-inflammatory M2 phenotype in ischemic myocardium. Interestingly, Periostin, which was highly expressed in Nes+cMSCs, could promote the polarization of M2-subtype macrophages, and knockdown or neutralization of Periostin remarkably reduced the therapeutic effects of Nes+cMSCs by decreasing M2 macrophages at lesion sites. Thus, the present work systemically shows that Nes+cMSCs have greater efficacy than do Nes+bmMSCs for cardiac healing after AMI, and that this occurs at least partly through Periostin-mediated M2 macrophage polarization.
Collapse
Affiliation(s)
- Yan Liao
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510623, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Guilan Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Xiaoran Zhang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510623, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weijun Huang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510623, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Dongmei Xie
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China; Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Gang Dai
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Shuanghua Zhu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Dihan Lu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhongyuan Zhang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510623, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Junyi Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Bingyuan Wu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wanwen Lin
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yang Chen
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhihong Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chaoquan Peng
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Maosheng Wang
- The Cardiovascular Center, Gaozhou People's Hospital, Maoming, Guangdong 525200, China
| | - Xinxin Chen
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510623, China.
| | - Mei Hua Jiang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510623, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Andy Peng Xiang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510623, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510080, China.
| |
Collapse
|
82
|
Wunderli SL, Blache U, Snedeker JG. Tendon explant models for physiologically relevant invitro study of tissue biology - a perspective. Connect Tissue Res 2020; 61:262-277. [PMID: 31931633 DOI: 10.1080/03008207.2019.1700962] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background: Tendon disorders increasingly afflict our aging society but we lack the scientific understanding to clinically address them. Clinically relevant models of tendon disease are urgently needed as established small animal models of tendinopathy fail to capture essential aspects of the disease. Two-dimensional and three-dimensional cell and tissue culture models are similarly limited, lacking many physiological extracellular matrix cues required to maintain tissue homeostasis or guide matrix remodeling. These cues reflect the biochemical and biomechanical status of the tissue, and encode information regarding the mechanical and metabolic competence of the tissue. Tendon explants overcome some of these limitations and have thus emerged as a valuable tool for the discovery and study of mechanisms associated with tendon homeostasis and pathophysiology. Tendon explants retain native cell-cell and cell-matrix connections, while allowing highly reproducible experimental control over extrinsic factors like mechanical loading and nutritional availability. In this sense tendon explant models can deliver insights that are otherwise impossible to obtain from in vivo animal or in vitro cell culture models. Purpose: In this review, we aimed to provide an overview of tissue explant models used in tendon research, with a specific focus on the value of explant culture systems for the controlled study of the tendon core tissue. We discuss their advantages, limitations and potential future utility. We include suggestions and technical recommendations for the successful use of tendon explant cultures and conclude with an outlook on how explant models may be leveraged with state-of-the-art biotechnologies to propel our understanding of tendon physiology and pathology.
Collapse
Affiliation(s)
- Stefania L Wunderli
- University Hospital Balgrist, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ulrich Blache
- University Hospital Balgrist, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Jess G Snedeker
- University Hospital Balgrist, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
83
|
Leong NL, Kator JL, Clemens TL, James A, Enamoto-Iwamoto M, Jiang J. Tendon and Ligament Healing and Current Approaches to Tendon and Ligament Regeneration. J Orthop Res 2020; 38:7-12. [PMID: 31529731 PMCID: PMC7307866 DOI: 10.1002/jor.24475] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 09/10/2019] [Indexed: 02/04/2023]
Abstract
Ligament and tendon injuries are common problems in orthopedics. There is a need for treatments that can expedite nonoperative healing or improve the efficacy of surgical repair or reconstruction of ligaments and tendons. Successful biologically-based attempts at repair and reconstruction would require a thorough understanding of normal tendon and ligament healing. The inflammatory, proliferative, and remodeling phases, and the cells involved in tendon and ligament healing will be reviewed. Then, current research efforts focusing on biologically-based treatments of ligament and tendon injuries will be summarized, with a focus on stem cells endogenous to tendons and ligaments. Statement of clinical significance: This paper details mechanisms of ligament and tendon healing, as well as attempts to apply stem cells to ligament and tendon healing. Understanding of these topics could lead to more efficacious therapies to treat ligament and tendon injuries. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:7-12, 2020.
