51
|
Nichols AEC, Werre SR, Dahlgren LA. Transient Scleraxis Overexpression Combined with Cyclic Strain Enhances Ligament Cell Differentiation. Tissue Eng Part A 2018; 24:1444-1455. [PMID: 29644940 DOI: 10.1089/ten.tea.2017.0481] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Efforts to generate tissue-engineered anterior cruciate ligament replacements are limited by a lack of methods to derive mature ligament cells. Viral overexpression of the tendon/ligament marker scleraxis (Scx) can drive cell differentiation; however, the use of viral vectors hampers translation to clinical use. In this study, C3H10T1/2 cells were transiently transfected with expression vectors containing the full-length murine Scx cDNA and cultured in three-dimensional collagen hydrogels under static or cyclic strain for up to 14 days. β-galactosidase (LacZ) transfected cells served as controls. Cell morphology and gene expression for ligament-related genes, in addition to contraction (hydrogel width), mechanical properties, and glycosaminoglycan (GAG) and DNA content of hydrogels, were quantified and compared over time, between Scx and LacZ groups, and between static and cyclically strained constructs. Increased Scx expression was maintained for the entire 14-day study in both static and cyclically strained constructs. In static culture, overexpression of Scx resulted in greater cell elongation and construct contraction compared to LacZ controls. There were no differences in gene expression, DNA, or GAG content between Scx and LacZ constructs cultured under static conditions and no differences in DNA content between Scx and LacZ constructs. When exposed to cyclic strain, Scx-overexpressing cells maintained the elongated phenotype exhibited in static constructs, increased GAG production compared to static culture, and increased expression of the ligament-related genes collagen type I, decorin, and tenascin-C compared to strained LacZ controls. Cyclically strained constructs containing Scx-overexpressing cells had increased maximum load and stiffness compared to LacZ controls. The maintenance of increased Scx expression throughout the 14 day study and subsequent increases in ligament marker gene expression and mechanical properties with cyclic, but not static strain, suggest that transient transfection may be a viable alternative to viral transduction of Scx for ligament engineering studies and support a synergistic effect of Scx and mechanical strain on driving early ligament cell differentiation.
Collapse
Affiliation(s)
- Anne E C Nichols
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine , Virginia Tech, Blacksburg, Virginia
| | - Stephen R Werre
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine , Virginia Tech, Blacksburg, Virginia
| | - Linda A Dahlgren
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine , Virginia Tech, Blacksburg, Virginia
| |
Collapse
|
52
|
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
|
53
|
Pöschke A, Krähling B, Failing K, Staszyk C. Molecular Characteristics of the Equine Periodontal Ligament. Front Vet Sci 2018; 4:235. [PMID: 29376061 PMCID: PMC5768624 DOI: 10.3389/fvets.2017.00235] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 12/14/2017] [Indexed: 02/04/2023] Open
Abstract
The equine periodontal ligament (PDL) is a fibrous connective tissue that covers the intra-alveolar parts of the tooth and anchors it to the alveolar bone-it, therefore, provides a similar function to a tendinous structure. While several studies have considered the formation and structure of tendons, there is insufficient information particularly on the molecular composition of the PDL. Especially for the equine PDL, there is limited knowledge concerning the expression of genes commonly regarded as typical for tendon tissue. In this study, the gene expression of, e.g., collagen type 1 alpha 1 (COL1), collagen type 3 alpha 1 (COL3), scleraxis (SCX), and fibrocartilage markers was examined in the functional mature equine PDL compared with immature and mature equine tendon tissue. PDL samples were obtained from incisor, premolar, and molar teeth from seven adult horses. Additionally, tendon samples were collected from four adult horses and five foals at different sampling locations. Analyses of gene expression were performed using real-time quantitative polymerase chain reaction (qRT-PCR). Significantly higher expression levels of COL1 and 3 were found in the mature equine PDL in comparison with mature tendon, indicating higher rates of collagen production and turnover in the mature equine PDL. The expression levels of SCX, a specific marker for tenogenic-differentiated cells, were on a similar level in functional mature PDL and in mature tendon tissue. Evidence of chondrogenic metaplasia, often found in tendon entheses or in pressurized regions of tendons, was not found in the mature equine PDL. The obtained results justify further experiments focused on the possible use of equine PDL cells for cell-based regenerative therapies.
Collapse
Affiliation(s)
- Antje Pöschke
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Bastian Krähling
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Klaus Failing
- Department of Biomathematics, Justus Liebig University Giessen, Giessen, Germany
| | - Carsten Staszyk
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
54
|
Chen J, Zhang W, Backman LJ, Kelk P, Danielson P. Mechanical stress potentiates the differentiation of periodontal ligament stem cells into keratocytes. Br J Ophthalmol 2018; 102:562-569. [PMID: 29306866 PMCID: PMC5890647 DOI: 10.1136/bjophthalmol-2017-311150] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/24/2017] [Accepted: 12/09/2017] [Indexed: 12/13/2022]
Abstract
Aims To explore the role of corneal-shaped static mechanical strain on the differentiation of human periodontal ligament stem cells (PDLSCs) into keratocytes and the possible synergistic effects of mechanics and inducing medium. Methods PDLSCs were exposed to 3% static dome-shaped mechanical strain in a Flexcell Tension System for 3 days and 7 days. Keratocyte phenotype was determined by gene expression of keratocyte markers. Keratocyte differentiation (inducing) medium was introduced in the Flexcell system, either continuously or intermittently combined with mechanical stimulation. The synergistic effects of mechanics and inducing medium on keratocyte differentiation was evaluated by gene and protein expression of keratocyte markers. Finally, a multilamellar cell sheet was assembled by seeding PDLSCs on a collagen membrane and inducing keratocyte differentiation. The transparency of the cell sheet was assessed, and typical markers of native human corneal stroma were evaluated by immunofluorescence staining. Results Dome-shaped mechanical stimulation promoted PDLSCs to differentiate into keratocytes, as shown by the upregulation of ALDH3A1, CD34, LUM, COL I and COL V. The expression of integrins were also upregulated after mechanical stimulation, including integrin alpha 1, alpha 2, beta 1 and non-muscle myosin II B. A synergistic effect of mechanics and inducing medium was found on keratocyte differentiation. The cell sheets were assembled under the treatment of mechanics and inducing medium simultaneously. The cell sheets were transparent, multilamellar and expressed typical markers of corneal stroma. Conclusion Dome-shaped mechanical stimulation promotes differentiation of PDLSCs into keratocytes and has synergistic effects with inducing medium. Multilamellar cell sheets that resemble native human corneal stroma show potential for future clinical applications.
Collapse
Affiliation(s)
- Jialin Chen
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Wei Zhang
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Peyman Kelk
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Patrik Danielson
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Unit of Ophthalmology, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| |
Collapse
|
55
|
The chemokines CXCL12 and CXCL14 differentially regulate connective tissue markers during limb development. Sci Rep 2017; 7:17279. [PMID: 29222527 PMCID: PMC5722906 DOI: 10.1038/s41598-017-17490-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 11/27/2017] [Indexed: 12/27/2022] Open
Abstract
Connective tissues (CT) support and connect organs together. Understanding the formation of CT is important, as CT deregulation leads to fibrosis. The identification of CT specific markers has contributed to a better understanding of CT function during development. In developing limbs, Osr1 transcription factor is involved in the differentiation of irregular CT while the transcription factor Scx labels tendon. In this study, we show that the CXCL12 and CXCL14 chemokines display distinct expression pattern in limb CT during chick development. CXCL12 positively regulates the expression of OSR1 and COL3A1, a collagen subtype of irregular CT, while CXCL14 activates the expression of the tendon marker SCX. We provide evidence that the CXCL12 effect on irregular CT involves CXCR4 receptor and vessels. In addition, the expression of CXCL12, CXCL14 and OSR genes is suppressed by the anti-fibrotic BMP signal. Finally, mechanical forces, known to be involved in adult fibrosis, control the expression of chemokines, CT-associated transcription factors and collagens during limb development. Such unexpected roles of CXCL12 and CXCL14 chemokines during CT differentiation can contribute to a better understanding of the fibrosis mechanisms in adult pathological conditions.
Collapse
|
56
|
Barboni B, Russo V, Gatta V, Bernabò N, Berardinelli P, Mauro A, Martelli A, Valbonetti L, Muttini A, Di Giacinto O, Turriani M, Silini A, Calabrese G, Abate M, Parolini O, Stuppia L, Mattioli M. Therapeutic potential of hAECs for early Achilles tendon defect repair through regeneration. J Tissue Eng Regen Med 2017; 12:e1594-e1608. [PMID: 29024514 DOI: 10.1002/term.2584] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/26/2022]
Abstract
Cell-based therapy holds great promise for tendon disorders, a widespread debilitating musculoskeletal condition. Even if the cell line remains to be defined, preliminary evidences have proven that amniotic-derived cells possess in vitro and in vivo a great tenogenic potential. This study investigated the efficacy of transplanted human amniotic epithelial cells (hAECs) by testing their early regenerative properties and mechanisms involved on a validated ovine Achilles tendon partial defect performed on 29 animals. The injured tendons treated with hAECs recovered rapidly, in 28 days, structural and biomechanical properties undertaking a programmed tissue regeneration, differently from the spontaneous healing tissues. hAECs remained viable within the host tendons establishing with the endogenous progenitor cells an active dialogue. Through the secretion of modulatory factors, hAECs inhibited the inflammatory cells infiltration, activated the M2 macrophage subpopulation early recruitment, and accelerated blood vessel as well as extracellular matrix remodelling. In parallel, some in situ differentiated hAECs displayed a tenocytelike phenotype. Both paracrine and direct hAECs stimulatory effects were confirmed analysing their genome profile before and after transplantation. The 49 human up-regulated transcripts recorded in transplanted hAECs belonged to tendon lineage differentiation (epithelial-mesenchymal transition, connective specific matrix components, and skeleton or muscle system development-related transcripts), as well as the in situ activation of paracrine signalling involved in inflammatory and immunomodulatory response. Altogether, these evidences support the hypothesis that hAECs are a practicable and efficient strategy for the acute treatment of tendinopathy, reinforcing the idea of a concrete use of amniotic epithelial cells towards the clinical practice.
