1
|
Malvandi AM, Gerosa L, Banfi G, Lombardi G. The bone-muscle unit: from mechanical coupling to soluble factors-mediated signaling. Mol Aspects Med 2025; 103:101367. [PMID: 40339487 DOI: 10.1016/j.mam.2025.101367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
Skeletal muscles (SKM) and bones form a morpho-functional unit, interconnected throughout life primarily through biomechanical coupling. This relationship serves as a key reciprocal stimulus, but they also interact via various hormones, such as sex steroids, growth hormone-insulin-like growth factor 1 (GH-IGF1) axis hormones, and adipokines like leptin and adiponectin. Additionally, myokines (released by muscles) and osteokines (released by bones) facilitate dense crosstalk, influencing each other's activity. Key myokines include interleukin (IL)-6, IL-7, IL-15, and myostatin, while osteocalcin (OC) and sclerostin are crucial bone-derived mediators affecting SKM cells. Moreover, miRNAs act as endocrine-like regulators, contributing to a complex network. This review covers the current understanding of bone-muscle crosstalk, which is essential for grasping the musculoskeletal apparatus's role in disease pathogenesis and may inform therapeutic development.
Collapse
Affiliation(s)
- Amir Mohammad Malvandi
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
| | - Laura Gerosa
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
| | - Giuseppe Banfi
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy; Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland.
| |
Collapse
|
2
|
Usami Y, Jiang X, Dyment NA, Kokubun T. Limb Motility and Ambulation as Mechanical Cues in Postnatal Murine Tendon Development. Dev Biol 2025:S0012-1606(25)00143-5. [PMID: 40414450 DOI: 10.1016/j.ydbio.2025.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 05/08/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
The musculoskeletal system provides structural stability and coordination to enable movement. Tendons have the essential role of efficiently transmitting force generated from muscle contraction to bone for ambulation. In doing so, they resist high mechanical loads. Muscle contraction during embryonic development is required for continued tendon growth and differentiation. Defining the types and magnitudes of loads that act on tendons during the early postnatal periods is quite difficult. This study aimed to reveal whether limb motility and spontaneous physical behaviors in the postnatal phase work as mechanical cues for tendon development, leading to mechanobiological phenomena during postnatal phases in the murine model. Neonatal mice showed gradual limb motility, rollover function, and ambulation patterns during early postnatal phases. Tendons showed lengthening, decreasing the cell density and nuclear roundness concurrently. Scx and Tnmd gene expression showed a tendency to increase with time as well. This study presented a comprehensive time course of limb movement and ambulation with corresponding tendon growth during the postnatal phase. Our chronological analysis of the relationship between changing limb loading and tendon development provides a foundation for future work focused on mechanobiology in tendon development.
Collapse
Affiliation(s)
- Yuna Usami
- Graduate School of Health, Medicine, and Welfare, Saitama Prefectural University, Saitama, Japan
| | - Xi Jiang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Takanori Kokubun
- Graduate School of Health, Medicine, and Welfare, Saitama Prefectural University, Saitama, Japan; Department of Physical Therapy, School of Health and Social Services, Saitama Prefectural University, Saitama, Japan.
| |
Collapse
|
3
|
Kwan KYC, Li K, Wang YY, Tse WY, Tong CY, Zhang X, Wang DM, Ker DFE. The Characterization of Serum-Free Media on Human Mesenchymal Stem Cell Fibrochondrogenesis. Bioengineering (Basel) 2025; 12:546. [PMID: 40428164 PMCID: PMC12109459 DOI: 10.3390/bioengineering12050546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/03/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Developing fibrochondrogenic serum-free media is important for regenerating diseased and injured fibrocartilage but no defined protocols exist. Towards this goal, we characterized the effect of four candidate fibrochondrogenic serum-free media containing transforming growth factor beta-3 (TGF-β3), insulin-like growth factor-1 (IGF-1), and fibroblast growth factor-2 (FGF-2) with high/low glucose and with/without dexamethasone on human mesenchymal stem cells (hMSCs) via proliferation and differentiation assays. In Ki67 proliferation assays, serum-free media containing low glucose and dexamethasone exhibited the highest growth. In gene expression assays, serum-free media containing low glucose and commercially available chondrogenic media (COM) induced high fibrochondrogenic transcription factor expression (scleraxis/SCX and SRY-Box Transcription Factor 9/SOX9) and extracellular matrix (ECM) protein levels (aggrecan/ACAN, collagen type I/COL1A1, and collagen type II/COL2A1), respectively. In immunofluorescence staining, serum-free media containing high glucose and COM induced high fibrochondrogenic transcription factor (SCX and SOX9) and ECM protein (COL1A1, COL2A1, and collagen type X/COL10A1) levels, respectively. In cytochemical staining, COM and serum-free media containing dexamethasone showed a high collagen content whereas serum-free media containing high glucose and dexamethasone exhibited high glycosaminoglycan (GAG) levels. Altogether, defined serum-free media containing high glucose exhibited the highest fibrochondrogenic potential. In summary, this work studied conditions conducive for fibrochondrogenesis, which may be further optimized for potential applications in fibrocartilage tissue engineering.
Collapse
Affiliation(s)
- Ka Yu Carissa Kwan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (K.Y.C.K.); (K.L.); (Y.Y.W.); (W.Y.T.); (C.Y.T.); (X.Z.); (D.M.W.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ke Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (K.Y.C.K.); (K.L.); (Y.Y.W.); (W.Y.T.); (C.Y.T.); (X.Z.); (D.M.W.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yu Yang Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (K.Y.C.K.); (K.L.); (Y.Y.W.); (W.Y.T.); (C.Y.T.); (X.Z.); (D.M.W.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wai Yi Tse
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (K.Y.C.K.); (K.L.); (Y.Y.W.); (W.Y.T.); (C.Y.T.); (X.Z.); (D.M.W.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Chung Yan Tong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (K.Y.C.K.); (K.L.); (Y.Y.W.); (W.Y.T.); (C.Y.T.); (X.Z.); (D.M.W.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xu Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (K.Y.C.K.); (K.L.); (Y.Y.W.); (W.Y.T.); (C.Y.T.); (X.Z.); (D.M.W.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (K.Y.C.K.); (K.L.); (Y.Y.W.); (W.Y.T.); (C.Y.T.); (X.Z.); (D.M.W.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, Hong Kong SAR, China
- Ministry of Education Key Laboratory for Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Dai Fei Elmer Ker
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| |
Collapse
|
4
|
Panebianco CJ, Essaidi M, Barnes E, Williams A, Vancíková K, Labberté MC, Brama P, Nowlan NC, Boerckel JD. Dynamics of postnatal bone development and epiphyseal synostosis in the caprine autopod. Dev Dyn 2025. [PMID: 40359336 DOI: 10.1002/dvdy.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Bones develop to structurally balance strength and mobility. Bone developmental dynamics are influenced by whether an animal is ambulatory at birth. Precocial species, which are ambulatory at birth, develop advanced skeletal maturity in utero and experience postnatal development under mechanical loading. Here, we characterized postnatal bone development in the lower forelimbs of precocial goats using microcomputed tomography and histology. Our analysis focused on the two phalanges 1 (P1) bones and the partially fused metacarpal bone of the goat autopod from birth through adulthood. RESULTS P1 cortical bone densified rapidly after birth, but cortical thickness increased continually through adulthood. Upon normalization by body mass, the P1 normalized polar moment of inertia was constant over time, suggestive of changes correlating with ambulatory loading. P1 trabecular bone increased in trabecular number and thickness until sexual maturity (12 months), while metacarpal trabeculae grew primarily through trabecular thickening. Unlike prenatal synostosis (i.e., bone fusion) of the metacarpal diaphysis, synostosis of the epiphyses occurred postnatally, prior to growth plate closure, through a unique fibrocartilaginous endochondral ossification. CONCLUSIONS These findings implicate ambulatory loading in postnatal bone development of precocial goats and identify a novel postnatal synostosis event in the caprine metacarpal epiphysis.
Collapse
Affiliation(s)
- Christopher J Panebianco
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maha Essaidi
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elijah Barnes
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- School of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, Alabama, USA
| | - Ashley Williams
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- School of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, Alabama, USA
| | - Karin Vancíková
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
- UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Margot C Labberté
- Translational Research Unit, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Pieter Brama
- Translational Research Unit, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Niamh C Nowlan
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
- UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Roelofs AJ, McClure JJ, Hay EA, De Bari C. Stem and progenitor cells in the synovial joint as targets for regenerative therapy. Nat Rev Rheumatol 2025; 21:211-220. [PMID: 40045009 DOI: 10.1038/s41584-025-01222-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 03/28/2025]
Abstract
Damage to articular cartilage, tendons, ligaments and entheses as a result of trauma, degeneration or inflammation in rheumatic diseases is prevalent. Regenerative medicine offers promising strategies for repairing damaged tissues, with the aim of restoring both their structure and function. While these strategies have traditionally relied on tissue engineering approaches using exogenous cells, interventions based on the activation of endogenous repair mechanisms are an attractive alternative. Key to advancing such approaches is a comprehensive understanding of the diversity of the stem and progenitor cells that reside in the adult synovial joint and how they function to repair damaged tissues. Advances in developmental biology have provided a lens through which to understand the origins, identities and functions of these cells, and insights into the roles of stem and progenitor cells in joint tissue repair, as well as their complex relationship with fibroblasts, have emerged. Integration of knowledge obtained through studies using advanced single-cell technologies will be crucial to establishing unified models of cell populations, lineage hierarchies and their molecular regulation. Ultimately, a more complete understanding of how cells repair tissues in adult life will guide the development of innovative pro-regenerative drugs, which are poised to enter clinical practice in musculoskeletal medicine.
Collapse
Affiliation(s)
- Anke J Roelofs
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Jessica J McClure
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Elizabeth A Hay
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Cosimo De Bari
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK.
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
6
|
Nayak PK, Subramanian A, Schilling TF. Transcriptome profiling of tendon fibroblasts at the onset of embryonic muscle contraction reveals novel force-responsive genes. eLife 2025; 14:e105802. [PMID: 40145570 PMCID: PMC12040314 DOI: 10.7554/elife.105802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Mechanical forces play a critical role in tendon development and function, influencing cell behavior through mechanotransduction signaling pathways and subsequent extracellular matrix (ECM) remodeling. Here, we investigate the molecular mechanisms by which tenocytes in developing zebrafish embryos respond to muscle contraction forces during the onset of swimming and cranial muscle activity. Using genome-wide bulk RNA sequencing of FAC-sorted tenocytes we identify novel tenocyte markers and genes involved in tendon mechanotransduction. Embryonic tendons show dramatic changes in expression of matrix remodeling associated 5b (mxra5b), matrilin 1 (matn1), and the transcription factor kruppel-like factor 2a (klf2a), as muscles start to contract. Using embryos paralyzed either by loss of muscle contractility or neuromuscular stimulation we confirm that muscle contractile forces influence the spatial and temporal expression patterns of all three genes. Quantification of these gene expression changes across tenocytes at multiple tendon entheses and myotendinous junctions reveals that their responses depend on force intensity, duration, and tissue stiffness. These force-dependent feedback mechanisms in tendons, particularly in the ECM, have important implications for improved treatments of tendon injuries and atrophy.