Collapse
Affiliation(s)
- Natalie L Leong
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
- Department of Surgery, Baltimore VA Medical Center, Baltimore, Maryland
| | - Jamie L Kator
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Aaron James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Motomi Enamoto-Iwamoto
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
| | - Jie Jiang
- Department of Orthopaedic Surgery, University of Maryland, 10 N. Greene St., Baltimore, Maryland, 21201
| |
Collapse
|
84
|
Abstract
Tendons connect muscles to bones to transfer the forces necessary for movement. Cell-cell junction proteins, cadherins and connexins, may play a role in tendon development and injury. In this review, we begin by highlighting current understanding of how cell-cell junctions may regulate embryonic tendon development and differentiation. We then examine cell-cell junctions in postnatal tendon, before summarizing the role of cadherins and connexins in adult tendons. More information exists regarding the role of cell-cell junctions in the formation and homeostasis of other musculoskeletal tissues, namely cartilage and bone. Therefore, to inform future tendon studies, we include a brief survey of cadherins and connexins in chondrogenesis and osteogenesis, and summarize how cell-cell junctions are involved in some musculoskeletal tissue pathologies. An enhanced understanding of how cell-cell junctions participate in tendon development, maintenance, and disease will benefit future regenerative strategies.
Collapse
Affiliation(s)
| | - Jett B Murray
- Biological Engineering, University of Idaho, Moscow, ID
| | | |
Collapse
|
85
|
Abstract
Tendons link muscle to bone and transfer forces necessary for normal movement. Tendon injuries can be debilitating and their intrinsic healing potential is limited. These challenges have motivated the development of model systems to study the factors that regulate tendon formation and tendon injury. Recent advances in understanding of embryonic and postnatal tendon formation have inspired approaches that aimed to mimic key aspects of tendon development. Model systems have also been developed to explore factors that regulate tendon injury and healing. We highlight current model systems that explore developmentally inspired cellular, mechanical, and biochemical factors in tendon formation and tenogenic stem cell differentiation. Next, we discuss in vivo, in vitro, ex vivo, and computational models of tendon injury that examine how mechanical loading and biochemical factors contribute to tendon pathologies and healing. These tendon development and injury models show promise for identifying the factors guiding tendon formation and tendon pathologies, and will ultimately improve regenerative tissue engineering strategies and clinical outcomes.
Collapse
Affiliation(s)
- Sophia K Theodossiou
- Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, Moscow, ID 83844, USA
| | - Nathan R Schiele
- Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, Moscow, ID 83844, USA
| |
Collapse
|
86
|
Tarafder S, Brito JA, Minhas S, Effiong L, Thomopoulos S, Lee CH. In situ tissue engineering of the tendon-to-bone interface by endogenous stem/progenitor cells. Biofabrication 2019; 12:015008. [PMID: 31561236 PMCID: PMC6904927 DOI: 10.1088/1758-5090/ab48ca] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The long-term success of surgical repair of rotator cuff tears is largely dependent on restoration of a functional tendon-to-bone interface. We implemented micro-precise spatiotemporal delivery of growth factors in three-dimensional printed scaffolds for integrative regeneration of a fibrocartilaginous tendon-to-bone interface. Sustained and spatially controlled release of tenogenic, chondrogenic and osteogenic growth factors was achieved using microsphere-based delivery carriers embedded in thin membrane-like scaffolds. In vitro, the scaffolds embedded with spatiotemporal delivery of growth factors successfully guided regional differentiation of mesenchymal progenitor cells, forming multiphase tissues with tendon-like, cartilage-like and bone-like regions. In vivo, when implanted at the interface between the supraspinatus tendon and the humeral head in a rat rotator cuff repair model, these scaffolds promoted recruitment of endogenous tendon progenitor cells followed by integrative healing of tendon and bone via re-formation of strong fibrocartilaginous interfaces. Our findings demonstrate the potential of in situ tissue engineering of tendon-to-bone interfaces by endogenous progenitor cells. The in situ tissue engineering approach shows translational potential for improving outcomes after rotator cuff repair.