Collapse
Affiliation(s)
- Barbara Barboni
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Valentina Gatta
- Medical Genetics, University "G. d'Annunzio" of Chieti Pescara, Chieti, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Paolo Berardinelli
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Alessandra Martelli
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Luca Valbonetti
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Aurelio Muttini
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Oriana Di Giacinto
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Maura Turriani
- Faculty of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Antonietta Silini
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza-Istituto Ospedaliero, Brescia, Italy
| | - Giuseppe Calabrese
- Medical Genetics, University "G. d'Annunzio" of Chieti Pescara, Chieti, Italy
| | - Michele Abate
- Department of Medicine and Science of Aging, University "G. d'Annunzio" Chieti Pescara, Chieti, Italy
| | - Ornella Parolini
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza-Istituto Ospedaliero, Brescia, Italy
| | - Liborio Stuppia
- Medical Genetics, University "G. d'Annunzio" of Chieti Pescara, Chieti, Italy
| | - Mauro Mattioli
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| |
Collapse
|
57
|
Chen J, Zhang E, Zhang W, Liu Z, Lu P, Zhu T, Yin Z, Backman LJ, Liu H, Chen X, Ouyang H. Fos Promotes Early Stage Teno-Lineage Differentiation of Tendon Stem/Progenitor Cells in Tendon. Stem Cells Transl Med 2017; 6:2009-2019. [PMID: 29024580 PMCID: PMC6430064 DOI: 10.1002/sctm.15-0146] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/19/2017] [Indexed: 12/31/2022] Open
Abstract
Stem cells have been widely used in tendon tissue engineering. The lack of refined and controlled differentiation strategy hampers the tendon repair and regeneration. This study aimed to find new effective differentiation factors for stepwise tenogenic differentiation. By microarray screening, the transcript factor Fos was found to be expressed in significantly higher amounts in postnatal Achilles tendon tissue derived from 1 day as compared with 7‐days‐old rats. It was further confirmed that expression of Fos decreased with time in postnatal rat Achilles tendon, which was accompanied with the decreased expression of multiply tendon markers. The expression of Fos also declined during regular in vitro cell culture, which corresponded to the loss of tendon phenotype. In a cell‐sheet and a three‐dimensional cell culture model, the expression of Fos was upregulated as compared with in regular cell culture, together with the recovery of tendon phenotype. In addition, significant higher expression of tendon markers was found in Fos‐overexpressed tendon stem/progenitor cells (TSPCs), and Fos knock‐down gave opposite results. In situ rat tendon repair experiments found more normal tendon‐like tissue formed and higher tendon markers expression at 4 weeks postimplantation of Fos‐overexpressed TSPCs derived nonscaffold engineering tendon (cell‐sheet), as compared with the control group. This study identifies Fos as a new marker and functional driver in the early stage teno‐lineage differentiation of tendon, which paves the way for effective stepwise tendon differentiation and future tendon regeneration. Stem Cells Translational Medicine2017;6:2009–2019
Collapse
Affiliation(s)
- Jialin Chen
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China.,Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
| | - Erchen Zhang
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Wei Zhang
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Zeyu Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Ping Lu
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Ting Zhu
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Department of Orthopedics, Second Affiliated Hospital, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Zi Yin
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
| | - Huanhuan Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Xiao Chen
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Hongwei Ouyang
- Center for Stem Cell and Tissue Engineering, School of Medicine.,Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
58
|
Gonçalves AI, Gershovich PM, Rodrigues MT, Reis RL, Gomes ME. Human adipose tissue-derived tenomodulin positive subpopulation of stem cells: A promising source of tendon progenitor cells. J Tissue Eng Regen Med 2017; 12:762-774. [PMID: 28593712 DOI: 10.1002/term.2495] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/14/2017] [Accepted: 06/03/2017] [Indexed: 01/05/2023]
Abstract
Cell-based therapies are of particular interest for tendon and ligament regeneration given the low regenerative potential of these tissues. Adipose tissue is an abundant source of stem cells, which may be employed for the healing of tendon lesions. However, human adult multipotent adipose-derived stem cells (hASCs) isolated from the stromal vascular fraction of adipose tissue originate highly heterogeneous cell populations that hinder their use in specific tissue-oriented applications. In this study, distinct subpopulations of hASCs were immunomagnetic separated and their tenogenic differentiation capacity evaluated in the presence of several growth factors (GFs), namely endothelial GF, basic-fibroblast GF, transforming GF-β1 and platelet-derived GF-BB, which are well-known regulators of tendon development, growth and healing. Among the screened hASCs subpopulations, tenomodulin-positive cells were shown to be more promising for tenogenic applications and therefore this subpopulation was further studied, assessing tendon-related markers (scleraxis, tenomodulin, tenascin C and decorin) both at gene and protein level. Additionally, the ability for depositing collagen type I and III forming extracellular matrix structures were weekly assessed up to 28 days. The results obtained indicated that tenomodulin-positive cells exhibit phenotypical features of tendon progenitor cells and can be biochemically induced towards tenogenic lineage, demonstrating that this subset of hASCs can provide a reliable source of progenitor cells for therapies targeting tendon regeneration.
Collapse
Affiliation(s)
- A I Gonçalves
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - P M Gershovich
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - M T Rodrigues
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - R L Reis
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - M E Gomes
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| |
Collapse
|
59
|
Asahara H, Inui M, Lotz MK. Tendons and Ligaments: Connecting Developmental Biology to Musculoskeletal Disease Pathogenesis. J Bone Miner Res 2017; 32:1773-1782. [PMID: 28621492 PMCID: PMC5585011 DOI: 10.1002/jbmr.3199] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/08/2017] [Accepted: 06/14/2017] [Indexed: 01/09/2023]
Abstract
Tendons and ligaments provide connections between muscle and bone or bone and bone to enable locomotion. Damage to tendons and ligaments caused by acute or chronic injury or associated with aging and arthritis is a prevalent cause of disability. Improvements in approaches for the treatment of these conditions depend on a better understanding of tendon and ligament development, cell biology, and pathophysiology. This review focuses on recent advances in the discovery of transcription factors that control ligament and tendon cell differentiation, how cell and extracellular matrix homeostasis are altered in disease, and how this new insight can lead to novel therapeutic approaches. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hiroshi Asahara
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Department of Systems BioMedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masafumi Inui
- Laboratory of Animal Regeneration Systemology, Department of Life Science, School of Agriculture, Meiji University, Kanagawa, 214-8571
| | - Martin K. Lotz
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
60
|
Islam A, Mbimba T, Younesi M, Akkus O. Effects of substrate stiffness on the tenoinduction of human mesenchymal stem cells. Acta Biomater 2017; 58:244-253. [PMID: 28602855 DOI: 10.1016/j.actbio.2017.05.058] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/02/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022]
Abstract
Extracellular matrix modulus plays an important role in regulating cell morphology, proliferation and differentiation during regular and diseased states. Although the effects of substrate topography and modulus on MSC differentiation are well known with respect to osteogenesis and adipogenesis, there has been relatively little investigation on the effects of this phenomenon on tenogenesis. Furthermore, relative roles of topographical factors (matrix alignment vs. matrix modulus) in inducing tenogenic differentiation is not well understood. In this study we investigated the effects of modulus and topographical alignment of type I collagen substrate on tendon differentiation. Type I collagen sheet substrates with random topographical alignment were fabricated with their moduli tuned in the range of 0.1, 1, 10 and 100MPa by using electrocompaction and controlled crosslinking. In one of the groups, topographical alignment was introduced at 10MPa stiffness, by controlled unidirectional stretching of the sheet. RT-PCR, immunohistochemistry and immunofluorescence results showed that mimicking the tendon topography, i.e. increasing the substrate modulus as well as alignment increased the tenogenic differentiation. Higher substrate modulus increased the expression of COLI, COLIII, COMP and TSP-4 about 2-3-fold and increased the production of COLI, COLIII and TSP-4 about 2-4-fold. Substrate alignment up regulated COLIII and COMP expression by 2-fold. Therefore, the tenoinductive collagen material model developed in this study can be used in the research and development of tissue engineering tendon repair constructs in future. STATEMENT OF SIGNIFICANCE Although the effects of substrate topography and modulus on MSC differentiation are well known with respect to osteogenesis and adipogenesis, there has been relatively little investigation on the effects of this phenomenon on tenogenesis. Furthermore, a relative role of topographical factors (matrix alignment vs. matrix modulus) in inducing tenogenic differentiation is not well understood. We investigated the effects of modulus and topographical alignment of type I collagen substrate on tendon differentiation. This study showed mimicking the tendon topography, i.e. increasing the substrate modulus as well as alignment increased the tenogenic differentiation. Therefore, the tenoinductive collagen material model developed in this study can be used in the research and development of tissue engineering tendon repair constructs in future.
Collapse
|
61
|
Wnt/β-catenin signaling suppresses expressions of Scx, Mkx, and Tnmd in tendon-derived cells. PLoS One 2017; 12:e0182051. [PMID: 28750046 PMCID: PMC5531628 DOI: 10.1371/journal.pone.0182051] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023] Open
Abstract
After tendon injuries, biomechanical properties of the injured tendon are not fully recovered in most cases. Modulation of signaling pathways, which are involved in tendon development and tendon repair, is one of attractive modalities to facilitate proper regeneration of the injured tendon. The roles of TGF-β signaling in tendon homeostasis and tendon development have been elucidated. In contrast, the roles of Wnt/β-catenin signaling in tendon remain mostly elusive. We found that the number of β-catenin-positive cells was increased at the injured site, suggesting involvement of Wnt/β-catenin signaling in tendon healing. Activation of Wnt/β-catenin signaling suppressed expressions of tenogenic genes of Scx, Mkx, and Tnmd in rat tendon-derived cells (TDCs) isolated from the Achilles tendons of 6-week old rats. Additionally, activation of Wnt/β-catenin reduced the amounts of Smad2 and Smad3, which are intracellular mediators for TGF-β signaling, and antagonized upregulation of Scx induced by TGF-β signaling in TDCs. We found that Wnt/β-catenin decreased Mkx and Tnmd expressions without suppressing Scx expression in Scx-programmed tendon progenitors. Our studies suggest that Wnt/β-catenin signaling is a repressor for tenogenic gene expressions.
Collapse
|
62
|
Dale TP, Mazher S, Webb WR, Zhou J, Maffulli N, Chen GQ, El Haj AJ, Forsyth NR. Tenogenic Differentiation of Human Embryonic Stem Cells. Tissue Eng Part A 2017; 24:361-368. [PMID: 28548630 DOI: 10.1089/ten.tea.2017.0017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tendon healing is complex to manage because of the limited regeneration capacity of tendon tissue; stem cell-based tissue engineering approaches may provide alternative healing strategies. We sought to determine whether human embryonic stem cells (hESC) could be induced to differentiate into tendon-like cells by the addition of exogenous bone morphogenetic protein (BMP)12 (growth differentiation factor[GDF]7) and BMP13 (GDF6). hESC (SHEF-1) were maintained with or without BMP12/13 supplementation, or supplemented with BMP12/13 and the Smad signaling cascade blocking agent, dorsomorphin. Primary rat tenocytes were included as a positive control in immunocytochemistry analysis. A tenocyte-like elongated morphology was observed in hESC after 40-days continuous supplementation with BMP12/13 and ascorbic acid (AA). These cells displayed a tenomodulin expression pattern and morphology consistent with that of the primary tenocyte control. Analysis of tendon-linked gene transcription in BMP12/13 supplemented hESC demonstrated consistent expression of COL1A2, COL3A1, DCN, TNC, THBS4, and TNMD levels. Conversely, when hESCs were cultured in the presence of BMP12/13 and dorsomorphin COL3A1, DCN, and TNC gene expression and tendon matrix formation were inhibited. Taken together, we have demonstrated that hESCs are responsive to tenogenic induction via BMP12/13 in the presence of AA. The directed in vitro generation of tenocytes from pluripotent stem cells may facilitate the development of novel repair approaches for this difficult to heal tissue.