Collapse
Affiliation(s)
- Pavan K Nayak
- Department of Developmental and Cell Biology, University of CaliforniaIrvineUnited States
| | - Arul Subramanian
- Department of Developmental and Cell Biology, University of CaliforniaIrvineUnited States
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of CaliforniaIrvineUnited States
| |
Collapse
|
7
|
Yu X, Kawakami R, Yambe S, Yoshimoto Y, Sasaki T, Higuchi S, Watanabe H, Akiyama H, Miura S, Hu K, Kondoh G, Sagasaki R, Inui M, Adachi T, Docheva D, Imamura T, Shukunami C. Dynamic interactions between cartilaginous and tendinous/ligamentous primordia during musculoskeletal integration. Development 2025; 152:dev204512. [PMID: 40135875 DOI: 10.1242/dev.204512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
Proper connections between cartilaginous and muscular primordia through tendinous/ligamentous primordia are essential for musculoskeletal integration. Herein, we report a novel double-reporter mouse model for investigating this process via fluorescently visualising scleraxis (Scx) and SRY-box containing gene 9 (Sox9) expression. We generated ScxTomato transgenic mice and crossed them with Sox9EGFP knock-in mice to obtain ScxTomato;Sox9EGFP mice. Deep imaging of optically cleared double-reporter embryos at E13.5 and E16.5 revealed previously unknown differences in the dynamic interactions between cartilaginous and tendinous/ligamentous primordia in control and Scx-deficient mice. Tendon/ligament maturation was evaluated through simultaneous detection of fluorescence and visualisation of collagen fibre formation using second harmonic generation imaging. Lack of deltoid tuberosity in Scx-deficient mice caused misdirected muscle attachment with morphological changes. Loss of Scx also dysregulated progenitor cell fate determination in the chondrotendinous junction, resulting in the formation of a rounded enthesis rather than the protruding enthesis observed in the control. Hence, our double-reporter mouse system, in combination with loss- or gain-of-function approaches, is a unique and powerful tool that could be used to gain a comprehensive understanding of musculoskeletal integration.
Collapse
Affiliation(s)
- Xinyi Yu
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Ryosuke Kawakami
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Ehime 791-0295, Japan
| | - Shinsei Yambe
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Takako Sasaki
- Department of Biochemistry II, Faculty of Medicine, Oita University, Oita 879-5593, Japan
| | - Shinnosuke Higuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Haruhiko Akiyama
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu 501-1193, Japan
| | - Shigenori Miura
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kadi Hu
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Ramu Sagasaki
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| | - Masafumi Inui
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| | - Taiji Adachi
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97074 Würzburg, Germany
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Ehime 791-0295, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
8
|
Tam KT, Baar K. Using load to improve tendon/ligament tissue engineering and develop novel treatments for tendinopathy. Matrix Biol 2025; 135:39-54. [PMID: 39645093 DOI: 10.1016/j.matbio.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Tendon and ligament injuries are highly prevalent but heal poorly, even with proper care. Restoration of native tissue function is complicated by the fact that these tissues vary anatomically in terms of their mechanical properties, composition, and structure. These differences develop as adaptations to diverse mechanical demands; however, pathology may alter the loads placed on the tissue. Musculoskeletal loads can be generally categorized into tension, compression, and shear. Each of these regulate distinct molecular pathways that are involved in tissue remodeling, including many of the canonical tenogenic genes. In this review, we provide a perspective on the stage-specific regulation of mechanically sensitive pathways during development and maturation of tendon and ligament tissue, including scleraxis, mohawk, and others. Furthermore, we discuss structural features of healing and diseased tendon that may contribute to aberrant loading profiles, and how the associated disturbance in molecular signaling may contribute to incomplete healing or the formation of degenerative phenotypes. The perspectives provided here draw from studies spanning in vitro, animal, and human experiments of healthy and diseased tendon to propose a more targeted approach to advance rehabilitation, orthobiologics, and tissue engineering.
Collapse
Affiliation(s)
- Kenneth T Tam
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA 95616, USA; Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | - Keith Baar
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA 95616, USA; Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; VA Northern California Health Care System, Mather, CA 95655, USA.
| |
Collapse
|
9
|
Panebianco CJ, Essaidi M, Barnes E, Williams A, Vancíková K, Labberté MC, Brama P, Nowlan NC, Boerckel JD. Dynamics of postnatal bone development and epiphyseal synostosis in the caprine autopod. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.26.630423. [PMID: 39763848 PMCID: PMC11703179 DOI: 10.1101/2024.12.26.630423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Bones develop to structurally balance strength and mobility. Bone developmental dynamics are influenced by whether an animal is ambulatory at birth (i.e., precocial). Precocial species, such as goats, develop advanced skeletal maturity in utero, making them useful models for studying the dynamics of bone formation under mechanical load. Here, we used microcomputed tomography and histology to characterize postnatal bone development in the autopod of the caprine lower forelimb. The caprine autopod features two toes, fused by metacarpal synostosis (i.e., bone fusion) prior to birth. Our analysis focused on the phalanges 1 (P1) and metacarpals of the goat autopod from birth through adulthood (3.5 years). P1 cortical bone densified rapidly after birth (half-life using one-phase exponential decay model (τ1/2 = 1.6 ± 0.4 months), but the P1 cortical thickness increased continually through adulthood (τ1/2 = 7.2 ± 2.7 mo). Upon normalization by body mass, the normalized polar moment of inertia of P1 cortical bone was constant over time, suggestive of structural load adaptation. P1 trabecular bone increased in trabecular number (τ1/2 = 6.7 ± 2.8 mo) and thickness (τ1/2 = 6.6 ± 2.0 mo) until skeletal maturity, while metacarpal trabeculae grew primarily through trabecular thickening (τ1/2 = 7.9 ± 2.2 mo). Unlike prenatal fusion of the metacarpal diaphysis, synostosis of the epiphyses occurred postnatally, prior to growth plate closure, through a unique fibrocartilaginous endochondral ossification. These findings implicate ambulatory loading in postnatal bone development of precocial goats and identify a novel postnatal synostosis event in the caprine metacarpal epiphysis.
Collapse
Affiliation(s)
- Christopher J. Panebianco
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Maha Essaidi
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Elijah Barnes
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- School of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL, USA
| | - Ashley Williams
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- School of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL, USA
| | - Karin Vancíková
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
- UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Margot C. Labberté
- Translational Research Unit, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Pieter Brama
- Translational Research Unit, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Niamh C. Nowlan
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
- UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Joel D. Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Dyment NA, Kamalitdinov TB, Kuntz AF. The 2024 Kappa Delta Young Investigator Award: Leveraging Insights From Development to Improve Adult Repair: Hedgehog Signaling as a Master Regulator of Enthesis Fibrocartilage Formation. J Am Acad Orthop Surg 2024; 32:1074-1086. [PMID: 39589737 PMCID: PMC11753257 DOI: 10.5435/jaaos-d-24-00996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 11/27/2024] Open
Abstract
The work in this article summarizes findings from our group on key biochemical cues that govern the formation and repair of tendons and ligaments. Specifically, we summarize the journey that started with a serendipitous discovery that is now being translated into novel therapies to improve tendon-to-bone repair outcomes. This journey began with the discovery that the Hedgehog (Hh) signaling pathway was expressed within the enthesis during development and that its primary role was to promote fibrocartilage production and maturation. Next, we developed an anterior cruciate ligament reconstruction model in novel transgenic mice that allowed us to discover that the Hh pathway promotes fibrocartilaginous tendon-to-bone attachments during the integration process. In addition, we established that the coordinated stages of zonal tendon-to-bone integration after anterior cruciate ligament reconstruction were comparable with the stages required for enthesis formation during development. Now that we have demonstrated that the Hh pathway is a potent therapeutic target, we are currently advancing these findings to develop drug delivery systems to improve tendon-to-bone repair. Ultimately, our group aims to establish key mechanisms that govern tendon and ligament formation that can be leveraged for novel regenerative therapies to improve clinical care.
Collapse
Affiliation(s)
- Nathaniel A Dyment
- From the Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA (Dr. Dyment, Dr. Kamalitdinov, and Dr. Kuntz), and the Department of Bioengineering, University of Pennsylvania, Philadelphia, PA (Dr. Dyment and Dr. Kamalitdinov)
| | | | | |
Collapse
|
11
|
Zhang B, He P, Lawrence JEG, Wang S, Tuck E, Williams BA, Roberts K, Kleshchevnikov V, Mamanova L, Bolt L, Polanski K, Li T, Elmentaite R, Fasouli ES, Prete M, He X, Yayon N, Fu Y, Yang H, Liang C, Zhang H, Blain R, Chedotal A, FitzPatrick DR, Firth H, Dean A, Bayraktar OA, Marioni JC, Barker RA, Storer MA, Wold BJ, Zhang H, Teichmann SA. A human embryonic limb cell atlas resolved in space and time. Nature 2024; 635:668-678. [PMID: 38057666 PMCID: PMC7616500 DOI: 10.1038/s41586-023-06806-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
Human limbs emerge during the fourth post-conception week as mesenchymal buds, which develop into fully formed limbs over the subsequent months1. This process is orchestrated by numerous temporally and spatially restricted gene expression programmes, making congenital alterations in phenotype common2. Decades of work with model organisms have defined the fundamental mechanisms underlying vertebrate limb development, but an in-depth characterization of this process in humans has yet to be performed. Here we detail human embryonic limb development across space and time using single-cell and spatial transcriptomics. We demonstrate extensive diversification of cells from a few multipotent progenitors to myriad differentiated cell states, including several novel cell populations. We uncover two waves of human muscle development, each characterized by different cell states regulated by separate gene expression programmes, and identify musculin (MSC) as a key transcriptional repressor maintaining muscle stem cell identity. Through assembly of multiple anatomically continuous spatial transcriptomic samples using VisiumStitcher, we map cells across a sagittal section of a whole fetal hindlimb. We reveal a clear anatomical segregation between genes linked to brachydactyly and polysyndactyly, and uncover transcriptionally and spatially distinct populations of the mesenchyme in the autopod. Finally, we perform single-cell RNA sequencing on mouse embryonic limbs to facilitate cross-species developmental comparison, finding substantial homology between the two species.