Collapse
Affiliation(s)
- Solaiman Tarafder
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| | - John A Brito
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| | - Sumeet Minhas
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| | - Linda Effiong
- Department of Orthopedic Surgery, Columbia University Medical Center, 650 W. 168th Street, BB14-1408, NY 10032, New York
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University Medical Center, 650 W. 168th Street, BB14-1408, NY 10032, New York
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, NY 10027, New York
| | - Chang H Lee
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| |
Collapse
|
87
|
Rui YF, Chen MH, Li YJ, Xiao LF, Geng P, Wang P, Xu ZY, Zhang XP, Dai GC. CTGF Attenuates Tendon-Derived Stem/Progenitor Cell Aging. Stem Cells Int 2019; 2019:6257537. [PMID: 31827530 PMCID: PMC6881574 DOI: 10.1155/2019/6257537] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/14/2019] [Accepted: 09/28/2019] [Indexed: 01/18/2023] Open
Abstract
Aged tendon-derived stem/progenitor cells (TSPCs) lead to age-related tendon disorders and impair tendon healing. However, the underlying molecular mechanisms of TSPC aging remain largely unknown. Here, we investigated the role of connective tissue growth factor (CTGF) in TSPC aging. CTGF protein and mRNA levels were markedly decreased in the aged TSPCs. Moreover, recombinant CTGF attenuates TSPC aging and restores the age-associated reduction of self-renewal and differentiation of TSPCs. In addition, cell cycle distribution of aged TSPCs was arrested in the G1/S phase while recombinant CTGF treatment promoted G1/S transition. Recombinant CTGF also rescued decreased levels of cyclin D1 and CDK4 and reduced p27kip1 expression in aged TSPCs. Our results demonstrated that CTGF plays a vital role in TSPC aging and might be a potential target for molecular therapy of age-related tendon disorders.
Collapse
Affiliation(s)
- Yun-feng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute, Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, Zhejiang 310000, China
| | - Min-hao Chen
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute, Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, Zhejiang 310000, China
| | - Ying-juan Li
- China Orthopedic Regenerative Medicine Group, Hangzhou, Zhejiang 310000, China
- Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Long-fei Xiao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute, Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Peng Geng
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute, Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Pei Wang
- School of Medicine, Southeast University, Nanjing, China
| | - Zheng-yuan Xu
- School of Medicine, Southeast University, Nanjing, China
| | - Xuan-pu Zhang
- School of Medicine, Southeast University, Nanjing, China
| | - Guang-chun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute, Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
88
|
Zhang C, Zhu J, Zhou Y, Thampatty BP, Wang JHC. Tendon Stem/Progenitor Cells and Their Interactions with Extracellular Matrix and Mechanical Loading. Stem Cells Int 2019; 2019:3674647. [PMID: 31737075 PMCID: PMC6815631 DOI: 10.1155/2019/3674647] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/04/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
Tendons are unique connective tissues in the sense that their biological properties are largely determined by their tendon-specific stem cells, extracellular matrix (ECM) surrounding the stem cells, mechanical loading conditions placed on the tendon, and the complex interactions among them. This review is aimed at providing an overview of recent advances in the identification and characterization of tendon stem/progenitor cells (TSPCs) and their interactions with ECM and mechanical loading. In addition, the effects of such interactions on the maintenance of tendon homeostasis and the initiation of tendon pathological conditions are discussed. Moreover, the challenges in further investigations of TSPC mechanobiology in vitro and in vivo are outlined. Finally, future research efforts are suggested, which include using specific gene knockout models and single-cell transcription profiling to enable a broad and deep understanding of the physiology and pathophysiology of tendons.
Collapse
Affiliation(s)
- Chuanxin Zhang
- Joint Surgery and Sports Medicine Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jun Zhu
- Joint Surgery and Sports Medicine Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yiqin Zhou
- Joint Surgery and Sports Medicine Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bhavani P. Thampatty
- MechanoBiology Laboratory, Departments of Orthopaedic Surgery, Bioengineering, and Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James H-C. Wang
- MechanoBiology Laboratory, Departments of Orthopaedic Surgery, Bioengineering, and Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
89
|
Yang F, Zhang A, Richardson DW. Regulation of the tenogenic gene expression in equine tenocyte-derived induced pluripotent stem cells by mechanical loading and Mohawk. Stem Cell Res 2019; 39:101489. [PMID: 31277043 PMCID: PMC7082636 DOI: 10.1016/j.scr.2019.101489] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/14/2019] [Accepted: 06/25/2019] [Indexed: 12/21/2022] Open
Abstract
Cell-based therapeutic strategies afford major potential advantages in the repair of injured tendons. Generation of induced pluripotent stem cells (iPSCs) expands cell sources for “regenerative” therapy. However, its application in tendon repair is still limited and the effects remain unclear. In this study, equine tenocyte-derived iPSCs (teno-iPSCs) were generated by expressing four Yamanaka factors. Compared to parental tenocytes and bone marrow derived mesenchymal stem cells (BMSCs), the transcriptional activities of lineage-specific genes, including Mkx, Col1A2, Col14, DCN, ELN, FMOD, and TNC, were highly repressed in the resulting teno-iPSCs. Exposure to cyclic uniaxial mechanical loading increased the expression of Scx, Egr1, Col1A2, DCN, and TNC in teno-iPSCs and the expression of Scx, Egr1, DCN, and TNC in BMSCs. Reintroduction of tenogenic transcription factor Mohawk (Mkx) upregulated the expression of DCN in teno-iPSCs and the expression of Scx, Col14, and FMOD in BMSCs. Mechanical loading combined with ectopic expression of equine Mkx further enhanced the expression of Egr1, Col1A2, DCN, and TNC in teno-iPSCs and the expression of Scx, Egr1, and TNC in BMSCs. These data suggest that the repressed lineage-specific genes in the teno-iPSCs can be re-activated by mechanical loading and ectopic expression of Mkx. Our findings offer new insights into the application of iPSCs for basic and clinic research in tendon repair.