Collapse
Affiliation(s)
- Tina P Dale
- 1 Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Faculty of Medicine and Health Sciences, Keele University , Thornburrow Drive, Stoke-on-Trent, Staffordshire, United Kingdom
| | - Shazia Mazher
- 1 Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Faculty of Medicine and Health Sciences, Keele University , Thornburrow Drive, Stoke-on-Trent, Staffordshire, United Kingdom
| | - William R Webb
- 1 Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Faculty of Medicine and Health Sciences, Keele University , Thornburrow Drive, Stoke-on-Trent, Staffordshire, United Kingdom
| | - Jing Zhou
- 2 School of Life Science, Tsinghua University , Beijing, China
| | - Nicola Maffulli
- 3 Centre for Sport and Exercise Medicine, Queen Mary University of London , United Kingdom
| | - Guo-Qiang Chen
- 2 School of Life Science, Tsinghua University , Beijing, China
| | - Alicia J El Haj
- 1 Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Faculty of Medicine and Health Sciences, Keele University , Thornburrow Drive, Stoke-on-Trent, Staffordshire, United Kingdom
| | - Nicholas R Forsyth
- 1 Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Faculty of Medicine and Health Sciences, Keele University , Thornburrow Drive, Stoke-on-Trent, Staffordshire, United Kingdom
| |
Collapse
|
63
|
Valdivia M, Vega-Macaya F, Olguín P. Mechanical Control of Myotendinous Junction Formation and Tendon Differentiation during Development. Front Cell Dev Biol 2017; 5:26. [PMID: 28386542 PMCID: PMC5362613 DOI: 10.3389/fcell.2017.00026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/07/2017] [Indexed: 01/01/2023] Open
Abstract
The development of the musculoskeletal system is a great model to study the interplay between chemical and mechanical inter-tissue signaling in cell adhesion, tissue morphogenesis and differentiation. In both vertebrates and invertebrates (e.g., Drosophila melanogaster) the formation of muscle-tendon interaction generates mechanical forces which are required for myotendinous junction maturation and tissue differentiation. In addition, these forces must be withstood by muscles and tendons in order to prevent detachment from each other, deformation or even losing their integrity. Extracellular matrix remodeling at the myotendinous junction is key to resist mechanical load generated by muscle contraction. Recent evidences in vertebrates indicate that mechanical forces generated during junction formation regulate chemical signaling leading to extracellular matrix remodeling, however, the mechanotransduction mechanisms associated to this response remains elusive. In addition to extracellular matrix remodeling, the ability of Drosophila tendon-cells to bear mechanical load depends on rearrangement of tendon cell cytoskeleton, thus studying the molecular mechanisms involved in this process is critical to understand the contribution of mechanical forces to the development of the musculoskeletal system. Here, we review recent findings regarding the role of chemical and mechanical signaling in myotendinous junction formation and tendon differentiation, and discuss molecular mechanisms of mechanotransduction that may allow tendon cells to withstand mechanical load during development of the musculoskeletal system.
Collapse
Affiliation(s)
- Mauricio Valdivia
- Program in Human Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Biomedical Neurosciences Institute, University of Chile Santiago, Chile
| | - Franco Vega-Macaya
- Program in Human Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Biomedical Neurosciences Institute, University of Chile Santiago, Chile
| | - Patricio Olguín
- Program in Human Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Biomedical Neurosciences Institute, University of Chile Santiago, Chile
| |
Collapse
|
64
|
Grier W, Moy A, Harley B. Cyclic tensile strain enhances human mesenchymal stem cell Smad 2/3 activation and tenogenic differentiation in anisotropic collagen-glycosaminoglycan scaffolds. Eur Cell Mater 2017; 33:227-239. [PMID: 28319248 PMCID: PMC5453510 DOI: 10.22203/ecm.v033a14] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Stem cell research arose from the need to explore new therapeutic possibilities for intractable and lethal diseases. Although musculoskeletal disorders are basically nonlethal, their high prevalence and relative ease of performing clinical trials have facilitated the clinical application of stem cells in this field. However, few reliable clinical studies have been published, despite the plethora of in vitro and preclinical studies in stem cell research for regenerative medicine in the musculoskeletal system. Stem cell therapy can be applied locally for bone, cartilage and tendon regeneration. Candidate disease modalities in bone regeneration include large bone defects, nonunion of fractures, and osteonecrosis. Focal osteochondral defect and osteoarthritis are current targets for cartilage regeneration. For tendon regeneration, bone-tendon junction problems such as rotator cuff tears are hot topics in clinical research. To date, the literature supporting stem cell-based therapies comprises mostly case reports or case series. Therefore, high-quality evidence, including from randomised clinical trials, is necessary to define the role of cell-based therapies in the treatment of musculoskeletal disorders. It is imperative that clinicians who adopt stem cell treatment into their practices possess a good understanding of the natural course of the disease. It is also highly recommended that treating physicians do not thrust aside the concomitant use of established measures until stem cell therapy is evidently proved worthy in terms of efficacy and cost. The purpose of this review is to summarise on the current status of stem cell application in the orthopaedic field along with the author's view of future prospects.
Collapse
Affiliation(s)
- W.K. Grier
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - A.S. Moy
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - B.A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA,Address for correspondence: B.A.C. Harley, Department of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL 61801, USA, Telephone number: +1 2172447112, Fax number: +1 2173335052,
| |
Collapse
|
65
|
Bavin EP, Atkinson F, Barsby T, Guest DJ. Scleraxis Is Essential for Tendon Differentiation by Equine Embryonic Stem Cells and in Equine Fetal Tenocytes. Stem Cells Dev 2017; 26:441-450. [DOI: 10.1089/scd.2016.0279] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Emma P. Bavin
- Centre for Preventive Medicine, Animal Health Trust, Newmarket, United Kingdom
| | - Francesca Atkinson
- Centre for Preventive Medicine, Animal Health Trust, Newmarket, United Kingdom
| | - Tom Barsby
- Centre for Preventive Medicine, Animal Health Trust, Newmarket, United Kingdom
| | - Debbie J. Guest
- Centre for Preventive Medicine, Animal Health Trust, Newmarket, United Kingdom
| |
Collapse
|
66
|
Hu JJ, Yin Z, Shen WL, Xie YB, Zhu T, Lu P, Cai YZ, Kong MJ, Heng BC, Zhou YT, Chen WS, Chen X, Ouyang HW. Pharmacological Regulation of In Situ Tissue Stem Cells Differentiation for Soft Tissue Calcification Treatment. Stem Cells 2017; 34:1083-96. [PMID: 26851078 DOI: 10.1002/stem.2306] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/25/2015] [Accepted: 11/29/2015] [Indexed: 01/24/2023]
Abstract
Calcification of soft tissues, such as heart valves and tendons, is a common clinical problem with limited therapeutics. Tissue specific stem/progenitor cells proliferate to repopulate injured tissues. But some of them become divergent to the direction of ossification in the local pathological microenvironment, thereby representing a cellular target for pharmacological approach. We observed that HIF-2alpha (encoded by EPAS1 inclined form) signaling is markedly activated within stem/progenitor cells recruited at calcified sites of diseased human tendons and heart valves. Proinflammatory microenvironment, rather than hypoxia, is correlated with HIF-2alpha activation and promoted osteochondrogenic differentiation of tendon stem/progenitor cells (TSPCs). Abnormal upregulation of HIF-2alpha served as a key switch to direct TSPCs differentiation into osteochondral-lineage rather than teno-lineage. Notably, Scleraxis (Scx), an essential tendon specific transcription factor, was suppressed on constitutive activation of HIF-2alpha and mediated the effect of HIF-2alpha on TSPCs fate decision. Moreover, pharmacological inhibition of HIF-2alpha with digoxin, which is a widely utilized drug, can efficiently inhibit calcification and enhance tenogenesis in vitro and in the Achilles's tendinopathy model. Taken together, these findings reveal the significant role of the tissue stem/progenitor cells fate decision and suggest that pharmacological regulation of HIF-2alpha function is a promising approach for soft tissue calcification treatment.
Collapse
Affiliation(s)
- Jia-Jie Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Zhejiang, 310009, China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Zhejiang, 310009, China
| | - Wei-Liang Shen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital , School of Medicine Zhejiang University, Zhejiang, 310009, China
| | - Yu-Bin Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Zhejiang, 310009, China
| | - Ting Zhu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Zhejiang, 310009, China
| | - Ping Lu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Zhejiang, 310009, China
| | - You-Zhi Cai
- Department of Orthopedic Surgery, 1st Affiliated Hospital, School of Medicine Zhejiang University, Zhejiang, 310009, China
| | - Min-Jian Kong
- Department of Orthopedic Surgery, 2nd Affiliated Hospital , School of Medicine Zhejiang University, Zhejiang, 310009, China
| | - Boon Chin Heng
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Yi-Ting Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Department of Biochemistry and Molecular Biology, Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Wei-Shan Chen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital , School of Medicine Zhejiang University, Zhejiang, 310009, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Zhejiang, 310009, China
| | - Hong-Wei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Zhejiang, 310009, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310000, China.,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, 310003, Hangzhou, China.,China Orthopedic Regenerative Medicine Group (CORMed)
| |
Collapse
|
67
|
Chen YY, He ST, Yan FH, Zhou PF, Luo K, Zhang YD, Xiao Y, Lin MK. Dental pulp stem cells express tendon markers under mechanical loading and are a potential cell source for tissue engineering of tendon-like tissue. Int J Oral Sci 2016; 8:213-222. [PMID: 27811845 PMCID: PMC5168414 DOI: 10.1038/ijos.2016.33] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2016] [Indexed: 12/29/2022] Open
Abstract
Postnatal mesenchymal stem cells have the capacity to differentiate into multiple cell lineages. This study explored the possibility of dental pulp stem cells (DPSCs) for potential application in tendon tissue engineering. The expression of tendon-related markers such as scleraxis, tenascin-C, tenomodulin, eye absent homologue 2, collagens I and VI was detected in dental pulp tissue. Interestingly, under mechanical stimulation, these tendon-related markers were significantly enhanced when DPSCs were seeded in aligned polyglycolic acid (PGA) fibre scaffolds. Furthermore, mature tendon-like tissue was formed after transplantation of DPSC-PGA constructs under mechanical loading conditions in a mouse model. This study demonstrates that DPSCs could be a potential stem cell source for tissue engineering of tendon-like tissue.
Collapse
Affiliation(s)
- Yu-Ying Chen
- Department of Stomatology, Fujian Provincial Hospital, Fuzhou, China.,School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Sheng-Teng He
- Department of Stomatology, Hainan Province Nongken Sanya Hospital, Sanya, China
| | - Fu-Hua Yan
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Australia
| | - Peng-Fei Zhou
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Kai Luo
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yan-Ding Zhang
- College of Life Science, Fujian Normal University, Fuzhou, China
| | - Yin Xiao
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Min-Kui Lin
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
68
|
Islam A, Younesi M, Mbimba T, Akkus O. Collagen Substrate Stiffness Anisotropy Affects Cellular Elongation, Nuclear Shape, and Stem Cell Fate toward Anisotropic Tissue Lineage. Adv Healthc Mater 2016; 5:2237-47. [PMID: 27377355 PMCID: PMC5203936 DOI: 10.1002/adhm.201600284] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/03/2016] [Indexed: 01/01/2023]
Abstract
Rigidity of substrates plays an important role in stem cell fate. Studies are commonly carried out on isotropically stiff substrate or substrates with unidirectional stiffness gradients. However, many native tissues are anisotropically stiff and it is unknown whether controlled presentation of stiff and compliant material axes on the same substrate governs cytoskeletal and nuclear morphology, as well as stem cell differentiation. In this study, electrocompacted collagen sheets are stretched to varying degrees to tune the stiffness anisotropy (SA) in the range of 1 to 8, resulting in stiff and compliant material axes orthogonal to each other. The cytoskeletal aspect ratio increased with increasing SA by about fourfold. Such elongation was absent on cellulose acetate replicas of aligned collagen surfaces indicating that the elongation was not driven by surface topography. Mesenchymal stem cells (MSCs) seeded on varying anisotropy sheets displayed a dose-dependent upregulation of tendon-related markers such as Mohawk and Scleraxis. After 21 d of culture, highly anisotropic sheets induced greater levels of production of type-I, type-III collagen, and thrombospondin-4. Therefore, SA has direct effects on MSC differentiation. These findings may also have ramifications of stem cell fate on other anisotropically stiff tissues, such as skeletal/cardiac muscles, ligaments, and bone.