Collapse
Affiliation(s)
- Bao Zhang
- The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Peng He
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - John E G Lawrence
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Trauma and Orthopaedics, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Shuaiyu Wang
- The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Obstetrics, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Elizabeth Tuck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Brian A Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kenny Roberts
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Lira Mamanova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Enhanc3D Genomics Ltd, Cambridge, UK
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Genomics England, London, UK
| | | | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Rasa Elmentaite
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Eirini S Fasouli
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Basic Research Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Xiaoling He
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Nadav Yayon
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Yixi Fu
- The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hao Yang
- The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chen Liang
- The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Raphael Blain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Alain Chedotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Institut de pathologie, groupe hospitalier Est, hospices civils de Lyon, Lyon, France
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | | | - Helen Firth
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Andrew Dean
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Foundation, Cambridge, UK
| | | | - John C Marioni
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Mekayla A Storer
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Barbara J Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Hongbo Zhang
- The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Advanced Medical Technology Center, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
12
|
Najafi Z, Rahmanian-Devin P, Baradaran Rahimi V, Nokhodchi A, Askari VR. Challenges and opportunities of medicines for treating tendon inflammation and fibrosis: A comprehensive and mechanistic review. Fundam Clin Pharmacol 2024; 38:802-841. [PMID: 38468183 DOI: 10.1111/fcp.12999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/20/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Tendinopathy refers to conditions characterized by collagen degeneration within tendon tissue, accompanied by the proliferation of capillaries and arteries, resulting in reduced mechanical function, pain, and swelling. While inflammation in tendinopathy can play a role in preventing infection, uncontrolled inflammation can hinder tissue regeneration and lead to fibrosis and impaired movement. OBJECTIVES The inability to regulate inflammation poses a significant limitation in tendinopathy treatment. Therefore, an ideal treatment strategy should involve modulation of the inflammatory process while promoting tissue regeneration. METHODS The current review article was prepared by searching PubMed, Scopus, Web of Science, and Google Scholar databases. Several treatment approaches based on biomaterials have been developed. RESULTS This review examines various treatment methods utilizing small molecules, biological compounds, herbal medicine-inspired approaches, immunotherapy, gene therapy, cell-based therapy, tissue engineering, nanotechnology, and phototherapy. CONCLUSION These treatments work through mechanisms of action involving signaling pathways such as transforming growth factor-beta (TGF-β), mitogen-activated protein kinases (MAPKs), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), all of which contribute to the repair of injured tendons.
Collapse
Affiliation(s)
- Zohreh Najafi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Rahmanian-Devin
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Nokhodchi
- Lupin Pharmaceutical Research Center, 4006 NW 124th Ave., Coral Springs, Florida, Florida, 33065, USA
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK
| | - Vahid Reza Askari
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Timmer KB, Killian ML, Harley BAC. Paracrine signals influence patterns of fibrocartilage differentiation in a lyophilized gelatin hydrogel for applications in rotator cuff repair. Biomater Sci 2024; 12:4806-4822. [PMID: 39150417 PMCID: PMC11404831 DOI: 10.1039/d4bm00543k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Rotator cuff injuries present a clinical challenge for repair due to current limitations in functional regeneration of the native tendon-to-bone enthesis. A biomaterial that can regionally instruct unique tissue-specific phenotypes offers potential to promote enthesis repair. We have recently demonstrated the mechanical benefits of a stratified triphasic biomaterial made up of tendon- and bone-mimetic collagen scaffold compartments connected via a continuous hydrogel, and we now explore the potential of a biologically favorable enthesis hydrogel for this application. Here we report in vitro behavior of human mesenchymal stem cells (hMSCs) within thiolated gelatin (Gel-SH) hydrogels in response to chondrogenic stimuli as well as paracrine signals derived from MSC-seeded bone and tendon scaffold compartments. Chondrogenic differentiation media promoted upregulation of cartilage and entheseal fibrocartilage matrix markers COL2, COLX, and ACAN as well as the enthesis-associated transcription factors SCX, SOX9, and RUNX2 in hMSCs within Gel-SH. Similar effects were observed in response to TGF-β3 and BMP-4, enthesis-associated growth factors known to play a role in entheseal development and maintenance. Conditioned media generated by hMSCs seeded in tendon- and bone-mimetic collagen scaffolds influenced patterns of gene expression regarding enthesis-relevant growth factors, matrix markers, and tendon-to-bone transcription factors for hMSCs within the material. Together, these findings demonstrate that a Gel-SH hydrogel provides a permissive environment for enthesis tissue engineering and highlights the significance of cellular crosstalk between adjacent compartments within a spatially graded biomaterial.
Collapse
Affiliation(s)
- Kyle B Timmer
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.
| | - Megan L Killian
- Department of Orthopaedic Surgery, University of Michigan Ann Arbor, Ann Arbor, Michigan 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, Ann Arbor, Michigan 48109, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
14
|
Wada S, Ideno H, Nakashima K, Komatsu K, Demura N, Tomonari H, Kimura H, Tachibana M, Nifuji A. The histone H3K9 methyltransferase G9a regulates tendon formation during development. Sci Rep 2024; 14:20771. [PMID: 39237663 PMCID: PMC11377446 DOI: 10.1038/s41598-024-71570-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
G9a is a histone methyltransferase that catalyzes the methylation of histone 3 lysine 9 (H3K9), which is involved in the regulation of gene expression. We had previously reported that G9a is expressed in developing tendons in vivo and in vitro and that G9a-deficient tenocytes show impaired proliferation and differentiation in vitro. In this study, we investigated the functions of G9a in tendon development in vivo by using G9a conditional knockout (G9a cKO) mice. We crossed Sox9Cre/+ mice with G9afl/fl mice to generate G9afl/fl; Sox9Cre/+ mice. The G9a cKO mice showed hypoplastic tendon formation at 3 weeks of age. Bromodeoxyuridine labeling on embryonic day 16.5 (E16.5) revealed decreased cell proliferation in the tenocytes of G9a cKO mice. Immunohistochemical analysis revealed decreased expression levels of G9a and its substrate, H3K9me2, in the vertebral tendons of G9a cKO mice. The tendon tissue of the vertebrae and limbs of G9a cKO mice showed reduced expression of a tendon marker, tenomodulin (Tnmd), and col1a1 genes, suggesting that tenocyte differentiation was suppressed. Overexpression of G9a resulted in enhancement of Tnmd and col1a1 expression in tenocytes in vitro. These results suggest that G9a regulates the proliferation and differentiation of tendon progenitor cells during tendon development. Thus, our results suggest that G9a plays an essential role in tendon development.
Collapse
Affiliation(s)
- Satoshi Wada
- Department of Pharmacology, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa, 230-8501, Japan
- Department of Oral and Maxillofacial Surgery, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa, 230-8501, Japan
| | - Hisashi Ideno
- Department of Pharmacology, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa, 230-8501, Japan
| | - Kazuhisa Nakashima
- Department of Pharmacology, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa, 230-8501, Japan
| | - Koichiro Komatsu
- Department of Pharmacology, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa, 230-8501, Japan
| | - Noboru Demura
- Department of Oral and Maxillofacial Surgery, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiroshi Tomonari
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa, 230-8501, Japan
| | - Hiroshi Kimura
- Department of Biological Sciences, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8501, Japan
| | - Makoto Tachibana
- Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Akira Nifuji
- Department of Pharmacology, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa, 230-8501, Japan.
| |
Collapse
|
15
|
Liang C, Fan Z, Zhang Z, Wang P, Deng H, Tao J. Electrospinning technology: a promising approach for tendon-bone interface tissue engineering. RSC Adv 2024; 14:26077-26090. [PMID: 39161449 PMCID: PMC11332360 DOI: 10.1039/d4ra04043k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/29/2024] [Indexed: 08/21/2024] Open
Abstract
The regeneration of tendon-bone interface tissue has become a topic of great interest in recent years. However, the complex nature of this interface has posed challenges in finding suitable solutions. Tissue engineering, with its potential to improve clinical outcomes and play a crucial role in musculoskeletal function, has been increasingly explored for tendon-bone interface regeneration. This review focuses on the research advancements of electrospinning technology in interface tissue engineering. By utilizing electrospinning, researchers have been able to fabricate scaffolds with tailored properties to promote the regeneration and integration of tendon and bone tissues. The review discusses the unique structure and function of the tendon-bone interface, the mechanisms involved in its healing, and the limitations currently faced in achieving successful regeneration. Additionally, it highlights the potential of electrospinning technology in scaffold fabrication and its role in facilitating the development of functional and integrated tendon-bone interface tissues. Overall, this review provides valuable insights into the application of electrospinning technology for tendon-bone interface tissue engineering, emphasizing its significance in addressing the challenges associated with regeneration in this complex interface.
Collapse
Affiliation(s)
- Chengzhi Liang
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Zaiwei Fan
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Zirui Zhang
- Department of Rehabilitation Medicine, The 960th Hospital of the Chinese People's Liberation Army Jinan 250000 China
| | - Pinkai Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Hui Deng
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| | - Jun Tao
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University Nanchang Jiangxi 330000 China
| |
Collapse
|
16
|
Hirsinger E, Blavet C, Bonnin MA, Bellenger L, Gharsalli T, Duprez D. Limb connective tissue is organized in a continuum of promiscuous fibroblast identities during development. iScience 2024; 27:110305. [PMID: 39050702 PMCID: PMC11267076 DOI: 10.1016/j.isci.2024.110305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Connective tissue (CT), which includes tendon and muscle CT, plays critical roles in development, in particular as positional cue provider. Nonetheless, our understanding of fibroblast developmental programs is hampered because fibroblasts are highly heterogeneous and poorly characterized. Combining single-cell RNA-sequencing-based strategies including trajectory inference and in situ hybridization analyses, we address the diversity of fibroblasts and their developmental trajectories during chicken limb fetal development. We show that fibroblasts switch from a positional information to a lineage diversification program at the fetal period onset. Muscle CT and tendon are composed of several fibroblast populations that emerge asynchronously. Once the final muscle pattern is set, transcriptionally close populations are found in neighboring locations in limbs, prefiguring the adult fibroblast layers. We propose that the limb CT is organized in a continuum of promiscuous fibroblast identities, allowing for the robust and efficient connection of muscle to bone and skin.
Collapse
Affiliation(s)
- Estelle Hirsinger
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Cédrine Blavet
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Marie-Ange Bonnin
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Léa Bellenger
- Sorbonne Université, CNRS FR3631, Inserm U1156, Institut de Biologie Paris Seine (IBPS), ARTbio Bioinformatics Analysis Facility, Paris, Institut Français de Bioinformatique (IFB), 75005 Paris, France
| | - Tarek Gharsalli
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
- Inovarion, 75005 Paris, France
| | - Delphine Duprez
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| |
Collapse
|
17
|
Papalamprou A, Yu V, Jiang W, Sheyn J, Stefanovic T, Chen A, Castaneda C, Chavez M, Sheyn D. Single Cell Transcriptomics-Informed Induced Pluripotent Stem Cells Differentiation to Tenogenic Lineage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.10.536240. [PMID: 37090543 PMCID: PMC10120682 DOI: 10.1101/2023.04.10.536240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
During vertebrate embryogenesis, axial tendons develop from the paraxial mesoderm and differentiate through specific developmental stages to reach the syndetome stage. While the main roles of signaling pathways in the earlier stages of the differentiation have been well established, pathway nuances in syndetome specification from the sclerotome stage have yet to be explored. Here, we show stepwise differentiation of human iPSCs to the syndetome stage using chemically defined media and small molecules that were modified based on single cell RNA-sequencing and pathway analysis. We identified a significant population of branching off-target cells differentiating towards a neural phenotype overexpressing Wnt. Further transcriptomics post-addition of a WNT inhibitor at the somite stage and onwards revealed not only total removal of the neural off-target cells, but also increased syndetome induction efficiency. Fine-tuning tendon differentiation in vitro is essential to address the current challenges in developing a successful cell-based tendon therapy.