Collapse
Affiliation(s)
- Feikun Yang
- Department of Clinic Studies at New Bolton Center, University of Pennsylvania, 382 West Street Road, Kennett Square, PA 19348, United States of America.
| | - Aiwu Zhang
- Department of Clinic Studies at New Bolton Center, University of Pennsylvania, 382 West Street Road, Kennett Square, PA 19348, United States of America.
| | - Dean W Richardson
- Department of Clinic Studies at New Bolton Center, University of Pennsylvania, 382 West Street Road, Kennett Square, PA 19348, United States of America.
| |
Collapse
|
90
|
Walia B, Huang AH. Tendon stem progenitor cells: Understanding the biology to inform therapeutic strategies for tendon repair. J Orthop Res 2019; 37:1270-1280. [PMID: 30270569 PMCID: PMC6823601 DOI: 10.1002/jor.24156] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
Tendon and ligament injuries are a leading cause of healthcare visits with significant impact in terms of economic cost and reduced quality of life. To date, reparative strategies remain largely restricted to conservative treatment or surgical repair. However, these therapies fail to restore native tendon structure and function; thus, the tissue may re-rupture or degenerate with time. To improve tendon healing, one promising strategy may be harnessing the innate potential of resident tendon stem/progenitor cells (TSPCs) to guide tenogenic regeneration. In this review, we outline recent advances in the identification and characterization of putative TSPC populations, and discuss biochemical, biomechanical, and biomaterial methods employed for their culture and differentiation. Finally, we identify limitations in our current understanding of TSPC biology, key challenges for their use, and potential therapeutic strategies to inform cell-based tendon repair. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1270-1280, 2019.
Collapse
Affiliation(s)
- Bhavita Walia
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice H. Huang
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
91
|
Chen Y, Xie Y, Liu M, Hu J, Tang C, Huang J, Qin T, Chen X, Chen W, Shen W, Yin Z. Controlled-release curcumin attenuates progression of tendon ectopic calcification by regulating the differentiation of tendon stem/progenitor cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109711. [PMID: 31349489 DOI: 10.1016/j.msec.2019.04.090] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 04/19/2019] [Accepted: 04/29/2019] [Indexed: 01/30/2023]
Abstract
Tendon calcification is a common but intractable problem leading to pain and activity limitation when injury or tendinopathy progresses into the late stage. This is because tendon stem/progenitor cells (TSPCs) can undergo aberrant osteogenic differentiation under inflammatory conditions. This study aims to investigate the effect of curcumin, a natural anti-inflammatory agent, on regulating the differentiation of TSPCs in tendon calcification. With inflammatory stimulation, TSPCs showed higher alkaline phosphatase activity and more frequent formation of mineralized nodules which were verified in the culture system; however, curcumin significantly alleviated these pathological changes. In in vivo function analysis, chitosan microsphere-encapsulated curcumin was delivered to injured sites of rat tendon ectopic calcification model. The inflammation in the tendon tissues of the curcumin group was significantly relieved. Controlled-release curcumin partially rescued tendon calcification and enhanced tendon regeneration in animal model. This study demonstrates that controlled-release curcumin can manipulate the fate decision of TSPCs, and that it promotes the tenogenesis and inhibits the osteogenesis of TSPCs in a pathological microenvironment, which provides a possible new therapeutic strategy for tendon disease.
Collapse
Affiliation(s)
- Yangwu Chen
- School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Orthopaedics Research Institute of Zhejiang Univerisity, China
| | - Yubin Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengfei Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajie Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenqi Tang
- School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Orthopaedics Research Institute of Zhejiang Univerisity, China
| | - Jiayun Huang
- School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Orthopaedics Research Institute of Zhejiang Univerisity, China
| | - Tian Qin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Chen
- School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; 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), China
| | - Weishan Chen
- School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Orthopaedics Research Institute of Zhejiang Univerisity, China.
| | - Weiliang Shen
- School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Orthopaedics Research Institute of Zhejiang Univerisity, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), China.