Collapse
Affiliation(s)
- Anowarul Islam
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mousa Younesi
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Thomas Mbimba
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ozan Akkus
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Orthopaedics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
69
|
Yin Z, Guo J, Wu TY, Chen X, Xu LL, Lin SE, Sun YX, Chan KM, Ouyang H, Li G. Stepwise Differentiation of Mesenchymal Stem Cells Augments Tendon-Like Tissue Formation and Defect Repair In Vivo. Stem Cells Transl Med 2016; 5:1106-1116. [PMID: 27280798 PMCID: PMC4954446 DOI: 10.5966/sctm.2015-0215] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 03/07/2016] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED : Tendon injuries are common and present a clinical challenge, as they often respond poorly to treatment and result in long-term functional impairment. Inferior tendon healing responses are mainly attributed to insufficient or failed tenogenesis. The main objective of this study was to establish an efficient approach to induce tenogenesis of bone marrow-derived mesenchymal stem cells (BMSCs), which are the most common seed cells in tendon tissue engineering. First, representative reported tenogenic growth factors were used as media supplementation to induce BMSC differentiation, and the expression of teno-lineage transcription factors and matrix proteins was compared. We found that transforming growth factor (TGF)-β1 significantly induced teno-lineage-specific gene scleraxis expression and collagen production. TGF-β1 combined with connective tissue growth factor (CTGF) elevated tenomodulin and Egr1 expression at day 7. Hence, a stepwise tenogenic differentiation approach was established by first using TGF-β1 stimulation, followed by combination with CTGF for another 7 days. Gene expression analysis showed that this stepwise protocol initiated and maintained highly efficient tenogenesis of BMSCs. Finally, regarding in situ rat patellar tendon repair, tendons treated with induced tenogenic BMSCs had better structural and mechanical properties than those of the control group, as evidenced by histological scoring, collagen I and tenomodulin immunohistochemical staining, and tendon mechanical testing. Collectively, these findings demonstrate a reliable and practical strategy of inducing tenogenesis of BMSCs for tendon regeneration and may enhance the effectiveness of cell therapy in treating tendon disorders. SIGNIFICANCE The present study investigated the efficiency of representative tenogenic factors on mesenchymal stem cells' tenogenic differentiation and established an optimized stepwise tenogenic differentiation approach to commit tendon lineage differentiation for functional tissue regeneration. The reliable tenogenic differentiation approach for stem cells not only serves as a platform for further studies of underlying molecular mechanisms but also can be used to enhance cell therapy outcome in treating tendon disorders and develop novel therapeutics for tendon injury.
Collapse
Affiliation(s)
- Zi Yin
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China China Orthopedic Regenerative Medicine Group, Hangzhou, People's Republic of China
| | - Jia Guo
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China
| | - Tian-Yi Wu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China
| | - Xiao Chen
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China China Orthopedic Regenerative Medicine Group, Hangzhou, People's Republic of China
| | - Liang-Liang Xu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China
| | - Si-En Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China
| | - Yun-Xin Sun
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China
| | - Kai-Ming Chan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China
| | - Hongwei Ouyang
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China China Orthopedic Regenerative Medicine Group, Hangzhou, People's Republic of China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China China Orthopedic Regenerative Medicine Group, Hangzhou, People's Republic of China Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 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, Hong Kong, People's Republic of China The Chinese University of Hong Kong-China Astronaut Research and Training Center Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
| |
Collapse
|
70
|
Langhans MT, Yu S, Tuan RS. Stem Cells in Skeletal Tissue Engineering: Technologies and Models. Curr Stem Cell Res Ther 2016; 11:453-474. [PMID: 26423296 DOI: 10.2174/1574888x10666151001115248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
This review surveys the use of pluripotent and multipotent stem cells in skeletal tissue engineering. Specific emphasis is focused on evaluating the function and activities of these cells in the context of development in vivo, and how technologies and methods of stem cell-based tissue engineering for stem cells must draw inspiration from developmental biology. Information on the embryonic origin and in vivo differentiation of skeletal tissues is first reviewed, to shed light on the persistence and activities of adult stem cells that remain in skeletal tissues after embryogenesis. Next, the development and differentiation of pluripotent stem cells is discussed, and some of their advantages and disadvantages in the context of tissue engineering are presented. The final section highlights current use of multipotent adult mesenchymal stem cells, reviewing their origin, differentiation capacity, and potential applications to tissue engineering.
Collapse
Affiliation(s)
| | | | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA 15219, USA.
| |
Collapse
|
71
|
Subramanian A, Schilling TF. Tendon development and musculoskeletal assembly: emerging roles for the extracellular matrix. Development 2016; 142:4191-204. [PMID: 26672092 DOI: 10.1242/dev.114777] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tendons and ligaments are extracellular matrix (ECM)-rich structures that interconnect muscles and bones. Recent work has shown how tendon fibroblasts (tenocytes) interact with muscles via the ECM to establish connectivity and strengthen attachments under tension. Similarly, ECM-dependent interactions between tenocytes and cartilage/bone ensure that tendon-bone attachments form with the appropriate strength for the force required. Recent studies have also established a close lineal relationship between tenocytes and skeletal progenitors, highlighting the fact that defects in signals modulated by the ECM can alter the balance between these fates, as occurs in calcifying tendinopathies associated with aging. The dynamic fine-tuning of tendon ECM composition and assembly thus gives rise to the remarkable characteristics of this unique tissue type. Here, we provide an overview of the functions of the ECM in tendon formation and maturation that attempts to integrate findings from developmental genetics with those of matrix biology.
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| |
Collapse
|
72
|
Gtf2ird1-Dependent Mohawk Expression Regulates Mechanosensing Properties of the Tendon. Mol Cell Biol 2016; 36:1297-309. [PMID: 26884464 PMCID: PMC4836271 DOI: 10.1128/mcb.00950-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/08/2016] [Indexed: 11/20/2022] Open
Abstract
Mechanoforces experienced by an organ are translated into biological information for cellular sensing and response. In mammals, the tendon connective tissue experiences and resists physical forces, with tendon-specific mesenchymal cells called tenocytes orchestrating extracellular matrix (ECM) turnover. We show that Mohawk (Mkx), a tendon-specific transcription factor, is essential in mechanoresponsive tenogenesis through regulation of its downstream ECM genes such as type I collagens and proteoglycans such as fibromodulin both in vivo and in vitro Wild-type (WT) mice demonstrated an increase in collagen fiber diameter and density in response to physical treadmill exercise, whereas in Mkx(-/-) mice, tendons failed to respond to the same mechanical stimulation. Furthermore, functional screening of the Mkx promoter region identified several upstream transcription factors that regulate Mkx In particular, general transcription factor II-I repeat domain-containing protein 1 (Gtf2ird1) that is expressed in the cytoplasm of unstressed tenocytes translocated into the nucleus upon mechanical stretching to activate the Mkx promoter through chromatin regulation. Here, we demonstrate that Gtf2ird1 is essential for Mkx transcription, while also linking mechanical forces to Mkx-mediated tendon homeostasis and regeneration.
Collapse
|
73
|
Retinoic acid receptor signaling preserves tendon stem cell characteristics and prevents spontaneous differentiation in vitrox. Stem Cell Res Ther 2016; 7:45. [PMID: 27001426 PMCID: PMC4802591 DOI: 10.1186/s13287-016-0306-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 01/12/2016] [Accepted: 03/04/2016] [Indexed: 02/07/2023] Open
Abstract
Background Previous studies have reported that adult mesenchymal stem cells (MSCs) tend to gradually lose their stem cell characteristics in vitro when placed outside their niche environment. They subsequently undergo spontaneous differentiation towards mesenchymal lineages after only a few passages. We observed a similar phenomenon with adult tendon stem cells (TSCs) where expression of key tendon genes such as Scleraxis (Scx), are being repressed with time in culture. We hypothesized that an environment able to restore or maintain Scleraxis expression could be of therapeutic interest for in vitro use and tendon cell-based therapies. Methods TSCs were isolated from human cadaveric Achilles tendon and expanded for 4 passages. A high content imaging assay that monitored the induction of Scx protein nuclear localization was used to screen ~1000 known drugs. Results We identified retinoic acid receptor (RAR) agonists as potent inducers of nuclear Scx in the small molecule screen. The upregulation correlated with improved maintenance of tendon stem cell properties through inhibition of spontaneous differentiation rather than the anticipated induction of tenogenic differentiation. Our results suggest that histone epigenetic modifications by RAR are driving this effect which is not likely only dependent on Scleraxis nuclear binding but also mediated through other key genes involved in stem cell self-renewal and differentiation. Furthermore, we demonstrate that the effect of RAR compounds on TSCs is reversible by revealing their multi-lineage differentiation ability upon withdrawal of the compound. Conclusion Based on these findings, RAR agonists could provide a valid approach for maintaining TSC stemness during expansion in vitro, thus improving their regenerative potential for cell-based therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0306-3) contains supplementary material, which is available to authorized users.
Collapse
|
74
|
Bagchi RA, Roche P, Aroutiounova N, Espira L, Abrenica B, Schweitzer R, Czubryt MP. The transcription factor scleraxis is a critical regulator of cardiac fibroblast phenotype. BMC Biol 2016; 14:21. [PMID: 26988708 PMCID: PMC4794909 DOI: 10.1186/s12915-016-0243-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/01/2016] [Indexed: 12/30/2022] Open
Abstract
Background Resident fibroblasts synthesize the cardiac extracellular matrix, and can undergo phenotype conversion to myofibroblasts to augment matrix production, impairing function and contributing to organ failure. A significant gap in our understanding of the transcriptional regulation of these processes exists. Given the key role of this phenotype conversion in fibrotic disease, the identification of such novel transcriptional regulators may yield new targets for therapies for fibrosis. Results Using explanted primary cardiac fibroblasts in gain- and loss-of-function studies, we found that scleraxis critically controls cardiac fibroblast/myofibroblast phenotype by direct transcriptional regulation of myriad genes that effectively define these cells, including extracellular matrix components and α-smooth muscle actin. Scleraxis furthermore potentiated the TGFβ/Smad3 signaling pathway, a key regulator of myofibroblast conversion, by facilitating transcription complex formation. While scleraxis promoted fibroblast to myofibroblast conversion, loss of scleraxis attenuated myofibroblast function and gene expression. These results were confirmed in scleraxis knockout mice, which were cardiac matrix-deficient and lost ~50 % of their complement of cardiac fibroblasts, with evidence of impaired epithelial-to-mesenchymal transition (EMT). Scleraxis directly transactivated several EMT marker genes, and was sufficient to induce mesenchymal/fibroblast phenotype conversion of A549 epithelial cells. Conversely, loss of scleraxis attenuated TGFβ-induced EMT marker expression. Conclusions Our results demonstrate that scleraxis is a novel and potent regulator of cellular progression along the continuum culminating in the cardiac myofibroblast phenotype. Scleraxis was both sufficient to drive conversion, and required for full conversion to occur. Scleraxis fulfills this role by direct transcriptional regulation of key target genes, and by facilitating TGFβ/Smad signaling. Given the key role of fibroblast to myofibroblast conversion in fibrotic diseases in the heart and other tissue types, scleraxis may be an important target for therapeutic development. Electronic supplementary material The online version of this article (doi:10.1186/s12915-016-0243-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rushita A Bagchi
- Institute of Cardiovascular Sciences, Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, University of Manitoba, R4008 St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Patricia Roche
- Institute of Cardiovascular Sciences, Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, University of Manitoba, R4008 St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Nina Aroutiounova
- Institute of Cardiovascular Sciences, Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, University of Manitoba, R4008 St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Leon Espira
- Institute of Cardiovascular Sciences, Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, University of Manitoba, R4008 St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Bernard Abrenica
- Institute of Cardiovascular Sciences, Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, University of Manitoba, R4008 St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada
| | - Ronen Schweitzer
- Shriners Hospital for Children, Research Division and Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Michael P Czubryt
- Institute of Cardiovascular Sciences, Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, University of Manitoba, R4008 St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada.