Collapse
Affiliation(s)
- Angela Papalamprou
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Victoria Yu
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Wensen Jiang
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Julia Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Tina Stefanovic
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Angel Chen
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Chloe Castaneda
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Melissa Chavez
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
18
|
Brooks EC, Han SJY, Bonatto Paese CL, Lewis AA, Aarnio-Peterson M, Brugmann SA. The ciliary protein C2cd3 is required for mandibular musculoskeletal tissue patterning. Differentiation 2024; 138:100782. [PMID: 38810379 PMCID: PMC11227401 DOI: 10.1016/j.diff.2024.100782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
The mandible is composed of several musculoskeletal tissues including bone, cartilage, and tendon that require precise patterning to ensure structural and functional integrity. Interestingly, most of these tissues are derived from one multipotent cell population called cranial neural crest cells (CNCCs). How CNCCs are properly instructed to differentiate into various tissue types remains nebulous. To better understand the mechanisms necessary for the patterning of mandibular musculoskeletal tissues we utilized the avian mutant talpid2 (ta2) which presents with several malformations of the facial skeleton including dysplastic tendons, mispatterned musculature, and bilateral ectopic cartilaginous processes extending off Meckel's cartilage. We found an ectopic epithelial BMP signaling domain in the ta2 mandibular prominence (MNP) that correlated with the subsequent expansion of SOX9+ cartilage precursors. These findings were validated with conditional murine models suggesting an evolutionarily conserved mechanism for CNCC-derived musculoskeletal patterning. Collectively, these data support a model in which cilia are required to define epithelial signal centers essential for proper musculoskeletal patterning of CNCC-derived mesenchyme.
Collapse
Affiliation(s)
- Evan C Brooks
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Simon J Y Han
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Christian Louis Bonatto Paese
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Amya A Lewis
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Megan Aarnio-Peterson
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA
| | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
19
|
Yambe S, Yoshimoto Y, Ikeda K, Maki K, Takimoto A, Tokuyama A, Higuchi S, Yu X, Uchibe K, Miura S, Watanabe H, Sakuma T, Yamamoto T, Tanimoto K, Kondoh G, Kasahara M, Mizoguchi T, Docheva D, Adachi T, Shukunami C. Sclerostin modulates mineralization degree and stiffness profile in the fibrocartilaginous enthesis for mechanical tissue integrity. Front Cell Dev Biol 2024; 12:1360041. [PMID: 38895158 PMCID: PMC11183276 DOI: 10.3389/fcell.2024.1360041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/19/2024] [Indexed: 06/21/2024] Open
Abstract
Fibrocartilaginous entheses consist of tendons, unmineralized and mineralized fibrocartilage, and subchondral bone, each exhibiting varying stiffness. Here we examined the functional role of sclerostin, expressed in mature mineralized fibrochondrocytes. Following rapid mineralization of unmineralized fibrocartilage and concurrent replacement of epiphyseal hyaline cartilage by bone, unmineralized fibrocartilage reexpanded after a decline in alkaline phosphatase activity at the mineralization front. Sclerostin was co-expressed with osteocalcin at the base of mineralized fibrocartilage adjacent to subchondral bone. In Scx-deficient mice with less mechanical loading due to defects of the Achilles tendon, sclerostin+ fibrochondrocyte count significantly decreased in the defective enthesis where chondrocyte maturation was markedly impaired in both fibrocartilage and hyaline cartilage. Loss of the Sost gene, encoding sclerostin, elevated mineral density in mineralized zones of fibrocartilaginous entheses. Atomic force microscopy analysis revealed increased fibrocartilage stiffness. These lines of evidence suggest that sclerostin in mature mineralized fibrochondrocytes acts as a modulator for mechanical tissue integrity of fibrocartilaginous entheses.
Collapse
Affiliation(s)
- Shinsei Yambe
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazutaka Ikeda
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Orthodontics and Craniofacial Developmental Biology, Applied Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koichiro Maki
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Aki Takimoto
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Shinnosuke Higuchi
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Xinyi Yu
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kenta Uchibe
- Department of Maxillofacial Anatomy and Neuroscience, Division of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigenori Miura
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tetsushi Sakuma
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Takashi Yamamoto
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Craniofacial Developmental Biology, Applied Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Taiji Adachi
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Division of Dental Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
20
|
Kitamura A, Yamamoto M, Hirouchi H, Watanabe G, Taniguchi S, Sekiya S, Ishizuka S, Jeong J, Higa K, Yamashita S, Abe S. Downregulation of SOX9 expression in developing entheses adjacent to intramembranous bone. PLoS One 2024; 19:e0301080. [PMID: 38728328 PMCID: PMC11086909 DOI: 10.1371/journal.pone.0301080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/08/2024] [Indexed: 05/12/2024] Open
Abstract
Entheses are classified into three types: fibrocartilaginous, fibrous, and periosteal insertions. However, the mechanism behind the development of fibrous entheses and periosteal insertions remains unclear. Since both entheses are part of the temporomandibular joint (TMJ), this study analyzes the TMJ entheses. Here, we show that SOX9 expression is negatively regulated during TMJ enthesis development, unlike fibrocartilage entheses which are modularly formed by SCX and SOX9 positive progenitors. The TMJ entheses was adjacent to the intramembranous bone rather than cartilage. SOX9 expression was diminished during TMJ enthesis development. To clarify the functional role of Sox9 in the development of TMJ entheses, we examined these structures in TMJ using Wnt1Cre;Sox9flox/+ reporter mice. Wnt1Cre;Sox9flox/+ mice showed enthesial deformation at the TMJ. Next, we also observed a diminished SOX9 expression area at the enthesis in contact with the clavicle's membranous bone portion, similar to the TMJ entheses. Together, these findings reveal that the timing of SOX9 expression varies with the ossification development mode.
Collapse
Affiliation(s)
- Asahi Kitamura
- Department of Removable Partial Prosthodontics, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| | - Masahito Yamamoto
- Division of Basic Medical Science, Department of Anatomy, Tokai University School of Medicine, Kanagawa, Japan
- Department of Anatomy, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| | - Hidetomo Hirouchi
- Department of Anatomy, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| | - Genji Watanabe
- Department of Anatomy, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| | | | - Sayo Sekiya
- Department of Anatomy, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| | - Satoshi Ishizuka
- Department of Pharmacology, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| | - Juhee Jeong
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States of America
| | - Kazunari Higa
- Ophthalmology/Cornea Center, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Shuichiro Yamashita
- Department of Removable Partial Prosthodontics, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
21
|
Steltzer SS, Abraham AC, Killian ML. Interfacial Tissue Regeneration with Bone. Curr Osteoporos Rep 2024; 22:290-298. [PMID: 38358401 PMCID: PMC11060924 DOI: 10.1007/s11914-024-00859-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE OF REVIEW Interfacial tissue exists throughout the body at cartilage-to-bone (osteochondral interface) and tendon-to-bone (enthesis) interfaces. Healing of interfacial tissues is a current challenge in regenerative approaches because the interface plays a critical role in stabilizing and distributing the mechanical stress between soft tissues (e.g., cartilage and tendon) and bone. The purpose of this review is to identify new directions in the field of interfacial tissue development and physiology that can guide future regenerative strategies for improving post-injury healing. RECENT FINDINGS Cues from interfacial tissue development may guide regeneration including biological cues such as cell phenotype and growth factor signaling; structural cues such as extracellular matrix (ECM) deposition, ECM, and cell alignment; and mechanical cues such as compression, tension, shear, and the stiffness of the cellular microenvironment. In this review, we explore new discoveries in the field of interfacial biology related to ECM remodeling, cellular metabolism, and fate. Based on emergent findings across multiple disciplines, we lay out a framework for future innovations in the design of engineered strategies for interface regeneration. Many of the key mechanisms essential for interfacial tissue development and adaptation have high potential for improving outcomes in the clinic.
Collapse
Affiliation(s)
- Stephanie S Steltzer
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Adam C Abraham
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Megan L Killian
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Suito H, Fujikawa K, Ohsako M. ENPP1 downregulation and FGF23 upregulation in growth-related calcification of the tibial tuberosity in rats. J Anat 2024; 244:333-342. [PMID: 37814911 PMCID: PMC10780148 DOI: 10.1111/joa.13958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/15/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023] Open
Abstract
During tibial tuberosity growth, superficial and deep portions can be observed; however, the deep portion is not observed after the growth period, as it develops into bone tissues. Calcification in vivo is known to be constitutively suppressed by ectonucleotide pyrophosphatase/phosphodiesterase-1 (ENPP1) but promoted by tissue-nonspecific alkaline phosphatase (TNAP). FGF23 promotes calcification of enthesis. Gene expression of FGF23 increased rapidly at 13W in this study. Therefore, the tibial tuberosity is speculated to develop via Enpp1 downregulation and Tnap upregulation; however, the understanding of these processes remains unclear. Hence, in the present study, we aimed to explore the age-related structural changes and underlying gene expression changes in the tibial tuberosity of rats. Male Wistar rats were divided into three groups (3-, 7-, and 13-week-old; eight each). The tibial tuberosity superficial and deep portions were clearly observed in 3- and 7-week-old rats, but the presence of the deep portion was not confirmed in 13-week-old rats. The extracellular matrix of hypertrophic chondrocytes was calcified. Furthermore, the Enpp1 expression was the highest in 3-week-old rats and decreased with growth. The TNAP expression did not differ significantly among the groups. The deep portion area was significantly lower in 3-week-old rats than in 7-week-old rats. Generally, the extracellular matrix of the immature chondrocytes is not calcified. Therefore, we speculated that the cartilaginous tibial tuberosity calcifies and ossifies with growth. The Enpp1 expression decreased with growth, whereas the Tnap expression remained unchanged. Thus, we surmise that the tibial tuberosity calcifies with growth and that this process involves Enpp1 downregulation and FGF23 upregulation. As Osgood-Schlatter disease is closely related to the calcification of the tibial tuberosity, these findings may help clarify the pathogenesis of this disease.