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
92
|
High-Dimensional Single-Cell Cartography Reveals Novel Skeletal Muscle-Resident Cell Populations. Mol Cell 2019; 74:609-621.e6. [PMID: 30922843 DOI: 10.1016/j.molcel.2019.02.026] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022]
Abstract
Adult tissue repair and regeneration require stem-progenitor cells that can self-renew and generate differentiated progeny. Skeletal muscle regenerative capacity relies on muscle satellite cells (MuSCs) and their interplay with different cell types within the niche. However, our understanding of skeletal muscle tissue cellular composition is limited. Here, using a combined approach of single-cell RNA sequencing and mass cytometry, we precisely mapped 10 different mononuclear cell types in adult mouse muscle. We also characterized gene signatures and determined key discriminating markers of each cell type. We identified two previously understudied cell populations in the interstitial compartment. One expresses the transcription factor scleraxis and generated tenocytes in vitro. The second expresses markers of smooth muscle and mesenchymal cells (SMMCs) and, while distinct from MuSCs, exhibited myogenic potential and promoted MuSC engraftment following transplantation. The blueprint presented here yields crucial insights into muscle-resident cell-type identities and can be exploited to study muscle diseases.
Collapse
|
93
|
Zhou Y, Sun B, Li W, Zhou J, Gao F, Wang X, Cai M, Sun Z. Pancreatic Stellate Cells: A Rising Translational Physiology Star as a Potential Stem Cell Type for Beta Cell Neogenesis. Front Physiol 2019; 10:218. [PMID: 30930789 PMCID: PMC6424017 DOI: 10.3389/fphys.2019.00218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/20/2019] [Indexed: 12/14/2022] Open
Abstract
The progressive decline and eventual loss of islet β-cell function underlies the pathophysiological mechanism of the development of both type 1 and type 2 diabetes mellitus. The recovery of functional β-cells is an important strategy for the prevention and treatment of diabetes. Based on similarities in developmental biology and anatomy, in vivo induction of differentiation of other types of pancreatic cells into β-cells is a promising avenue for future diabetes treatment. Pancreatic stellate cells (PSCs), which have attracted intense research interest due to their effects on tissue fibrosis over the last decade, express multiple stem cell markers and can differentiate into various cell types. In particular, PSCs can successfully differentiate into insulin- secreting cells in vitro and can contribute to tissue regeneration. In this article, we will brings together the main concepts of the translational physiology potential of PSCs that have emerged from work in the field and discuss possible ways to develop the future renewable source for clinical treatment of pancreatic diseases.
Collapse
Affiliation(s)
- Yunting Zhou
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Bo Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Wei Li
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Junming Zhou
- Department of Outpatient, Army Engineering University, Jingling Hospital, Nanjing University, Nanjing, China
| | - Feng Gao
- Graduate Innovation Platform of Southeast University, Nanjing, China
| | - Xiaohang Wang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Min Cai
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| | - Zilin Sun
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
94
|
Multiscale Stem Cell Technologies for Osteonecrosis of the Femoral Head. Stem Cells Int 2019; 2019:8914569. [PMID: 30728843 PMCID: PMC6341242 DOI: 10.1155/2019/8914569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/21/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
The last couple of decades have seen brilliant progress in stem cell therapies, including native, genetically modified, and engineered stem cells, for osteonecrosis of the femoral head (ONFH). In vitro studies evaluate the effect of endogenous or exogenous factor or gene regulation on osteogenic phenotype maintenance and/or differentiation towards osteogenic lineage. The preclinical and clinical outcomes accelerate the clinical translation. Bone marrow mesenchymal stem cells and adipose-derived stem cells have demonstrated better effects in the treatment of femoral head necrosis. Various materials have been used widely in the ONFH treatment in both preclinical and clinical trials. In a word, in vivo and multiscale efforts are expected to overcome obstacles in the approaches for treating ONFH and provide clinical relevance and commercial strategies in the future. Therefore, we will discuss the above aspects in this paper and present our opinions.