| |
Collapse
|
75
|
Chen J, Zhang W, Liu Z, Zhu T, Shen W, Ran J, Tang Q, Gong X, Backman LJ, Chen X, Chen X, Wen F, Ouyang H. Characterization and comparison of post-natal rat Achilles tendon-derived stem cells at different development stages. Sci Rep 2016; 6:22946. [PMID: 26972579 PMCID: PMC4789738 DOI: 10.1038/srep22946] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/24/2016] [Indexed: 12/21/2022] Open
Abstract
Tendon stem/progenitor cells (TSPCs) are a potential cell source for tendon tissue engineering. The striking morphological and structural changes of tendon tissue during development indicate the complexity of TSPCs at different stages. This study aims to characterize and compare post-natal rat Achilles tendon tissue and TSPCs at different stages of development. The tendon tissue showed distinct differences during development: the tissue structure became denser and more regular, the nuclei became spindle-shaped and the cell number decreased with time. TSPCs derived from 7 day Achilles tendon tissue showed the highest self-renewal ability, cell proliferation, and differentiation potential towards mesenchymal lineage, compared to TSPCs derived from 1 day and 56 day tissue. Microarray data showed up-regulation of several groups of genes in TSPCs derived from 7 day Achilles tendon tissue, which may account for the unique cell characteristics during this specific stage of development. Our results indicate that TSPCs derived from 7 day Achilles tendon tissue is a superior cell source as compared to TSPCs derived from 1 day and 56 day tissue, demonstrating the importance of choosing a suitable stem cell source for effective tendon tissue engineering and regeneration.
Collapse
Affiliation(s)
- Jialin Chen
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China.,Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå, 90187, Sweden
| | - Wei Zhang
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China
| | - Zeyu Liu
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China
| | - Ting Zhu
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China.,Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, China
| | - Weiliang Shen
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China.,Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, China
| | - Jisheng Ran
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, China
| | - Qiaomei Tang
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China.,Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, China
| | - Xiaonan Gong
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå, 90187, Sweden
| | - Xiao Chen
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| | - Xiaowen Chen
- Division of haematology and oncology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Feiqiu Wen
- Division of haematology and oncology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Hongwei Ouyang
- Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Provincial Key Lab for tissue engineering and regenerative medicine, 310000, Hangzhou, China.,Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| |
Collapse
|
76
|
Lin X, Shi Y, Cao Y, Liu W. Recent progress in stem cell differentiation directed by material and mechanical cues. ACTA ACUST UNITED AC 2016; 11:014109. [PMID: 26836059 DOI: 10.1088/1748-6041/11/1/014109] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cells play essential roles in tissue regeneration in vivo via specific lineage differentiation induced by environmental factors. In the past, biochemical signals were the focus of induced stem cell differentiation. As reported by Engler et al (2006 Cell 126 677-89), biophysical signal mediated stem cell differentiation could also serve as an important inducer. With the advancement of material science, it becomes a possible strategy to generate active biophysical signals for directing stem cell fate through specially designed material microstructures. In the past five years, significant progress has been made in this field, and these designed biophysical signals include material elasticity/rigidity, micropatterned structure, extracellular matrix (ECM) coated materials, material transmitted extracellular mechanical force etc. A large number of investigations involved material directed differentiation of mesenchymal stem cells, neural stem/progenitor cells, adipose derived stem cells, hematopoietic stem/progenitor cells, embryonic stem cells and other cells. Hydrogel based materials were commonly used to create varied mechanical properties via modifying the ratio of different components, crosslinking levels, matrix concentration and conjugation with other components. Among them, polyacrylamide (PAM) and polydimethylsiloxane (PDMS) hydrogels remained the major types of material. Specially designed micropatterning was not only able to create a unique topographical surface to control cell shape, alignment, cell-cell and cell-matrix contact for basic stem cell biology study, but also could be integrated with 3D bioprinting to generate micropattered 3D structure and thus to induce stem cell based tissue regeneration. ECM coating on a specific topographical structure was capable of inducing even more specific and potent stem cell differentiation along with soluble factors and mechanical force. The article overviews the progress of the past five years in this particular field.
Collapse
Affiliation(s)
- Xunxun Lin
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Rd, People's Republic of China. Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of China, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
77
|
Engineering Tendon: Scaffolds, Bioreactors, and Models of Regeneration. Stem Cells Int 2015; 2016:3919030. [PMID: 26839559 PMCID: PMC4709784 DOI: 10.1155/2016/3919030] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 09/20/2015] [Indexed: 12/23/2022] Open
Abstract
Tendons bridge muscle and bone, translating forces to the skeleton and increasing the safety and efficiency of locomotion. When tendons fail or degenerate, there are no effective pharmacological interventions. The lack of available options to treat damaged tendons has created a need to better understand and improve the repair process, particularly when suitable autologous donor tissue is unavailable for transplantation. Cells within tendon dynamically react to loading conditions and undergo phenotypic changes in response to mechanobiological stimuli. Tenocytes respond to ultrastructural topography and mechanical deformation via a complex set of behaviors involving force-sensitive membrane receptor activity, changes in cytoskeletal contractility, and transcriptional regulation. Effective ex vivo model systems are needed to emulate the native environment of a tissue and to translate cell-matrix forces with high fidelity. While early bioreactor designs have greatly expanded our knowledge of mechanotransduction, traditional scaffolds do not fully model the topography, composition, and mechanical properties of native tendon. Decellularized tendon is an ideal scaffold for cultivating replacement tissue and modeling tendon regeneration. Decellularized tendon scaffolds (DTS) possess high clinical relevance, faithfully translate forces to the cellular scale, and have bulk material properties that match natural tissue. This review summarizes progress in tendon tissue engineering, with a focus on DTS and bioreactor systems.
Collapse
|
78
|
Maman E, Somjen D, Maman E, Katzburg S, Sharfman ZT, Stern N, Dolkart O. The response of cells derived from the supraspinatus tendon to estrogen and calciotropic hormone stimulations: in vitro study. Connect Tissue Res 2015; 57:124-30. [PMID: 26646255 DOI: 10.3109/03008207.2015.1114615] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE The most frequent complications after rotator cuff repair (RCR) are non-healing and re-tear. Age and gender are both proven risk factors for faulty RCR. This study analyzed the effects of female sex steroids and calciotropic hormones on tendon-derived cell characteristics. METHODS Tendon-derived cells from rat supraspinatus were treated with estradiol-17β (E2); soy isoflavones (daidzein, genistein, biochainin A); raloxifene and estrogen receptors α and β agonists and antagonists; and less-calcemic vitamin-D analog, parathyroid hormone, and vehicle control for 24 h. Cell proliferation and mRNA expression of estrogen receptor α and β, vitamin-D receptor (VDR), scleraxis, and collagen-1 were assessed. RESULTS E2, Biochainin A, raloxifene, and vitamin-D significantly increased tendon-derived cell proliferation. Estrogen receptor α antagonists neutralized tendon-derived cells response to estradiol 17-β; however, estrogen receptor β antagonists did not have an effect. Scleraxis expression decreased following estradiol 17-β and vitamin-D treatments. Vitamin-D significantly reduced collagen-1 expression, while estradiol 17-β had no effect. Vitamin-D and estradiol 17-β upregulated VDR expression. CONCLUSIONS Significant tendon-derived cell proliferation can be achieved with commonly prescribed female sex and calciotropic hormones. However, collagen-1 expression remained constant or decreased following the administration of these hormones. Female sex steroids and vitamin-D promoted tendon-derived cell proliferation via estrogen receptor α and VDR, not estrogen receptor β. Amplified cell proliferation was not associated with increased scleraxis and collagen-1 expression. These results have important implications to the properties of healing tendon and possible pharmaceutical therapies for patients with torn RC. Further research is warranted to expose the underling mechanisms of these effects.
Collapse
Affiliation(s)
- Eran Maman
- a Shoulder Unit, Division of Orthopedic Surgery, Tel-Aviv Medical Center and the Sackler Faculty of Medicine , Tel-Aviv University , Tel-Aviv , Israel
| | - Dalia Somjen
- b Institute of Endocrinology, Metabolism and Hypertension, Tel-Aviv Medical Center and the Sackler Faculty of Medicine , Tel-Aviv University , Tel-Aviv , Israel
| | - Ettie Maman
- c IVF unit, Department of Obstetrics and Gynecology , Sheba Medical Center, Tel Hashomer, Tel-Aviv University , Tel-Aviv , Israel
| | - Sarah Katzburg
- b Institute of Endocrinology, Metabolism and Hypertension, Tel-Aviv Medical Center and the Sackler Faculty of Medicine , Tel-Aviv University , Tel-Aviv , Israel
| | - Zachary T Sharfman
- a Shoulder Unit, Division of Orthopedic Surgery, Tel-Aviv Medical Center and the Sackler Faculty of Medicine , Tel-Aviv University , Tel-Aviv , Israel
| | - Naftali Stern
- b Institute of Endocrinology, Metabolism and Hypertension, Tel-Aviv Medical Center and the Sackler Faculty of Medicine , Tel-Aviv University , Tel-Aviv , Israel
| | - Oleg Dolkart
- a Shoulder Unit, Division of Orthopedic Surgery, Tel-Aviv Medical Center and the Sackler Faculty of Medicine , Tel-Aviv University , Tel-Aviv , Israel
| |
Collapse
|
79
|
Liu H, Zhang C, Zhu S, Lu P, Zhu T, Gong X, Zhang Z, Hu J, Yin Z, Heng BC, Chen X, Ouyang HW. Mohawk promotes the tenogenesis of mesenchymal stem cells through activation of the TGFβ signaling pathway. Stem Cells 2015; 33:443-55. [PMID: 25332192 DOI: 10.1002/stem.1866] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 09/09/2014] [Accepted: 09/18/2014] [Indexed: 12/14/2022]
Abstract
The transcription factor Mohawk (Mkx) is expressed in developing tendons and is an important regulator of tenogenic differentiation. However, the exact roles of Mkx in tendinopathy and tendon repair remain unclear. Using gene expression Omnibus datasets and immunofluorescence assays, we found that Mkx expression level was dramatically lower in human tendinopathy tissue and it is activated at specific stages of tendon development. In mesenchymal stem cells (MSCs), ectopic Mkx expression strikingly promoted tenogenesis more efficiently than Scleraxis (Scx), a well-known master transcription factor of tendon. Significantly higher levels of tenogenic gene expression and collagen fibril growth were observed with Mkx versus Scx. Interestingly, it was observed that Mkx dramatically upregulated Scx through binding to the Tgfb2 promoter. Additionally, the transplantation of Mkx-expressing-MSC sheets promoted tendon repair in a mouse model of Achilles-tendon defect. Taken together, these data shed light on previously unrecognized roles of Mkx in tendinopathy, tenogenesis, and tendon repair as well as in regulating the TGFβ pathway.