Collapse
Affiliation(s)
- Hirai Suito
- Graduate School of Human Life Design, Toyo University, Tokyo, Japan
- Japan Society for the Promotion of Science Research Fellowships DC, Tokyo, Japan
| | - Kaoru Fujikawa
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, Tokyo, Japan
| | - Masafumi Ohsako
- Graduate School of Health and Sports Science, Toyo University, Tokyo, Japan
| |
Collapse
|
23
|
Wang X, Gong W, Li R, Li L, Wang J. Preparation of genetically or chemically engineered exosomes and their therapeutic effects in bone regeneration and anti-inflammation. Front Bioeng Biotechnol 2024; 12:1329388. [PMID: 38314353 PMCID: PMC10834677 DOI: 10.3389/fbioe.2024.1329388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/11/2024] [Indexed: 02/06/2024] Open
Abstract
The treatment of bone or cartilage damage and inflammation-related diseases has been a long-standing research hotspot. Traditional treatments such as surgery and cell therapy have only displayed limited efficacy because they can't avoid potential deterioration and ensure cell activity. Recently, exosomes have become a favorable tool for various tissue reconstruction due to their abundant content of proteins, lipids, DNA, RNA and other substances, which can promote bone regeneration through osteogenesis, angiogenesis and inflammation modulation. Besides, exosomes are also promising delivery systems because of stability in the bloodstream, immune stealth capacity, intrinsic cell-targeting property and outstanding intracellular communication. Despite having great potential in therapeutic delivery, exosomes still show some limitations in clinical studies, such as inefficient targeting ability, low yield and unsatisfactory therapeutic effects. In order to overcome the shortcomings, increasing studies have prepared genetically or chemically engineered exosomes to improve their properties. This review focuses on different methods of preparing genetically or chemically engineered exosomes and the therapeutic effects of engineering exosomes in bone regeneration and anti-inflammation, thereby providing some references for future applications of engineering exosomes.
Collapse
Affiliation(s)
- Xinyue Wang
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Weitao Gong
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Rongrong Li
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Lin Li
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Jing Wang
- School of Stomatology, Lanzhou University, Lanzhou, China
- Clinical Research Center for Oral Diseases, Lanzhou, China
| |
Collapse
|
24
|
Liu X, Zhou M, Tan J, Ma L, Tang H, He G, Tao X, Guo L, Kang X, Tang K, Bian X. Inhibition of CX3CL1 by treadmill training prevents osteoclast-induced fibrocartilage complex resorption during TBI healing. Front Immunol 2024; 14:1295163. [PMID: 38283363 PMCID: PMC10811130 DOI: 10.3389/fimmu.2023.1295163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction The healing of tendon-bone injuries is very difficult, often resulting in poor biomechanical performance and unsatisfactory functional recovery. The tendon-bone insertion has a complex four distinct layers structure, and previous studies have often focused on promoting the regeneration of the fibrocartilage layer, neglecting the role of its bone end repair in tendon-bone healing. This study focuses on the role of treadmill training in promoting bone regeneration at the tendon-bone insertion and its related mechanisms. Methods After establishing the tendon-bone insertion injury model, the effect of treadmill training on tendon-bone healing was verified by Micro CT and HE staining; then the effect of CX3CL1 on osteoclast differentiation was verified by TRAP staining and cell culture; and finally the functional recovery of the mice was verified by biomechanical testing and behavioral test. Results Treadmill training suppresses the secretion of CX3CL1 and inhibits the differentiation of local osteoclasts after tendon-bone injury, ultimately reducing osteolysis and promoting tendon bone healing. Discussion Our research has found the interaction between treadmill training and the CX3CL1-C3CR1 axis, providing a certain theoretical basis for rehabilitation training.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Mei Zhou
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jindong Tan
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Lin Ma
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Hong Tang
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Gang He
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xu Tao
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Lin Guo
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xia Kang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Kanglai Tang
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xuting Bian
- Department of Sports Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
25
|
Brown ME, Puetzer JL. Enthesis maturation in engineered ligaments is differentially driven by loads that mimic slow growth elongation and rapid cyclic muscle movement. Acta Biomater 2023; 172:106-122. [PMID: 37839633 DOI: 10.1016/j.actbio.2023.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/17/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
Entheses are complex attachments that translate load between elastic-ligaments and stiff-bone via organizational and compositional gradients. Neither natural healing, repair, nor engineered replacements restore these gradients, contributing to high re-tear rates. Previously, we developed a culture system which guides ligament fibroblasts in high-density collagen gels to develop early postnatal-like entheses, however further maturation is needed. Mechanical cues, including slow growth elongation and cyclic muscle activity, are critical to enthesis development in vivo but these cues have not been widely explored in engineered entheses and their individual contribution to maturation is largely unknown. Our objective here was to investigate how slow stretch, mimicking ACL growth rates, and intermittent cyclic loading, mimicking muscle activity, individually drive enthesis maturation in our system so to shed light on the cues governing enthesis development, while further developing our tissue engineered replacements. Interestingly, we found these loads differentially drive organizational maturation, with slow stretch driving improvements in the interface/enthesis region, and cyclic load improving the ligament region. However, despite differentially affecting organization, both loads produced improvements to interface mechanics and zonal composition. This study provides insight into how mechanical cues differentially affect enthesis development, while producing some of the most organized engineered enthesis to date. STATEMENT OF SIGNIFICANCE: Entheses attach ligaments to bone and are critical to load transfer; however, entheses do not regenerate with repair or replacement, contributing to high re-tear rates. Mechanical cues are critical to enthesis development in vivo but their individual contribution to maturation is largely unknown and they have not been widely explored in engineered replacements. Here, using a novel culture system, we provide new insight into how slow stretch, mimicking ACL growth rates, and intermittent cyclic loading, mimicking muscle activity, differentially affect enthesis maturation in engineered ligament-to-bone tissues, ultimately producing some of the most organized entheses to date. This system is a promising platform to explore cues regulating enthesis formation so to produce functional engineered replacements and better drive regeneration following repair.
Collapse
Affiliation(s)
- M Ethan Brown
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, 23284, United States.
| |
Collapse
|
26
|
Yeung CYC, Garva R, Pickard A, Lu Y, Mallikarjun V, Swift J, Taylor SH, Rai J, Eyre DR, Chaturvedi M, Itoh Y, Meng QJ, Mauch C, Zigrino P, Kadler KE. Mmp14 is required for matrisome homeostasis and circadian rhythm in fibroblasts. Matrix Biol 2023; 124:8-22. [PMID: 37913834 DOI: 10.1016/j.matbio.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 10/03/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
The circadian clock in tendon regulates the daily rhythmic synthesis of collagen-I and the appearance and disappearance of small-diameter collagen fibrils in the extracellular matrix. How the fibrils are assembled and removed is not fully understood. Here, we first showed that the collagenase, membrane type I-matrix metalloproteinase (MT1-MMP, encoded by Mmp14), is regulated by the circadian clock in postnatal mouse tendon. Next, we generated tamoxifen-induced Col1a2-Cre-ERT2::Mmp14 KO mice (Mmp14 conditional knockout (CKO)). The CKO mice developed hind limb dorsiflexion and thickened tendons, which accumulated narrow-diameter collagen fibrils causing ultrastructural disorganization. Mass spectrometry of control tendons identified 1195 proteins of which 212 showed time-dependent abundance. In Mmp14 CKO mice 19 proteins had reversed temporal abundance and 176 proteins lost time dependency. Among these, the collagen crosslinking enzymes lysyl oxidase-like 1 (LOXL1) and lysyl hydroxylase 1 (LH1; encoded by Plod2) were elevated and had lost time-dependent regulation. High-pressure chromatography confirmed elevated levels of hydroxylysine aldehyde (pyridinoline) crosslinking of collagen in CKO tendons. As a result, collagen-I was refractory to extraction. We also showed that CRISPR-Cas9 deletion of Mmp14 from cultured fibroblasts resulted in loss of circadian clock rhythmicity of period 2 (PER2), and recombinant MT1-MMP was highly effective at cleaving soluble collagen-I but less effective at cleaving collagen pre-assembled into fibrils. In conclusion, our study shows that circadian clock-regulated Mmp14 controls the rhythmic synthesis of small diameter collagen fibrils, regulates collagen crosslinking, and its absence disrupts the circadian clock and matrisome in tendon fibroblasts.
Collapse
Affiliation(s)
- Ching-Yan Chloé Yeung
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK; Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Denmark.
| | - Richa Garva
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Adam Pickard
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Yinhui Lu
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Venkatesh Mallikarjun
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Joe Swift
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Susan H Taylor
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Jyoti Rai
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | - David R Eyre
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | | | - Yoshifumi Itoh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Qing-Jun Meng
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Cornelia Mauch
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Paola Zigrino
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Karl E Kadler
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
27
|
Donderwinkel I, Tuan RS, Cameron NR, Frith JE. A systematic investigation of the effects of TGF-β3 and mechanical stimulation on tenogenic differentiation of mesenchymal stromal cells in a poly(ethylene glycol)/gelatin-based hydrogel. J Orthop Translat 2023; 43:1-13. [PMID: 37929240 PMCID: PMC10622696 DOI: 10.1016/j.jot.2023.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/31/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023] Open
Abstract
Background High post-surgical failure rates following tendon injury generate high medical costs and poor patient recovery. Cell-based tendon tissue engineering has the potential to produce fully functional replacement tissue and provide new strategies to restore tendon function and healing. In this endeavour, the application of mesenchymal stromal cells (MSCs) encapsulated in biomaterial scaffolds has shown great promise. However, a consensus on optimal promotion of tenogenic differentiation of MSCs has yet to be reached, although growth factors and mechanical cues are generally acknowledged as important factors. Methods In this study, we prepared a hydrogel cell culture system consisting of methacrylated poly(d,l-lactic acid-ethylene glycol-d,l-lactic acid) (P(LA-EG-LA)) and gelatin methacrylate (GelMA) to encapsulate human bone marrow-derived MSCs (hBMSCs). We further systematically investigated the influence of static and intermittent cyclic uniaxial strain mechanical stimulation, in combination with transforming growth factor-β3 (TGF-β3) supplementation, on tenogenic differentiation of hBMSCs. Results Increased TGF-β3 concentration upregulated the tenogenic genes Scleraxis (SCX) and collagen type I (COL1A1) but showed no effects on tenascin-c (TNC) and collagen type III (COL3A1) expression. Mechanical stimulation had no observable effect on gene expression, but intermittent cyclic uniaxial strain stimulation improved matrix deposition. Together, these data provide new insights into how TGF-β3 and mechanical stimulation regulate MSC tenogenesis, with TGF-β3 promoting the expression of key tenogenic genes whilst mechanical stimulation aided matrix deposition in the engineered tissue. Furthermore, intermittent cyclic uniaxial strain at 3% elongation and 0.33 Hz for 1 h/day showed improved matrix effects compared to static strain. Conclusion Together, the most promising result for tenogenic differentiation of hBMSCs was identified as treatment with 5 ng/ml TGF-β3 under intermittent cyclic uniaxial strain (3% strain; 0.33 Hz; 1 h/day). This knowledge is of importance for the development of an improved protocol for tenogenic differentiation of MSCs and thereby for tendon tissue engineering. The translational potential of this article Tissue-engineered strategies for tendon repair require a consensus on the differentiation of mesenchymal stromal cells to tenocytes, which is currently lacking. This article provides a systematic investigation of two main tenogenic differentiation conditions to further development of a tenogenic differentiation protocol.