Collapse
|
95
|
Ho TC, Tsai SH, Yeh SI, Chen SL, Tung KY, Chien HY, Lu YC, Huang CH, Tsao YP. PEDF-derived peptide promotes tendon regeneration through its mitogenic effect on tendon stem/progenitor cells. Stem Cell Res Ther 2019; 10:2. [PMID: 30606221 PMCID: PMC6318926 DOI: 10.1186/s13287-018-1110-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/04/2018] [Accepted: 12/13/2018] [Indexed: 01/15/2023] Open
Abstract
Background Tendon stem/progenitor cells (TSPC) exhibit a low proliferative response to heal tendon injury, leading to limited regeneration outcomes. Exogenous growth factors that activate TSPC proliferation have emerged as a promising approach for treatment. Here, we evaluated the pigment epithelial-derived factor (PEDF)-derived short peptide (PSP; 29-mer) for treating acute tendon injury and to determine the timing and anatomical features of CD146- and necleostemin-positive TSPC in the tendon healing process. Methods Tendon cells were isolated from rabbit Achilles tendons, stimulated by the 29-mer and analyzed for colony-forming capacity. The expression of the TSPC markers CD146, Oct4, and nestin, induced by the 29-mer, was examined by immunostaining and western blotting. Tendo-Achilles injury was induced in rats by full-thickness insertion of an 18-G needle and immediately treated topically with an alginate gel, loaded with 29-mer. The distribution of TSPC in the injured tendon and their proliferation were monitored using immunohistochemistry with antibodies to CD146 and nucleostemin and by BrdU labeling. Results TSPC markers were enriched among the primary tendon cells when stimulated by the 29-mer. The 29-mer also induced the clonogenicity of CD146+ TSPC, implying TSPC stemness was retained during TSPC expansion in culture. Correspondingly, the expanded TSPC differentiated readily into tenocyte-like cells after removal of the 29-mer from culture. 29-mer/alginate gel treatment caused extensive expansion of CD146+ TSPC in their niche on postoperative day 2, followed by infiltration of CD146+/BrdU− TSPC into the injured tendon on day 7. The nucleostemin+ TSPC were located predominantly in the healing region of the injured tendon in the later phase (day 7) and exhibited proliferative capacity. By 3 weeks, 29-mer-treated tendons showed more organized collagen fiber regeneration and higher tensile strength than control tendons. In culture, the mitogenic effect of the 29-mer was found to be mediated by the phosphorylation of ERK2 and STAT3 in nucleostemin+ TSPC. Conclusions The anatomical analysis of TSPC populations in the wound healing process supports the hypothesis that substantial expansion of resident TSPC by exogenous growth factor is beneficial for tendon healing. The study suggests that synthetic 29-mer peptide may be an innovative therapy for acute tendon rupture.
Collapse
Affiliation(s)
- Tsung-Chuan Ho
- Department of Medical Research, Mackay Memorial Hospital, No. 45, Minsheng Rd., Tamsui District, New Taipei City, 25160, Taiwan
| | - Shawn H Tsai
- Department of Ophthalmology, Mackay Memorial Hospital, No. 92, Sec. 2, Chung Shan North Road, Taipei, 10449, Taiwan.,Department of Optometry, Chung Shan Medical University, Taichung, 40201, Taiwan
| | - Shu-I Yeh
- Department of Ophthalmology, Mackay Memorial Hospital, No. 92, Sec. 2, Chung Shan North Road, Taipei, 10449, Taiwan
| | - Show-Li Chen
- Department of Microbiology, School of Medicine, National Taiwan University, No. 1 Jen Ai road, section 1, Taipei, 100, Taiwan
| | - Kwang-Yi Tung
- Department of Plastic Surgery, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Taipei, 10449, Taiwan
| | - Hsin-Yu Chien
- Department of Ophthalmology, Mackay Memorial Hospital, No. 92, Sec. 2, Chung Shan North Road, Taipei, 10449, Taiwan
| | - Yung-Chang Lu
- Departments of Biomechanics Laboratory, and Orthopaedic Surgery, Mackay Memorial Hospital, No. 45, Minsheng Rd., Tamsui District, New Taipei City, 25160, Taiwan
| | - Chang-Hung Huang
- Departments of Biomechanics Laboratory, and Orthopaedic Surgery, Mackay Memorial Hospital, No. 45, Minsheng Rd., Tamsui District, New Taipei City, 25160, Taiwan.,Department of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yeou-Ping Tsao
- Department of Medical Research, Mackay Memorial Hospital, No. 45, Minsheng Rd., Tamsui District, New Taipei City, 25160, Taiwan. .,Department of Ophthalmology, Mackay Memorial Hospital, No. 92, Sec. 2, Chung Shan North Road, Taipei, 10449, Taiwan.