Collapse
Affiliation(s)
- Huanhuan Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Li Y, Ramcharan M, Zhou Z, Leong DJ, Akinbiyi T, Majeska RJ, Sun HB. The Role of Scleraxis in Fate Determination of Mesenchymal Stem Cells for Tenocyte Differentiation. Sci Rep 2015; 5:13149. [PMID: 26289033 PMCID: PMC4542341 DOI: 10.1038/srep13149] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/02/2015] [Indexed: 01/02/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent cells that primarily differentiate into osteocytes, chondrocytes, and adipocytes. Recent studies indicate that MSCs can also be induced to generate tenocyte-like cells; moreover, MSCs have been suggested to have great therapeutic potential for tendon pathologies. Yet the precise molecular cascades governing tenogenic differentiation of MSCs remain unclear. We demonstrate scleraxis, a transcription factor critically involved in embryonic tendon development and formation, plays a pivotal role in the fate determination of MSC towards tenocyte differentiation. Using murine C3H10T1/2 pluripotent stem cells as a model system, we show scleraxis is extensively expressed in the early phase of bone morphogenetic protein (BMP)-12-triggered tenocytic differentiation. Once induced, scleraxis directly transactivates tendon lineage-related genes such as tenomodulin and suppresses osteogenic, chondrogenic, and adipogenic capabilities, thus committing C3H10T1/2 cells to differentiate into the specific tenocyte-like lineage, while eliminating plasticity for other lineages. We also reveal that mechanical loading-mediated tenocytic differentiation follows a similar pathway and that BMP-12 and cyclic uniaxial strain act in an additive fashion to augment the maximal response by activating signal transducer Smad8. These results provide critical insights into the determination of multipotent stem cells to the tenocyte lineage induced by both chemical and physical signals.
Collapse
Affiliation(s)
- Yonghui Li
- Leni and Peter W. May Department of Orthopedics, Mount Sinai School of Medicine, New York, NY 10029
| | - Melissa Ramcharan
- Leni and Peter W. May Department of Orthopedics, Mount Sinai School of Medicine, New York, NY 10029.,Department of Biomedical Engineering, City College of New York, New York, NY 10031
| | - Zuping Zhou
- Leni and Peter W. May Department of Orthopedics, Mount Sinai School of Medicine, New York, NY 10029
| | - Daniel J Leong
- Leni and Peter W. May Department of Orthopedics, Mount Sinai School of Medicine, New York, NY 10029.,Department of Biomedical Engineering, City College of New York, New York, NY 10031.,Department of Orthopedic Surgery, Albert Einstein College of Medicine, Bronx, NY 10461.,Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Takintope Akinbiyi
- Leni and Peter W. May Department of Orthopedics, Mount Sinai School of Medicine, New York, NY 10029
| | - Robert J Majeska
- Department of Biomedical Engineering, City College of New York, New York, NY 10031
| | - Hui B Sun
- Leni and Peter W. May Department of Orthopedics, Mount Sinai School of Medicine, New York, NY 10029.,Department of Orthopedic Surgery, Albert Einstein College of Medicine, Bronx, NY 10461.,Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
81
|
Chen W, Tang H, Zhou M, Hu C, Zhang J, Tang K. Dexamethasone inhibits the differentiation of rat tendon stem cells into tenocytes by targeting the scleraxis gene. J Steroid Biochem Mol Biol 2015; 152:16-24. [PMID: 25906952 DOI: 10.1016/j.jsbmb.2015.04.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/24/2015] [Accepted: 04/08/2015] [Indexed: 11/27/2022]
Abstract
Glucocorticoid-induced tendon rupture is very common in clinical practice, and the overall outcome of surgical suture repair is rather poor. The mechanism remains unclear, and effective treatments are still lacking. In the present study, we investigated the effect of dexamethasone on the differentiation of rat tendon stem cells (TSCs) to tenocytes and the underlying molecular mechanisms and found that dexamethasone inhibits the differentiation of TSCs to tenocytes by analyzing the development of long, spindle-shaped cells and detecting the expression of tenocyte markers type I collagen and tenomodulin (TNMD) at both the mRNA and protein levels. We also discovered that after treatment with dexamethasone, the scleraxis expression level is downregulated in vitro and in human specimen. Chromatin immunoprecipitation (ChIP)-PCR showed that dexamethasone promotes glucocorticoid receptor interacted with the TGGAAGCC sequence located between -734 and -726 base pairs (bp) upstream of the start codon of the scleraxis gene. Furthermore, TSCs were transfected with scleraxis knockdown or overexpression plasmids, and the results indicated that scleraxis plays a pivotal role in the differentiation of TSCs to tenocytes. In conclusion, dexamethasone inhibits the differentiation of TSCs to tenocytes by inhibiting the scleraxis gene.
Collapse
Affiliation(s)
- Wan Chen
- Department of Orthopedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Hong Tang
- Department of Orthopedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Mei Zhou
- Department of Orthopedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Chao Hu
- Department of Orthopedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiqiang Zhang
- Department of Neurology, Third Military Medical University, Chongqing 400038, China.
| | - Kanglai Tang
- Department of Orthopedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
82
|
Milet C, Duprez D. The Mkx homeoprotein promotes tenogenesis in stem cells and improves tendon repair. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:S33. [PMID: 26046080 DOI: 10.3978/j.issn.2305-5839.2015.03.64] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Cécile Milet
- 1 CNRS UMR 7622, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 2 Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 3 Inserm U1156, F-75005, Paris, France
| | - Delphine Duprez
- 1 CNRS UMR 7622, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 2 Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 3 Inserm U1156, F-75005, Paris, France
| |
Collapse
|
83
|
Markers for the identification of tendon-derived stem cells in vitro and tendon stem cells in situ - update and future development. Stem Cell Res Ther 2015; 6:106. [PMID: 26031740 PMCID: PMC4451873 DOI: 10.1186/s13287-015-0097-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The efficacy of tendon-derived stem cells (TDSCs) for the promotion of tendon and tendon-bone junction repair has been reported in animal studies. Modulation of the tendon stem cell niche in vivo has also been reported to influence tendon structure. There is a need to have specific and reliable markers that can define TDSCs in vitro and tendon stem cells in situ for several reasons: to understand the basic biology of TDSCs and their subpopulations in vitro; to understand the identity, niches and functions of tendon/progenitor stem cells in vivo; to meet the governmental regulatory requirements for quality of TDSCs when translating the exciting preclinical findings into clinical trial/practice; and to develop new treatment strategies for mobilizing endogenous stem/progenitor cells in tendon. TDSCs were reported to express the common mesenchymal stem cell (MSC) markers and some embryonic stem cell (ESC) markers, and there were attempts to use these markers to label tendon stem cells in situ. Are these stem cell markers useful for the identification of TDSCs in vitro and tracking of tendon stem cells in situ? This review aims to discuss the values of the panel of MSC, ESC and tendon-related markers for the identification of TDSCs in vitro. Important factors influencing marker expression by TDSCs are discussed. The usefulness and limitations of the panel of MSC, ESC and tendon-related markers for tracking stem cells in tendon, especially tendon stem cells, in situ are then reviewed. Future research directions are proposed.
Collapse
|
84
|
Valencia Mora M, Ruiz Ibán MA, Díaz Heredia J, Barco Laakso R, Cuéllar R, García Arranz M. Stem cell therapy in the management of shoulder rotator cuff disorders. World J Stem Cells 2015; 7:691-9. [PMID: 26029341 PMCID: PMC4444610 DOI: 10.4252/wjsc.v7.i4.691] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 01/26/2015] [Accepted: 02/04/2015] [Indexed: 02/07/2023] Open
Abstract
Rotator cuff tears are frequent shoulder problems that are usually dealt with surgical repair. Despite improved surgical techniques, the tendon-to-bone healing rate is unsatisfactory due to difficulties in restoring the delicate transitional tissue between bone and tendon. It is essential to understand the molecular mechanisms that determine this failure. The study of the molecular environment during embryogenesis and during normal healing after injury is key in devising strategies to get a successful repair. Mesenchymal stem cells (MSC) can differentiate into different mesodermal tissues and have a strong paracrine, anti-inflammatory, immunoregulatory and angiogenic potential. Stem cell therapy is thus a potentially effective therapy to enhance rotator cuff healing. Promising results have been reported with the use of autologous MSC of different origins in animal studies: they have shown to have better healing properties, increasing the amount of fibrocartilage formation and improving the orientation of fibrocartilage fibers with less immunologic response and reduced lymphocyte infiltration. All these changes lead to an increase in biomechanical strength. However, animal research is still inconclusive and more experimental studies are needed before human application. Future directions include expanded stem cell therapy in combination with growth factors or different scaffolds as well as new stem cell types and gene therapy.
Collapse
Affiliation(s)
- Maria Valencia Mora
- Maria Valencia Mora, Miguel A Ruiz Ibán, Jorge Díaz Heredia, Unidad de Hombro y Codo, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Miguel A Ruiz Ibán
- Maria Valencia Mora, Miguel A Ruiz Ibán, Jorge Díaz Heredia, Unidad de Hombro y Codo, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Jorge Díaz Heredia
- Maria Valencia Mora, Miguel A Ruiz Ibán, Jorge Díaz Heredia, Unidad de Hombro y Codo, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Raul Barco Laakso
- Maria Valencia Mora, Miguel A Ruiz Ibán, Jorge Díaz Heredia, Unidad de Hombro y Codo, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Ricardo Cuéllar
- Maria Valencia Mora, Miguel A Ruiz Ibán, Jorge Díaz Heredia, Unidad de Hombro y Codo, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Mariano García Arranz
- Maria Valencia Mora, Miguel A Ruiz Ibán, Jorge Díaz Heredia, Unidad de Hombro y Codo, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| |
Collapse
|
85
|
Gaspar D, Spanoudes K, Holladay C, Pandit A, Zeugolis D. Progress in cell-based therapies for tendon repair. Adv Drug Deliv Rev 2015; 84:240-56. [PMID: 25543005 DOI: 10.1016/j.addr.2014.11.023] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/08/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
The last decade has seen significant developments in cell therapies, based on permanently differentiated, reprogrammed or engineered stem cells, for tendon injuries and degenerative conditions. In vitro studies assess the influence of biophysical, biochemical and biological signals on tenogenic phenotype maintenance and/or differentiation towards tenogenic lineage. However, the ideal culture environment has yet to be identified due to the lack of standardised experimental setup and readout system. Bone marrow mesenchymal stem cells and tenocytes/dermal fibroblasts appear to be the cell populations of choice for clinical translation in equine and human patients respectively based on circumstantial, rather than on hard evidence. Collaborative, inter- and multi-disciplinary efforts are expected to provide clinically relevant and commercially viable cell-based therapies for tendon repair and regeneration in the years to come.