Collapse
Affiliation(s)
- Ilze Donderwinkel
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Neil R. Cameron
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- School of Engineering, University of Warwick, Coventry, CV4 7AL, United Kingdom
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - Jessica E. Frith
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
28
|
Fukuma Y, Tokunaga T, Tanimura S, Yoshimoto Y, Mashimo T, Kaneko T, Tian X, Ideo K, Yonemitsu R, Matsushita K, Sugimoto K, Yugami M, Hisanaga S, Nakamura T, Uehara Y, Masuda T, Shukunami C, Karasugi T, Miyamoto T. Potential function of Scx+/Sox9+ cells as progenitor cells in rotator cuff tear repair in rats. Biochem Biophys Res Commun 2023; 676:84-90. [PMID: 37499368 DOI: 10.1016/j.bbrc.2023.07.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Tendons and their attachment sites to bone, fibrocartilaginous tissues, have poor self-repair capacity when they rupture, and have risks of retear even after surgical repair. Thus, defining mechanisms underlying their repair is required in order to stimulate tendon repairing capacity. Here we used a rat surgical rotator cuff tear repair model and identified cells expressing the transcription factors Scleraxis (Scx) and SRY-box 9 (Sox9) as playing a crucial role in rotator cuff tendon-to-bone repair. Given the challenges of establishing stably reproducible models of surgical rotator cuff tear repair in mice, we newly established Scx-GFP transgenic rats in which Scx expression can be monitored by GFP. We observed tissue-specific GFP expression along tendons in developing ScxGFP transgenic rats and were able to successfully monitor tissue-specific Scx expression based on GFP signals. Among 3-, 6-, and 12-week-old ScxGFP rats, Scx+/Sox9+ cells were most abundant in 3-week-old rats near the site of humerus bone attachment to the rotator cuff tendon, while we observed significantly fewer cells in the same area in 6- or 12-week-old rats. We then applied a rotator cuff repair model using ScxGFP rats and observed the largest number of Scx+/Sox9+ cells at postoperative repair sites of 3-week-old relative to 6- or 12-week-old rats. Tendons attach to bone via fibrocartilaginous tissue, and cartilage-like tissue was seen at repair sites of 3-week-old but not 6- or 12-week-old rats during postoperative evaluation. Our findings suggest that Scx+/Sox9+ cells may function in rotator cuff repair, and that ScxGFP rats could serve as useful tools to develop therapies to promote rotator cuff repair by enabling analysis of these activities.
Collapse
Affiliation(s)
- Yuko Fukuma
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takuya Tokunaga
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| | - Shuntaro Tanimura
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yuki Yoshimoto
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan; Department of Molecular Biology and Biochemistry, Basic Life Sciences, Graduate School of Biomedical and Health Sciences, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Takehito Kaneko
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Xiao Tian
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Katsumasa Ideo
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Ryuji Yonemitsu
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kozo Matsushita
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kazuki Sugimoto
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Masaki Yugami
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Satoshi Hisanaga
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takayuki Nakamura
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yusuke Uehara
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Tetsuro Masuda
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Basic Life Sciences, Graduate School of Biomedical and Health Sciences, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Tatsuki Karasugi
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| | - Takeshi Miyamoto
- Department of Orthopedic Surgery, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan; Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
29
|
Chatterjee M, Evans MK, Bell R, Nguyen PK, Kamalitdinov TB, Korntner S, Kuo CK, Dyment NA, Andarawis-Puri N. Histological and immunohistochemical guide to tendon tissue. J Orthop Res 2023; 41:2114-2132. [PMID: 37321983 DOI: 10.1002/jor.25645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 06/02/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Tendons are unique dense connective tissues with discrete zones having specific structure and function. They are juxtaposed with other tissues (e.g., bone, muscle, and fat) with different compositional, structural, and mechanical properties. Additionally, tendon properties change drastically with growth and development, disease, aging, and injury. Consequently, there are unique challenges to performing high quality histological assessment of this tissue. To address this need, histological assessment was one of the breakout session topics at the 2022 Orthopaedic Research Society (ORS) Tendon Conference hosted at the University of Pennsylvania. The purpose of the breakout session was to discuss needs from members of the ORS Tendon Section related to histological procedures, data presentation, knowledge dissemination, and guidelines for future work. Therefore, this review provides a brief overview of the outcomes of this discussion and provides a set of guidelines, based on the perspectives from our laboratories, for histological assessment to assist researchers in their quest to utilize these techniques to enhance the outcomes and interpretations of their studies.
Collapse
Affiliation(s)
- Monideepa Chatterjee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Mary K Evans
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rebecca Bell
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| | - Phong K Nguyen
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| | - Timur B Kamalitdinov
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stefanie Korntner
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Catherine K Kuo
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Nathaniel A Dyment
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nelly Andarawis-Puri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
- Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
30
|
Zhang T, Wan L, Xiao H, Wang L, Hu J, Lu H. Single-cell RNA sequencing reveals cellular and molecular heterogeneity in fibrocartilaginous enthesis formation. eLife 2023; 12:e85873. [PMID: 37698466 PMCID: PMC10513478 DOI: 10.7554/elife.85873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 09/10/2023] [Indexed: 09/13/2023] Open
Abstract
The attachment site of the rotator cuff (RC) is a classic fibrocartilaginous enthesis, which is the junction between bone and tendon with typical characteristics of a fibrocartilage transition zone. Enthesis development has historically been studied with lineage tracing of individual genes selected a priori, which does not allow for the determination of single-cell landscapes yielding mature cell types and tissues. Here, in together with open-source GSE182997 datasets (three samples) provided by Fang et al., we applied Single-cell RNA sequencing (scRNA-seq) to delineate the comprehensive postnatal RC enthesis growth and the temporal atlas from as early as postnatal day 1 up to postnatal week 8. And, we furtherly performed single-cell spatial transcriptomic sequencing on postnatal day 1 mouse enthesis, in order to deconvolute bone-tendon junction (BTJ) chondrocytes onto spatial spots. In summary, we deciphered the cellular heterogeneity and the molecular dynamics during fibrocartilage differentiation. Combined with current spatial transcriptomic data, our results provide a transcriptional resource that will support future investigations of enthesis development at the mechanistic level and may shed light on the strategies for enhanced RC healing outcomes.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Liyang Wan
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Han Xiao
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Linfeng Wang
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Jianzhong Hu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South UniversityChangshaChina
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
31
|
Kamalitdinov TB, Fujino K, Keith Lang S, Jiang X, Madi R, Evans MK, Zgonis MH, Kuntz AF, Dyment NA. Targeting the hedgehog signaling pathway to improve tendon-to-bone integration. Osteoarthritis Cartilage 2023; 31:1202-1213. [PMID: 37146960 PMCID: PMC10524548 DOI: 10.1016/j.joca.2023.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/24/2023] [Accepted: 04/29/2023] [Indexed: 05/07/2023]
Abstract
OBJECTIVE While the role of hedgehog (Hh) signaling in promoting zonal fibrocartilage production during development is well-established, whether this pathway can be leveraged to improve tendon-to-bone repair in adults is unknown. Our objective was to genetically and pharmacologically stimulate the Hh pathway in cells that give rise to zonal fibrocartilaginous attachments to promote tendon-to-bone integration. DESIGN Hh signaling was stimulated genetically via constitutive Smo (SmoM2 construct) activation of bone marrow stromal cells or pharmacologically via systemic agonist delivery to mice following anterior cruciate ligament reconstruction (ACLR). To assess tunnel integration, we measured mineralized fibrocartilage (MFC) formation in these mice 28 days post-surgery and performed tunnel pullout testing. RESULTS Hh pathway-related genes increased in cells forming the zonal attachments in wild-type mice. Both genetic and pharmacologic stimulation of the Hh pathway increased MFC formation and integration strength 28 days post-surgery. We next conducted studies to define the role of Hh in specific stages of the tunnel integration process. We found Hh agonist treatment increased the proliferation of the progenitor pool in the first week post-surgery. Additionally, genetic stimulation led to continued MFC production in the later stages of the integration process. These results indicate that Hh signaling plays an important biphasic role in cell proliferation and differentiation towards fibrochondrocytes following ACLR. CONCLUSION This study reveals a biphasic role for Hh signaling during the tendon-to-bone integration process after ACLR. In addition, the Hh pathway is a promising therapeutic target to improve tendon-to-bone repair outcomes.
Collapse
Affiliation(s)
- Timur B Kamalitdinov
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Keitaro Fujino
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Osaka Medical and Pharmaceutical University, Takatsuki, Osaka Prefecture, Japan
| | - Sinaia Keith Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Xi Jiang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Rashad Madi
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Kate Evans
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Miltiadis H Zgonis
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew F Kuntz
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel A Dyment
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Wernlé KK, Sonnenfelt MA, Leek CC, Ganji E, Sullivan AL, Offutt C, Shuff J, Ornitz DM, Killian ML. Loss of Fgfr1 and Fgfr2 in Scleraxis-lineage cells leads to enlarged bone eminences and attachment cell death. Dev Dyn 2023; 252:1180-1188. [PMID: 37212424 PMCID: PMC10524747 DOI: 10.1002/dvdy.600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 05/23/2023] Open
Abstract
BACKGROUND Tendons and ligaments attach to bone are essential for joint mobility and stability in vertebrates. Tendon and ligament attachments (ie, entheses) are found at bony protrusions (ie, eminences), and the shape and size of these protrusions depend on both mechanical forces and cellular cues during growth. Tendon eminences also contribute to mechanical leverage for skeletal muscle. Fibroblast growth factor receptor (FGFR) signaling plays a critical role in bone development, and Fgfr1 and Fgfr2 are highly expressed in the perichondrium and periosteum of bone where entheses can be found. RESULTS AND CONCLUSIONS We used transgenic mice for combinatorial knockout of Fgfr1 and/or Fgfr2 in tendon/attachment progenitors (ScxCre) and measured eminence size and shape. Conditional deletion of both, but not individual, Fgfr1 and Fgfr2 in Scx progenitors led to enlarged eminences in the postnatal skeleton and shortening of long bones. In addition, Fgfr1/Fgfr2 double conditional knockout mice had more variation collagen fibril size in tendon, decreased tibial slope, and increased cell death at ligament attachments. These findings identify a role for FGFR signaling in regulating growth and maintenance of tendon/ligament attachments and the size and shape of bony eminences.
Collapse
Affiliation(s)
- Kendra K. Wernlé
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
- Institute of Anatomy, University of Zürich, Winterthurerstrasse 190, Zürich, Switzerland
| | - Michael A. Sonnenfelt
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
| | - Connor C. Leek
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
- Department of Orthopaedic Surgery, University of Michigan, 109 Zina Pitcher Pl, Ann Arbor, MI 48109
| | - Elahe Ganji
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
- Department of Orthopaedic Surgery, University of Michigan, 109 Zina Pitcher Pl, Ann Arbor, MI 48109
- Department of Mechanical Engineering, University of Delaware, 130 Academy St, Newark, DE 19716
| | - Anna Lia Sullivan
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
| | - Claudia Offutt
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
| | - Jordan Shuff
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, St Louis, Missouri, 63110
| | - Megan L. Killian
- Department of Biomedical Engineering, University of Delaware, 150 Academy St, Newark, Delaware, 19716
- Department of Orthopaedic Surgery, University of Michigan, 109 Zina Pitcher Pl, Ann Arbor, MI 48109
| |
Collapse
|
33
|
Subramanian A, Kanzaki LF, Schilling TF. Mechanical force regulates Sox9 expression at the developing enthesis. Development 2023; 150:dev201141. [PMID: 37497608 PMCID: PMC10445799 DOI: 10.1242/dev.201141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
Entheses transmit force from tendons and ligaments to the skeleton. Regional organization of enthesis extracellular matrix (ECM) generates differences in stiffness required for force transmission. Two key transcription factors co-expressed in entheseal tenocytes, scleraxis (Scx) and Sox9, directly control production of enthesis ECM components. Formation of embryonic craniofacial entheses in zebrafish coincides with onset of jaw movements, possibly in response to the force of muscle contraction. We show dynamic changes in scxa and sox9a mRNA levels in subsets of entheseal tenocytes that correlate with their roles in force transmission. We also show that transcription of a direct target of Scxa, Col1a, in enthesis ECM is regulated by the ratio of scxa to sox9a expression. Eliminating muscle contraction by paralyzing embryos during early stages of musculoskeletal differentiation alters relative levels of scxa and sox9a in entheses, primarily owing to increased sox9a expression. Force-dependent TGF-β (TGFβ) signaling is required to maintain this balance of scxa and sox9a expression. Thus, force from muscle contraction helps establish a balance of transcription factor expression that controls specialized ECM organization at the tendon enthesis and its ability to transmit force.