| |
Collapse
|
96
|
Zhang H, Liu MF, Liu RC, Shen WL, Yin Z, Chen X. Physical Microenvironment-Based Inducible Scaffold for Stem Cell Differentiation and Tendon Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:443-453. [PMID: 29724151 DOI: 10.1089/ten.teb.2018.0018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tendon injuries are common musculoskeletal system disorders, but the tendons have poor regeneration ability. To address this issue, tendon tissue engineering provides potential strategies for future therapeutic treatment. Elements of the physical microenvironment, such as the mechanical force and surface topography, play a vital role in regulating stem cell fate, enhancing the differentiation efficiency of seed cells in tendon tissue engineering. Various inducible scaffolds have been widely explored for tendon regeneration, and scaffold-enhancing modifications have been extensively studied. In this review, we systematically summarize the effects of the physical microenvironment on stem cell differentiation and tendon regeneration; we also provide an overview of the inducible scaffolds for stem cell tenogenic differentiation. Finally, we suggest some potential scaffold-based therapies for tendon injuries, presenting an interesting perspective on tendon regenerative medicine.
Collapse
Affiliation(s)
- Hong Zhang
- 1 School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China .,2 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,3 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Meng-Fei Liu
- 1 School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China .,2 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,3 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China
| | - Ri-Chun Liu
- 4 Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University , Nanning, China
| | - Wei-Liang Shen
- 2 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,5 Department of Sports Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,6 China Orthopedic Regenerative Medicine Group (CORMed) , Hangzhou, China .,7 State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China
| | - Zi Yin
- 1 School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China .,2 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,3 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,6 China Orthopedic Regenerative Medicine Group (CORMed) , Hangzhou, China
| | - Xiao Chen
- 1 School of Basic Medical Sciences, and Department of Orthopedic Surgery of The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China .,2 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,3 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,4 Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University , Nanning, China .,5 Department of Sports Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,6 China Orthopedic Regenerative Medicine Group (CORMed) , Hangzhou, China
| |
Collapse
|
97
|
Piuzzi NS, Dominici M, Long M, Pascual-Garrido C, Rodeo S, Huard J, Guicheux J, McFarland R, Goodrich LR, Maddens S, Robey PG, Bauer TW, Barrett J, Barry F, Karli D, Chu CR, Weiss DJ, Martin I, Jorgensen C, Muschler GF. Proceedings of the signature series symposium "cellular therapies for orthopaedics and musculoskeletal disease proven and unproven therapies-promise, facts and fantasy," international society for cellular therapies, montreal, canada, may 2, 2018. Cytotherapy 2018; 20:1381-1400. [PMID: 30316562 PMCID: PMC8487641 DOI: 10.1016/j.jcyt.2018.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/06/2018] [Indexed: 12/17/2022]
Abstract
The Signature Series Symposium "Cellular Therapies for Orthopaedics and Musculoskeletal Disease Proven and Unproven Therapies-Promise, Facts and Fantasy" was held as a pre-meeting of the 26th International Society for Cellular Therapy (ISCT) annual congress in Montreal, Canada, May 2, 2018. This was the first ISCT program that was entirely dedicated to the advancement of cell-based therapies for musculoskeletal diseases. Cellular therapies in musculoskeletal medicine are a source of great promise and opportunity. They are also the source of public controversy, confusion and misinformation. Patients, clinicians, scientists, industry and government share a commitment to clear communication and responsible development of the field. Therefore, this symposium convened thought leaders from around the world in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value to patients with musculoskeletal conditions.
Collapse
Affiliation(s)
- Nicolas S Piuzzi
- Department of Orthopedic Surgery and Biomedical Engineering Cleveland Clinic, Cleveland, Ohio, USA; Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Marc Long
- MTF Biologics, Edison, New Jersey, USA
| | - Cecilia Pascual-Garrido
- Adult Reconstruction-Adolescent and Young Adult Hip Service, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Scott Rodeo
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, UTHealth Medical School, Houston, Texas, USA; Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Jérome Guicheux
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Nantes University School of Dental Medicine, ONIRIS, Nantes, France; CHU Nantes, PHU4 OTONN, Nantes, France
| | - Richard McFarland
- Advanced Regenerative Manufacturing Institute, Manchester, New Hampshire, USA, and Standards Coordinating Body, Gaithersburg, Maryland, USA
| | - Laurie R Goodrich
- Orthopaedic Research Center and Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | | | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Thomas W Bauer
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, New York, USA
| | - John Barrett
- Stem Cell Allogeneic Transplant Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Frank Barry
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - David Karli
- Steadman Philippon Research Institute, Vail, Colorado, USA; Greyledge Technologies, LLC, Vail, Colorado, USA
| | - Constance R Chu
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Daniel J Weiss
- University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Ivan Martin