Collapse
Affiliation(s)
- Diana Gaspar
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Kyriakos Spanoudes
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Carolyn Holladay
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Dimitrios Zeugolis
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
86
|
Chen JL, Zhang W, Liu ZY, Heng BC, Ouyang HW, Dai XS. Physical regulation of stem cells differentiation into teno-lineage: current strategies and future direction. Cell Tissue Res 2014; 360:195-207. [DOI: 10.1007/s00441-014-2077-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/17/2014] [Indexed: 12/18/2022]
|
87
|
Russo V, Mauro A, Martelli A, Di Giacinto O, Di Marcantonio L, Nardinocchi D, Berardinelli P, Barboni B. Cellular and molecular maturation in fetal and adult ovine calcaneal tendons. J Anat 2014; 226:126-42. [PMID: 25546075 PMCID: PMC4304568 DOI: 10.1111/joa.12269] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2014] [Indexed: 02/06/2023] Open
Abstract
Processes of development during fetal life profoundly transform tendons from a plastic tissue into a highly differentiated structure, characterised by a very low ability to regenerate after injury in adulthood. Sheep tendon is frequently used as a translational model to investigate cell-based regenerative approaches. However, in contrast to other species, analytical and comparative baseline studies on the normal developmental maturation of sheep tendons from fetal through to adult life are not currently available. Thus, a detailed morphological and biochemical study was designed to characterise tissue maturation during mid- (2 months of pregnancy: 14 cm of length) and late fetal (4 months: 40 cm of length) life, through to adulthood. The results confirm that ovine tendon morphology undergoes profound transformations during this period. Endotenon was more developed in fetal tendons than in adult tissues, and its cell phenotype changed through tendon maturation. Indeed, groups of large rounded cells laying on smaller and more compacted ones expressing osteocalcin, vascular endothelial growth factor (VEGF) and nerve growth factor (NGF) were identified exclusively in fetal mid-stage tissues, and not in late fetal or adult tendons. VEGF, NGF as well as blood vessels and nerve fibers showed decreased expression during tendon development. Moreover, the endotenon of mid- and late fetuses contained identifiable cells that expressed several pluripotent stem cell markers [Telomerase Reverse Transcriptase (TERT), SRY Determining Region Y Box-2 (SOX2), Nanog Homeobox (NANOG) and Octamer Binding Transcription Factor-4A (OCT-4A)]. These cells were not identifiable in adult specimens. Ovine tendon development was also accompanied by morphological modifications to cell nuclei, and a progressive decrease in cellularity, proliferation index and expression of connexins 43 and 32. Tendon maturation was similarly characterised by modulation of several other gene expression profiles, including Collagen type I, Collagen type III, Scleraxis B, Tenomodulin, Trombospondin 4 and Osteocalcin. These gene profiles underwent a dramatic reduction in adult tissues. Transforming growth factor-1 expression (involved in collagen synthesis) underwent a similar decrease. In conclusion, these morphological studies carried out on sheep tendons at different stages of development and aging offer normal structural and molecular baseline data to allow accurate evaluation of data from subsequent interventional studies investigating tendon healing and regeneration in ovine experimental models.
Collapse
|
88
|
Spanoudes K, Gaspar D, Pandit A, Zeugolis DI. The biophysical, biochemical, and biological toolbox for tenogenic phenotype maintenance in vitro. Trends Biotechnol 2014; 32:474-82. [PMID: 25043371 DOI: 10.1016/j.tibtech.2014.06.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/16/2014] [Accepted: 06/25/2014] [Indexed: 12/14/2022]
Abstract
Tendon injuries constitute an unmet clinical need, with 3 to 5 million new incidents occurring annually worldwide. Tissue grafting and biomaterial-based approaches fail to provide environments that are conducive to regeneration; instead they lead to nonspecific cell adhesion and scar tissue formation, which collectively impair functionality. Cell based therapies may potentially recover native tendon function, if tenocyte trans-differentiation can be evaded and stem cell differentiation towards tenogenic lineage is attained. To this end, recreating an artificial in vivo tendon niche by engineering functional in vitro microenvironments is a research priority. Clinically relevant cell based therapies for tendon repair and regeneration could be created using tools that harness biophysical beacons (surface topography, mechanical loading), biochemical cues (oxygen tension), and biological signals (growth factors).
Collapse
Affiliation(s)
- Kyriakos Spanoudes
- Network of Excellence for Functional Biomaterials (NFB), Biosciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Diana Gaspar
- Network of Excellence for Functional Biomaterials (NFB), Biosciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials (NFB), Biosciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Dimitrios I Zeugolis
- Network of Excellence for Functional Biomaterials (NFB), Biosciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
89
|
Czubryt MP. A tale of 2 tissues: the overlapping role of scleraxis in tendons and the heart. Can J Physiol Pharmacol 2014; 92:707-12. [PMID: 25083735 DOI: 10.1139/cjpp-2013-0489] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue integrity in the face of external physical forces requires the production of a strong extracellular matrix (ECM) composed primarily of the protein collagen. Tendons and the heart both withstand large and changing physical forces, and emerging evidence suggests that the transcription factor scleraxis plays a central role in responding to these forces by directly regulating the production of ECM components and (or) by determining the fate of matrix-producing cell types. Thus, despite the highly disparate inherent nature of these tissues, a common response mechanism may exist to govern the development, growth, and remodeling of the ECM in response to external force.
Collapse
Affiliation(s)
- Michael P Czubryt
- R4008 St. Boniface Research Centre, 351 Tache Avenue, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
90
|
Tang QM, Chen JL, Shen WL, Yin Z, Liu HH, Fang Z, Heng BC, Ouyang HW, Chen X. Fetal and adult fibroblasts display intrinsic differences in tendon tissue engineering and regeneration. Sci Rep 2014; 4:5515. [PMID: 24992450 PMCID: PMC4080701 DOI: 10.1038/srep05515] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 06/12/2014] [Indexed: 12/14/2022] Open
Abstract
Injured adult tendons do not exhibit optimal healing through a regenerative process, whereas fetal tendons can heal in a regenerative fashion without scar formation. Hence, we compared FFs (mouse fetal fibroblasts) and AFs (mouse adult fibroblasts) as seed cells for the fabrication of scaffold-free engineered tendons. Our results demonstrated that FFs had more potential for tendon tissue engineering, as shown by higher levels of tendon-related gene expression. In the in situ AT injury model, the FFs group also demonstrated much better structural and functional properties after healing, with higher levels of collagen deposition and better microstructure repair. Moreover, fetal fibroblasts could increase the recruitment of fibroblast-like cells and reduce the infiltration of inflammatory cells to the injury site during the regeneration process. Our results suggest that the underlying mechanisms of better regeneration with FFs should be elucidated and be used to enhance adult tendon healing. This may assist in the development of future strategies to treat tendon injuries.
Collapse
Affiliation(s)
- Qiao-Mei Tang
- 1] Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, China, 310058 [2] Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine School of Medicine, Zhejiang University, Hangzhou China. 310058
| | - Jia Lin Chen
- 1] Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, China, 310058 [2] Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine School of Medicine, Zhejiang University, Hangzhou China. 310058
| | - Wei Liang Shen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China, 310058
| | - Zi Yin
- 1] Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, China, 310058 [2] Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine School of Medicine, Zhejiang University, Hangzhou China. 310058
| | - Huan Huan Liu
- 1] Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, China, 310058 [2] Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine School of Medicine, Zhejiang University, Hangzhou China. 310058
| | - Zhi Fang
- 1] Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, China, 310058 [2] Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine School of Medicine, Zhejiang University, Hangzhou China. 310058
| | - Boon Chin Heng
- Department of Biosystems Science & Engineering (D-BSSE), ETH-Zurich, Mattenstrasse 26, Basel 4058, Switzerland
| | - Hong Wei Ouyang
- 1] Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, China, 310058 [2] Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine School of Medicine, Zhejiang University, Hangzhou China. 310058
| | - Xiao Chen
- 1] Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, China, 310058 [2] Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine School of Medicine, Zhejiang University, Hangzhou China. 310058
| |
Collapse
|
91
|
Tan C, Lui PPY, Lee YW, Wong YM. Scx-transduced tendon-derived stem cells (tdscs) promoted better tendon repair compared to mock-transduced cells in a rat patellar tendon window injury model. PLoS One 2014; 9:e97453. [PMID: 24831949 PMCID: PMC4022525 DOI: 10.1371/journal.pone.0097453] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 04/21/2014] [Indexed: 02/06/2023] Open
Abstract
We hypothesized that the transplantation of Scx-transduced tendon-derived stem cells (TDSCs) promoted better tendon repair compared to the transplantation of mock-transduced cells. This study thus aimed to investigate the effect of Scx transduction on the expression of lineage markers in TDSCs and the effect of the resulting cell line in the promotion of tendon repair. Rat non-GFP or GFP-TDSCs were transduced with Scx or empty lentiviral vector (Mock) and selected by blasticidin. The mRNA expressions of Scx and different lineage markers were examined by qRT-PCR. The effect of the transplantation of GFP-TDSC-Scx on tendon repair was then tested in a rat unilateral patellar tendon window injury model. The transplantation of GFP-TDSC-Mock and scaffold-only served as controls. At week 2, 4 and 8 post-transplantation, the repaired patellar tendon was harvested for ex vivo fluorescent imaging, vivaCT imaging, histology, immunohistochemistry and biomechanical test. GFP-TDSC-Scx consistently showed higher expressions of most of tendon- and cartilage- related markers compared to the GFP-TDSC-Mock. However, the effect of Scx transduction on the expressions of bone-related markers was inconclusive. The transplanted GFP-TDSCs could be detected in the window wound at week 2 but not at week 4. Ectopic mineralization was detected in some samples at week 8 but there was no difference among different groups. The GFP-TDSC-Scx group only statistically significantly improved tendon repair histologically and biomechanically compared to the Scaffold-only group and the GFP-TDSC-Mock group at the early stage of tendon repair. There was significant higher expression of collagen type I in the window wound in the GFP-TDSC-Scx group compared to the other two groups at week 2. The transplantation of GFP-TDSC-Scx promoted healing at the early stage of tendon repair in a rat patellar tendon window injury model.
Collapse
Affiliation(s)
- Chunlai Tan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Yuk Wa Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yin Mei Wong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Hong Kong Jockey Club Sports Medicine and Health Sciences Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
92
|
Miyabara S, Yuda Y, Kasashima Y, Kuwano A, Arai K. Regulation of Tenomodulin Expression Via Wnt/β-catenin Signaling in Equine Bone Marrow-derived Mesenchymal Stem Cells. J Equine Sci 2014; 25:7-13. [PMID: 24834008 PMCID: PMC4019198 DOI: 10.1294/jes.25.7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 01/30/2014] [Indexed: 01/06/2023] Open
Abstract
Tenomodulin has been recognized as a biomarker for tendon differentiation, and its gene expression is regulated by several
transcription factors including Scleraxis and Mohawk. In this study, we found a novel regulatory mechanism of tenomodulin
expression. Equine bone marrow-derived mesenchymal stem cells (BMSCs) in monolayer culture showed a low mRNA level of tenomodulin
in comparison with the level in the tendon. When cultured in collagen gel containing a glycogen synthase kinase-3 (GSK-3)
inhibitor (BIO), expression of tenomodulin in BMSCs increased up to the level in the tendon. Participation of GSK-3 in its gene
expression was further demonstrated by a gene silencing experiment with small interference RNA corresponding to GSK-3, suggesting
that Wnt/β-catenin signaling mediated expression of tenomodulin. These results were confirmed by nuclear translocation of
β-catenin in BIO-treated BMSCs cultured in collagen gel. Under this culture condition, expression of tenomodulin-related
transcription factors including Scleraxis and Mohawk was not affected, suggesting that Wnt/β-catenin signaling was independent
from these transcription factors. Additionally, BIO strongly enhanced expression of type XIV collagen in collagen-embedded BMSCs
up to the level in the tendon, and other tendon-related extracellular matrix components such as decorin and fibromodulin were also
upregulated. Taken together, these results indicated that activation of Wnt/β-catenin signaling could induce differentiation of
BMSCs into tenomodulin-expressing tendon cells in collagen gel.