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Lauren F. Kanzaki
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
34
|
Watanabe G, Yamamoto M, Taniguchi S, Sugiyama Y, Hirouchi H, Ishizuka S, Kitamura K, Mizoguchi T, Takayama T, Hayashi K, Abe S. Chronological Changes in the Expression and Localization of Sox9 between Achilles Tendon Injury and Functional Recovery in Mice. Int J Mol Sci 2023; 24:11305. [PMID: 37511063 PMCID: PMC10379325 DOI: 10.3390/ijms241411305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Tendons help transmit forces from the skeletal muscles and bones. However, tendons have inferior regenerative ability compared to muscles. Despite studies on the regeneration of muscles and bone tissue, only a few have focused on tendinous tissue regeneration, especially tendon regeneration. Sex-determining region Y-box transcription factor 9 (Sox9) is an SRY-related transcription factor with a DNA-binding domain and is an important control factor for cartilage formation. Sox9 is critical to the early-to-middle stages of tendon development. However, how Sox9 participates in the healing process after tendon injury is unclear. We hypothesized that Sox9 is expressed in damaged tendons and is crucially involved in restoring tendon functions. We constructed a mouse model of an Achilles tendon injury by performing a 0.3 mm wide partial excision in the Achilles tendon of mice, and chronologically evaluated the function restoration and localization of the Sox9 expressed in the damaged sites. The results reveal that Sox9 was expressed simultaneously with the formation of the pre-structure of the epitenon, an essential part of the tendinous tissue, indicating that its expression is linked to the functional restoration of tendons. Lineage tracing for Sox9 expressed during tendon restoration revealed the tendon restoration involvement of cells that switched into Sox9-expressing cells after tendon injury. The stem cells involved in tendon regeneration may begin to express Sox9 after injury.
Collapse
Affiliation(s)
- Genji Watanabe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Masahito Yamamoto
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Shuichirou Taniguchi
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Yuki Sugiyama
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Hidetomo Hirouchi
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Satoshi Ishizuka
- Department of Pharmacology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kei Kitamura
- Department of Histology and Developmental Biology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Toshihide Mizoguchi
- Oral Health Science Center, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Takashi Takayama
- Department of Dentistry, The Jikei University School of Medicine, 3-19-18 Nishi-shinnbashi, Minato, Tokyo 105-8471, Japan
| | - Katsuhiko Hayashi
- Department of Dentistry, The Jikei University School of Medicine, 3-19-18 Nishi-shinnbashi, Minato, Tokyo 105-8471, Japan
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
35
|
Buss DJ, Rechav K, Reznikov N, McKee MD. Mineral tessellation in mouse enthesis fibrocartilage, Achilles tendon, and Hyp calcifying enthesopathy: A shared 3D mineralization pattern. Bone 2023:116818. [PMID: 37295663 DOI: 10.1016/j.bone.2023.116818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/17/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
The hallmark of enthesis architecture is the 3D compositional and structural gradient encompassing four tissue zones - tendon/ligament, uncalcified fibrocartilage, calcified fibrocartilage and bone. This functional gradient accommodates the large stiffness differential between calcified bone and uncalcified tendon/ligament. Here we analyze in 3D the organization of the mouse Achilles enthesis and mineralizing Achilles tendon in comparison to lamellar bone. We use correlative, multiscale high-resolution volume imaging methods including μCT with submicrometer resolution and FIB-SEM tomography (both with deep learning-based image segmentation), and TEM and SEM imaging, to describe ultrastructural features of physiologic, age-related and aberrant mineral patterning. We applied these approaches to murine wildtype (WT) Achilles enthesis tissues to describe in normal calcifying fibrocartilage a crossfibrillar mineral tessellation pattern similar to that observed in lamellar bone, but with greater variance in mineral tesselle morphology and size. We also examined Achilles enthesis structure in Hyp mice, a murine model for the inherited osteomalacic disease X-linked hypophosphatemia (XLH) with calcifying enthesopathy. In Achilles enthesis fibrocartilage of Hyp mice, we show defective crossfibrillar mineral tessellation similar to that which occurs in Hyp lamellar bone. At the cellular level in fibrocartilage, unlike in bone where enlarged osteocyte mineral lacunae are found as peri-osteocytic lesions, mineral lacunar volumes for fibrochondrocytes did not differ between WT and Hyp mice. While both WT and Hyp aged mice demonstrate Achilles tendon midsubstance ectopic mineralization, a consistently defective mineralization pattern was observed in Hyp mice. Strong immunostaining for osteopontin was observed at all mineralization sites examined in both WT and Hyp mice. Taken together, this new 3D ultrastructural information describes details of common mineralization trajectories for enthesis, tendon and bone, which in Hyp/XLH are defective.
Collapse
Affiliation(s)
- Daniel J Buss
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Katya Rechav
- Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Natalie Reznikov
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, Quebec, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Marc D McKee
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
36
|
Zou M, Wang J, Shao Z. Therapeutic Potential of Exosomes in Tendon and Tendon-Bone Healing: A Systematic Review of Preclinical Studies. J Funct Biomater 2023; 14:299. [PMID: 37367263 PMCID: PMC10299056 DOI: 10.3390/jfb14060299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Exosomes have been proven to play a positive role in tendon and tendon-bone healing. Here, we systematically review the literature to evaluate the efficacy of exosomes in tendon and tendon-bone healing. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a systematic and comprehensive review of the literature was performed on 21 January 2023. The electronic databases searched included Medline (through PubMed), Web of Science, Embase, Scopus, Cochrane Library and Ovid. In the end, a total of 1794 articles were systematically reviewed. Furthermore, a "snowball" search was also carried out. Finally, forty-six studies were included for analysis, with the total sample size being 1481 rats, 416 mice, 330 rabbits, 48 dogs, and 12 sheep. In these studies, exosomes promoted tendon and tendon-bone healing and displayed improved histological, biomechanical and morphological outcomes. Some studies also suggested the mechanism of exosomes in promoting tendon and tendon-bone healing, mainly through the following aspects: (1) suppressing inflammatory response and regulating macrophage polarization; (2) regulating gene expression, reshaping cell microenvironment and reconstructing extracellular matrix; (3) promoting angiogenesis. The risk of bias in the included studies was low on the whole. This systematic review provides evidence of the positive effect of exosomes on tendon and tendon-bone healing in preclinical studies. The unclear-to-low risk of bias highlights the significance of standardization of outcome reporting. It should be noted that the most suitable source, isolation methods, concentration and administration frequency of exosomes are still unknown. Additionally, few studies have used large animals as subjects. Further studies may be required on comparing the safety and efficacy of different treatment parameters in large animal models, which would be conducive to the design of clinical trials.
Collapse
Affiliation(s)
- Mingrui Zou
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (M.Z.); (J.W.)
- Beijing Key Laboratory of Sports Injuries, Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Jingzhou Wang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (M.Z.); (J.W.)
- Beijing Key Laboratory of Sports Injuries, Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Zhenxing Shao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (M.Z.); (J.W.)
- Beijing Key Laboratory of Sports Injuries, Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| |
Collapse
|
37
|
Sunadome K, Erickson AG, Kah D, Fabry B, Adori C, Kameneva P, Faure L, Kanatani S, Kaucka M, Dehnisch Ellström I, Tesarova M, Zikmund T, Kaiser J, Edwards S, Maki K, Adachi T, Yamamoto T, Fried K, Adameyko I. Directionality of developing skeletal muscles is set by mechanical forces. Nat Commun 2023; 14:3060. [PMID: 37244931 PMCID: PMC10224984 DOI: 10.1038/s41467-023-38647-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/05/2023] [Indexed: 05/29/2023] Open
Abstract
Formation of oriented myofibrils is a key event in musculoskeletal development. However, the mechanisms that drive myocyte orientation and fusion to control muscle directionality in adults remain enigmatic. Here, we demonstrate that the developing skeleton instructs the directional outgrowth of skeletal muscle and other soft tissues during limb and facial morphogenesis in zebrafish and mouse. Time-lapse live imaging reveals that during early craniofacial development, myoblasts condense into round clusters corresponding to future muscle groups. These clusters undergo oriented stretch and alignment during embryonic growth. Genetic perturbation of cartilage patterning or size disrupts the directionality and number of myofibrils in vivo. Laser ablation of musculoskeletal attachment points reveals tension imposed by cartilage expansion on the forming myofibers. Application of continuous tension using artificial attachment points, or stretchable membrane substrates, is sufficient to drive polarization of myocyte populations in vitro. Overall, this work outlines a biomechanical guidance mechanism that is potentially useful for engineering functional skeletal muscle.
Collapse
Affiliation(s)
- Kazunori Sunadome
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Alek G Erickson
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Delf Kah
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Csaba Adori
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
- Department of Molecular Biosciences, Wenner Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Polina Kameneva
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Shigeaki Kanatani
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Marketa Kaucka
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Str.2, 24306, Plön, Germany
| | | | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Steven Edwards
- KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden
| | - Koichiro Maki
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Taiji Adachi
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Takuya Yamamoto
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, 606-8501, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden.
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria.
| |
Collapse
|
38
|
Arostegui M, Scott RW, Underhill TM. Hic1 identifies a specialized mesenchymal progenitor population in the embryonic limb responsible for bone superstructure formation. Cell Rep 2023; 42:112325. [PMID: 37002923 DOI: 10.1016/j.celrep.2023.112325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/21/2022] [Accepted: 03/17/2023] [Indexed: 06/19/2023] Open
Abstract
The musculoskeletal system relies on the integration of multiple components with diverse physical properties, such as striated muscle, tendon, and bone, that enable locomotion and structural stability. This relies on the emergence of specialized, but poorly characterized, interfaces between these various elements during embryonic development. Within the appendicular skeleton, we show that a subset of mesenchymal progenitors (MPs), identified by Hic1, do not contribute to the primary cartilaginous anlagen but represent the MP population, whose progeny directly contribute to the interfaces that connect bone to tendon (entheses), tendon to muscle (myotendinous junctions), and the associated superstructures. Furthermore, deletion of Hic1 leads to skeletal defects reflective of deficient muscle-bone coupling and, consequently, perturbation of ambulation. Collectively, these findings show that Hic1 identifies a unique MP population that contributes to a secondary wave of bone sculpting critical to skeletal morphogenesis.