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Christian Jorgensen
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - George F Muschler
- Department of Orthopedic Surgery and Biomedical Engineering Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
98
|
Delgado Caceres M, Pfeifer CG, Docheva D. Understanding Tendons: Lessons from Transgenic Mouse Models. Stem Cells Dev 2018; 27:1161-1174. [PMID: 29978741 PMCID: PMC6121181 DOI: 10.1089/scd.2018.0121] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/05/2018] [Indexed: 12/26/2022] Open
Abstract
Tendons and ligaments are connective tissues that have been comparatively less studied than muscle and cartilage/bone, even though they are crucial for proper function of the musculoskeletal system. In tendon biology, considerable progress has been made in identifying tendon-specific genes (Scleraxis, Mohawk, and Tenomodulin) in the past decade. However, besides tendon function and the knowledge of a small number of important players in tendon biology, neither the ontogeny of the tenogenic lineage nor signaling cascades have been fully understood. This results in major drawbacks in treatment and repair options following tendon degeneration. In this review, we have systematically evaluated publications describing tendon-related genes, which were studied in depth and characterized by using knockout technologies and the subsequently generated transgenic mouse models (Tg) (knockout mice, KO). We report in a tabular manner, that from a total of 24 tendon-related genes, in 22 of the respective knockout mouse models, phenotypic changes were detected. Additionally, in some of the models it was described at which developmental stages these changes appeared and progressed. To summarize, only loss of Scleraxis and TGFβ signaling led to severe tendon developmental phenotypes, while mice deficient for various proteoglycans, Mohawk, EGR1 and 2, and Tenomodulin presented mild phenotypes. These data suggest that the tendon developmental system is well organized, orchestrated, and backed up; this is even more evident among the members of the proteoglycan family, where the compensatory effects are much clearer. In future, it will be of great importance to discover additional master tendon transcription factors and the genes that play crucial roles in tendon development. This would improve our understanding of the genetic makeup of tendons, and will increase the chances of generating tendon-specific drugs to advance overall treatment strategies.
Collapse
Affiliation(s)
- Manuel Delgado Caceres
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Christian G. Pfeifer
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Department of Medical Biology, Medical University-Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
99
|
Bianco ST, Moser HL, Galatz LM, Huang AH. Biologics and stem cell-based therapies for rotator cuff repair. Ann N Y Acad Sci 2018; 1442:35-47. [PMID: 30008172 DOI: 10.1111/nyas.13918] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
The rotator cuff is composed of several distinct muscles and tendons that function in concert to coordinate shoulder motion. Injuries to these tendons frequently result in permanent dysfunction and persistent pain. Despite considerable advances in operation techniques, surgical repair alone still does not fully restore rotator cuff function. This review focuses on recent research in the use of biologics and stem cell-based therapies to augment repair, highlighting promising avenues for future work and remaining challenges. While a number of animal models are used for rotator cuff studies, the anatomy of the rotator cuff varies dramatically between species. Since the rodent rotator cuff shares the most anatomical features with the human, this review will focus primarily on rodent models to enable consistent interpretation of outcome measures.
Collapse
Affiliation(s)
- Spencer T Bianco
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Helen L Moser
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York.,Shoulder, Elbow and Orthopaedic Sports Medicine, Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Leesa M Galatz
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
100
|
Zhang YJ, Chen X, Li G, Chan KM, Heng BC, Yin Z, Ouyang HW. Concise Review: Stem Cell Fate Guided By Bioactive Molecules for Tendon Regeneration. Stem Cells Transl Med 2018; 7:404-414. [PMID: 29573225 PMCID: PMC5905226 DOI: 10.1002/sctm.17-0206] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022] Open
Abstract
Tendon disorders, which are commonly presented in the clinical setting, disrupt the patients' normal work and life routines, and they damage the careers of athletes. However, there is still no effective treatment for tendon disorders. In the field of tissue engineering, the potential of the therapeutic application of exogenous stem cells to treat tendon pathology has been demonstrated to be promising. With the development of stem cell biology and chemical biology, strategies that use inductive tenogenic factors to program stem cell fate in situ are the most easily and readily translatable to clinical applications. In this review, we focus on bioactive molecules that can potentially induce tenogenesis in adult stem cells, and we summarize the various differentiation factors found in comparative studies. Moreover, we discuss the molecular regulatory mechanisms of tenogenesis, and we examine the various challenges in developing standardized protocols for achieving efficient and reproducible tenogenesis. Finally, we discuss and predict future directions for tendon regeneration. Stem Cells Translational Medicine 2018;7:404-414.
Collapse
Affiliation(s)
- Yan-Jie Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| | - Gang Li
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Kai-Ming Chan
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Boon Chin Heng
- Faculty of Dentistry, Department of Endodontology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Hong-Wei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| |
Collapse
|