Collapse
Affiliation(s)
- Shihori Miyabara
- Department of Tissue Physiology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Yohei Yuda
- Department of Tissue Physiology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Yoshinori Kasashima
- Laboratory of Clinical Science and Pathobiology, Equine Research Institute, Japan Racing Association, Tochigi 320-8056, Japan
| | - Atsutoshi Kuwano
- Laboratory of Clinical Science and Pathobiology, Equine Research Institute, Japan Racing Association, Tochigi 320-8056, Japan
| | - Katsuhiko Arai
- Department of Tissue Physiology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
93
|
Barsby T, Bavin EP, Guest DJ. Three-dimensional culture and transforming growth factor beta3 synergistically promote tenogenic differentiation of equine embryo-derived stem cells. Tissue Eng Part A 2014; 20:2604-13. [PMID: 24628376 DOI: 10.1089/ten.tea.2013.0457] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The natural reparative mechanisms triggered by tendon damage often lead to the formation of biomechanically inferior scar tissue that is prone to re-injury. Before the efficient application of stem cell-based regenerative therapies, the processes regulating tenocyte differentiation should first be better understood. Three-dimensional (3D) growth environments under strain and the exogenous addition of transforming growth factor beta3 (TGF-β3) have separately been shown to promote tendon differentiation. The aim of this study was to determine the ability of both of these factors to induce tendon differentiation of equine embryo-derived stem cells (ESCs). ESCs seeded into 3D collagen constructs can contract the matrix to a similar degree to that of tenocyte-seeded constructs and histologically appear nearly identical, with no areas of cartilage or bone tissue deposition. Tendon-associated genes and proteins Tenascin-C, Collagen Type I, and COMP are significantly up-regulated in the 3D ESC constructs compared with tenogenic induction in monolayer ESC cultures. The addition of TGF-β3 to the 3D cultures further up-regulates the expression of these genes and also induces the expression of mature tenocyte markers Tenomodulin and Thrombospondin-4. Our results show that when ESCs are exposed to the intrinsic forces exerted by a 3D culture environment, they express tendon-associated genes and proteins which are indicative of tenocyte lineage differentiation and that this effect is synergistically enhanced and accelerated by the addition of TGF-β3.
Collapse
Affiliation(s)
- Tom Barsby
- Animal Health Trust, Centre for Preventive Medicine , Newmarket, Suffolk, United Kingdom
| | | | | |
Collapse
|
94
|
Yang G, Rothrauff BB, Tuan RS. Tendon and ligament regeneration and repair: clinical relevance and developmental paradigm. ACTA ACUST UNITED AC 2014; 99:203-222. [PMID: 24078497 DOI: 10.1002/bdrc.21041] [Citation(s) in RCA: 295] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 07/27/2013] [Accepted: 07/27/2013] [Indexed: 12/18/2022]
Abstract
As dense connective tissues connecting bone to muscle and bone to bone, respectively, tendon and ligament (T/L) arise from the somitic mesoderm, originating in a recently discovered somitic compartment, the syndetome. Inductive signals from the adjacent sclerotome and myotome upregulate expression of Scleraxis, a key transcription factor for tenogenic and ligamentogenic differentiation. Understanding T/L development is critical to establishing a knowledge base for improving the healing and repair of T/L injuries, a high-burden disease due to the intrinsically poor natural healing response. Current treatment of the three most common tendon injuries-tearing of the rotator cuff of the shoulder, flexor tendon of the hand, and Achilles tendon-include mostly surgical repair and/or conservative approaches, including biophysical modalities such as rehabilitation and cryotherapy. Unfortunately, the fibrovascular scar formed during healing possesses inferior mechanical and biochemical properties, resulting in compromised tissue functionality. Regenerative approaches have sought to augment the injured tissue with cells, scaffolds, bioactive agents, and mechanical stimulation to improve the natural healing response. The key challenges in restoring full T/L function following injury include optimal combination of these biological agents as well as their delivery to the injury site. A greater understanding of the molecular mechanisms involved in T/L development and natural healing, coupled with the capability of producing complex biomaterials to deliver multiple biofactors with high spatiotemporal resolution and specificity, should lead to regenerative procedures that more closely recapitulate T/L morphogenesis, thereby offering future patients the prospect of T/L regeneration, as opposed to simple tissue repair.
Collapse
Affiliation(s)
- Guang Yang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Benjamin B Rothrauff
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
95
|
Liu H, Zhu S, Zhang C, Lu P, Hu J, Yin Z, Ma Y, Chen X, OuYang H. Crucial transcription factors in tendon development and differentiation: their potential for tendon regeneration. Cell Tissue Res 2014; 356:287-98. [PMID: 24705622 DOI: 10.1007/s00441-014-1834-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/30/2014] [Indexed: 12/22/2022]
Abstract
Tendons that connect muscles to bone are often the targets of sports injuries. The currently unsatisfactory state of tendon repair is largely attributable to the limited understanding of basic tendon biology. A number of tendon lineage-related transcription factors have recently been uncovered and provide clues for the better understanding of tendon development. Scleraxis and Mohawk have been identified as critical transcription factors in tendon development and differentiation. Other transcription factors, such as Sox9 and Egr1/2, have also been recently reported to be involved in tendon development. However, the molecular mechanisms and application of these transcription factors remain largely unclear and this prohibits their use in tendon therapy. Here, we systematically review and analyze recent findings and our own data concerning tendon transcription factors and tendon regeneration. Based on these findings, we provide interaction and temporal programming maps of transcription factors, as a basis for future tendon therapy. Finally, we discuss future directions for tendon regeneration with differentiation and trans-differentiation approaches based on transcription factors.
Collapse
Affiliation(s)
- Huanhuan Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou, 310058, China
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Tang SW, Tong WY, Shen W, Yeung KWK, Lam YW. Stringent requirement for spatial arrangement of extracellular matrix in supporting cell morphogenesis and differentiation. BMC Cell Biol 2014; 15:10. [PMID: 24661496 PMCID: PMC3987840 DOI: 10.1186/1471-2121-15-10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 03/19/2014] [Indexed: 01/30/2023] Open
Abstract
Background In vitro experiments on the functional roles of extracellular matrix (ECM) components usually involve the culture of cells on surfaces coated with purified ECM components. These experiments can seldom recuperate the spatial arrangement of ECM found in vivo. In this study, we have overcome this obstacle by using histological sections of bovine Achilles tendon as cell culture substrates. Results We found that tendon sections can be viewed as a pre-formed block of ECM in which the collagen fibrils exhibited a spatial regularity unraveled in any artificially constructed scaffold. By carving the tendon at different angles relative to its main axis, we created different surfaces with distinct spatial arrangements of collagen fibrils. To assess the cellular responses to these surfaces, human mesenchymal stem cells (MSCs) were directly cultured on these sections, hence exposed to the collagen with different spatial orientations. Cells seeded on longitudinal tendon sections adopted a highly elongated and aligned morphology, and expressed an increased level of tenomodulin, suggesting that the collagen fibrils present in this section provide a microenvironment that facilitates cell morphogenesis and differentiation. However, MSC elongation, alignment and induction of tenomodulin diminished dramatically even as the sectioned angle changed slightly. Conclusion Our results suggest that cell functions are influenced not only by the type or concentration of ECM components, but also by the precise spatial arrangements of these molecules. The method developed in this study offers a simple and robust way for the studying of cell-ECM interactions, and opens many research avenues in the field of matrix biology.
Collapse
Affiliation(s)
| | | | | | - Kelvin W K Yeung
- Department of Biology & Chemistry, City University of Hong Kong, Hong Kong, China.
| | | |
Collapse
|
97
|
Moshaverinia A, Xu X, Chen C, Ansari S, Zadeh HH, Snead ML, Shi S. Application of stem cells derived from the periodontal ligament or gingival tissue sources for tendon tissue regeneration. Biomaterials 2014; 35:2642-50. [PMID: 24397989 DOI: 10.1016/j.biomaterials.2013.12.053] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 12/19/2013] [Indexed: 12/24/2022]
Abstract
Tendon injuries are often associated with significant dysfunction and disability due to tendinous tissue's very limited self-repair capacity and propensity for scar formation. Dental-derived mesenchymal stem cells (MSCs) in combination with appropriate scaffold material present an alternative therapeutic option for tendon repair/regeneration that may be advantageous compared to other current treatment modalities. The MSC delivery vehicle is the principal determinant for successful implementation of MSC-mediated regenerative therapies. In the current study, a co-delivery system based on TGF-β3-loaded RGD-coupled alginate microspheres was developed for encapsulating periodontal ligament stem cells (PDLSCs) or gingival mesenchymal stem cells (GMSCs). The capacity of encapsulated dental MSCs to differentiate into tendon tissue was investigated in vitro and in vivo. Encapsulated dental-derived MSCs were transplanted subcutaneously into immunocompromised mice. Our results revealed that after 4 weeks of differentiation in vitro, PDLSCs and GMSCs as well as the positive control human bone marrow mesenchymal stem cells (hBMMSCs) exhibited high levels of mRNA expression for gene markers related to tendon regeneration (Scx, DCn, Tnmd, and Bgy) via qPCR measurement. In a corresponding in vivo animal model, ectopic neo-tendon regeneration was observed in subcutaneous transplanted MSC-alginate constructs, as confirmed by histological and immunohistochemical staining for protein markers specific for tendons. Interestingly, in our quantitative PCR and in vivo histomorphometric analyses, PDLSCs showed significantly greater capacity for tendon regeneration than GMSCs or hBMMSCs (P < 0.05). Altogether, these findings indicate that periodontal ligament and gingival tissues can be considered as suitable stem cell sources for tendon engineering. PDLSCs and GMSCs encapsulated in TGF-β3-loaded RGD-modified alginate microspheres are promising candidates for tendon regeneration.
Collapse
Affiliation(s)
- Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.
| | - Xingtian Xu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Chider Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Sahar Ansari
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Homayoun H Zadeh
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Malcolm L Snead
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Songtao Shi
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
98
|
Tanabe S. Perspectives of gene combinations in phenotype presentation. World J Stem Cells 2013; 5:61-67. [PMID: 23951387 PMCID: PMC3744131 DOI: 10.4252/wjsc.v5.i3.61] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/22/2013] [Accepted: 06/08/2013] [Indexed: 02/06/2023] Open
Abstract
Cells exhibit a variety of phenotypes in different stages and diseases. Although several markers for cellular phenotypes have been identified, gene combinations denoting cellular phenotypes have not been completely elucidated. Recent advances in gene analysis have revealed that various gene expression patterns are observed in each cell species and status. In this review, the perspectives of gene combinations in cellular phenotype presentation are discussed. Gene expression profiles change during cellular processes, such as cell proliferation, cell differentiation, and cell death. In addition, epigenetic regulation increases the complexity of the gene expression profile. The role of gene combinations and panels of gene combinations in each cellular condition are also discussed.
Collapse
Affiliation(s)
- Shihori Tanabe
- Shihori Tanabe, Division of Safety Information on Drug, Food and Chemicals, National Institute of Health Sciences, Tokyo 158-8501, Japan
| |
Collapse
|
99
|
Leung M, Jana S, Tsao CT, Zhang M. Tenogenic differentiation of human bone marrow stem cells via a combinatory effect of aligned chitosan–poly-caprolactone nanofibers and TGF-β3. J Mater Chem B 2013; 1:6516-6524. [DOI: 10.1039/c3tb20825g] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|