Collapse
Affiliation(s)
- Martin Arostegui
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - R Wilder Scott
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - T Michael Underhill
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
39
|
Fu S, Lan Y, Wang G, Bao D, Qin B, Zheng Q, Liu H, Wong VKW. External stimulation: A potential therapeutic strategy for tendon-bone healing. Front Bioeng Biotechnol 2023; 11:1150290. [PMID: 37064229 PMCID: PMC10102526 DOI: 10.3389/fbioe.2023.1150290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Injuries at the tendon-bone interface are very common in the field of sports medicine, and healing at the tendon-bone interface is complex. Injuries to the tendon-bone interface can seriously affect a patient’s quality of life, so it is essential to restore stability and promote healing of the tendon-bone interface. In addition to surgical treatment, the healing of tendons and bones can also be properly combined with extracorporeal stimulation therapy during the recovery process. In this review, we discuss the effects of extracorporeal shock waves (ESWs), low-intensity pulsed ultrasound (LIPUS), and mechanical stress on tendon-bone healing, focusing on the possible mechanisms of action of mechanical stress on tendon-bone healing in terms of transcription factors and biomolecules. The aim is to provide possible therapeutic approaches for subsequent clinical treatment.
Collapse
Affiliation(s)
- Shijie Fu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Yujian Lan
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Guoyou Wang
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Dingsu Bao
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Qin
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiu Zheng
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Huan Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
- *Correspondence: Huan Liu, ; Vincent Kam Wai Wong,
| | - Vincent Kam Wai Wong
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
- *Correspondence: Huan Liu, ; Vincent Kam Wai Wong,
| |
Collapse
|
40
|
Markman S, Zada M, David E, Giladi A, Amit I, Zelzer E. A single-cell census of mouse limb development identifies complex spatiotemporal dynamics of skeleton formation. Dev Cell 2023; 58:565-581.e4. [PMID: 36931270 DOI: 10.1016/j.devcel.2023.02.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 10/20/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023]
Abstract
Limb development has long served as a model system for coordinated spatial patterning of progenitor cells. Here, we identify a population of naive limb progenitors and show that they differentiate progressively to form the skeleton in a complex, non-consecutive, three-dimensional pattern. Single-cell RNA sequencing of the developing mouse forelimb identified three progenitor states: naive, proximal, and autopodial, as well as Msx1 as a marker for the naive progenitors. In vivo lineage tracing confirmed this role and localized the naive progenitors to the outer margin of the limb, along the anterior-posterior axis. Sequential pulse-chase experiments showed that the progressive transition of Msx1+ naive progenitors into proximal and autopodial progenitors coincides with their differentiation to Sox9+ chondroprogenitors, which occurs along all the forming skeletal segments. Indeed, tracking the spatiotemporal sequence of differentiation showed that the skeleton forms progressively in a complex pattern. These findings suggest an alternative model for limb skeleton development.
Collapse
Affiliation(s)
- Svetlana Markman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mor Zada
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
41
|
Insights into the Molecular and Hormonal Regulation of Complications of X-Linked Hypophosphatemia. ENDOCRINES 2023. [DOI: 10.3390/endocrines4010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
X-linked hypophosphatemia (XLH) is characterized by mutations in the PHEX gene, leading to elevated serum levels of FGF23, decreased production of 1,25 dihydroxyvitamin D3 (1,25D), and hypophosphatemia. Those affected with XLH manifest impaired growth and skeletal and dentoalveolar mineralization as well as increased mineralization of the tendon–bone attachment site (enthesopathy), all of which lead to decreased quality of life. Many molecular and murine studies have detailed the role of mineral ions and hormones in regulating complications of XLH, including how they modulate growth and growth plate maturation, bone mineralization and structure, osteocyte-mediated mineral matrix resorption and canalicular organization, and enthesopathy development. While these studies have provided insight into the molecular underpinnings of these skeletal processes, current therapies available for XLH do not fully prevent or treat these complications. Therefore, further investigations are needed to determine the molecular pathophysiology underlying the complications of XLH.
Collapse
|
42
|
Ganji E, Leek C, Duncan W, Patra D, Ornitz DM, Killian ML. Targeted deletion of Fgf9 in tendon disrupts mineralization of the developing enthesis. FASEB J 2023; 37:e22777. [PMID: 36734881 PMCID: PMC10108073 DOI: 10.1096/fj.202201614r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023]
Abstract
The enthesis is a transitional tissue between tendon and bone that matures postnatally. The development and maturation of the enthesis involve cellular processes likened to an arrested growth plate. In this study, we explored the role of fibroblast growth factor 9 (Fgf9), a known regulator of chondrogenesis and vascularization during bone development, on the structure and function of the postnatal enthesis. First, we confirmed spatial expression of Fgf9 in the tendon and enthesis using in situ hybridization. We then used Cre-lox recombinase to conditionally knockout Fgf9 in mouse tendon and enthesis (Scx-Cre) and characterized enthesis morphology as well as mechanical properties in Fgf9ScxCre and wild-type (WT) entheses. Fgf9ScxCre mice had smaller calcaneal and humeral apophyses, thinner cortical bone at the attachment, increased cellularity, and reduced failure load in mature entheses compared to WT littermates. During postnatal development, we found reduced chondrocyte hypertrophy and disrupted type X collagen (Col X) in Fgf9ScxCre entheses. These findings support that tendon-derived Fgf9 is important for functional development of the enthesis, including its postnatal mineralization. Our findings suggest the potential role of FGF signaling during enthesis development.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Orthopaedic Surgery, Michigan Medicine, Michigan, Ann Arbor, USA.,Department of Mechanical Engineering, University of Delaware, Delaware, Newark, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 61801, IL, Urbana, United States.,Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| | - Connor Leek
- Department of Orthopaedic Surgery, Michigan Medicine, Michigan, Ann Arbor, USA.,Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| | - William Duncan
- Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| | - Debabrata Patra
- Department of Developmental Biology, Washington University School of Medicine, Missouri, St Louis, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Missouri, St Louis, USA
| | - Megan L Killian
- Department of Orthopaedic Surgery, Michigan Medicine, Michigan, Ann Arbor, USA.,Department of Biomedical Engineering, University of Delaware, Delaware, Newark, USA
| |
Collapse
|
43
|
Ozone K, Minegishi Y, Takahata K, Takahashi H, Yoneno M, Hattori S, Xianglan L, Oka Y, Murata K, Kanemura N. Eccentric contraction-dominant exercise leads to molecular biological changes in enthesis and enthesopathy-like morphological changes. J Orthop Res 2023; 41:511-523. [PMID: 35716158 DOI: 10.1002/jor.25399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 06/02/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023]
Abstract
Entheses, which are tendon-to-bone attachment sites in the musculoskeletal system, play important roles in optimizing the mechanical stress and force transmitted from the muscle to the bone. Sports-related enthesopathy shows pathological features, including hyperplasia of the fibrocartilage (FC) region in the enthesis. The amount of exercise and type of muscle contraction during movement is involved in the pathogenesis of sports-related enthesopathy; however, the details of this condition are unclear. Here we examined the molecular pathways involved in the morphological changes of the muscle-tendon-enthesis complex and enthesis FC region in the supraspinatus muscle enthesis of mice under different exercise conditions. Following intervention, morphological changes in the muscle-tendon-enthesis complex were initiated in the eccentric contraction-dominant exercise group at 2 weeks, with activation of the transforming growth factor-β (TGFβ) superfamily pathway predicted by proteome and ingenuity pathway analyses. Histological and molecular biological analyses confirmed the activation of the TGFβ/bone morphogenetic protein (BMP)-Smad pathway. The concentric contraction-dominant exercise group showed no change in the morphology of the muscle-tendon-enthesis complex or activation of the TGFβ/BMP-Smad pathway, despite overuse exercise. Statement of Clinical Significance: These results suggest that eccentric contraction-dominant exercise induces sports-related enthesopathy-like morphological changes in the early stages as well as molecular biological changes, mainly in the transforming growth factor-β superfamily pathway in enthesis. Statement of Clinical Significance: These results suggest that eccentric contraction-dominant exercise induces sports-related enthesopathy-like morphological changes in the early stages as well as molecular biological changes, mainly in the transforming growth factor-β superfamily pathway in enthesis.
Collapse
Affiliation(s)
- Kaichi Ozone
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Koshigaya, Saitama, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yuki Minegishi
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Koshigaya, Saitama, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Kei Takahata
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Koshigaya, Saitama, Japan
| | - Haruna Takahashi
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Koshigaya, Saitama, Japan
| | - Moe Yoneno
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Koshigaya, Saitama, Japan
| | - Shinya Hattori
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Li Xianglan
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Yuichiro Oka
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Koshigaya, Saitama, Japan
| | - Kenji Murata
- Department of Physical Therapy, Health and Social Services, Saitama Prefectural University, Koshigaya, Saitama, Japan
| | - Naohiko Kanemura
- Department of Physical Therapy, Health and Social Services, Saitama Prefectural University, Koshigaya, Saitama, Japan
| |
Collapse
|
44
|
Mesenchymal stem cells and macrophages and their interactions in tendon-bone healing. J Orthop Translat 2023; 39:63-73. [PMID: 37188000 PMCID: PMC10175706 DOI: 10.1016/j.jot.2022.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023] Open
Abstract
Tendon-bone insertion injuries (TBI), such as anterior cruciate ligament (ACL) and rotator cuff injuries, are common degenerative or traumatic pathologies with a negative impact on the patient's daily life, and they cause huge economic losses every year. The healing process after an injury is complex and is dependent on the surrounding environment. Macrophages accumulate during the entire process of tendon and bone healing and their phenotypes progressively transform as they regenerate. As the "sensor and switch of the immune system", mesenchymal stem cells (MSCs) respond to the inflammatory environment and exert immunomodulatory effects during the tendon-bone healing process. When exposed to appropriate stimuli, they can differentiate into different tissues, including chondrocytes, osteocytes, and epithelial cells, promoting reconstruction of the complex transitional structure of the enthesis. It is well known that MSCs and macrophages communicate with each other during tissue repair. In this review, we discuss the roles of macrophages and MSCs in TBI injury and healing. Reciprocal interactions between MSCs and macrophages and some biological processes utilizing their mutual relations in tendon-bone healing are also described. Additionally, we discuss the limitations in our understanding of tendon-bone healing and propose feasible ways to exploit MSC-macrophage interplay to develop an effective therapeutic strategy for TBI injuries. The Translational potential of this article This paper reviewed the important functions of macrophages and mesenchymal stem cells in tendon-bone healing and described the reciprocal interactions between them during the healing process. By managing macrophage phenotypes, mesenchymal stem cells and the interactions between them, some possible novel therapies for tendon-bone injury may be proposed to promote tendon-bone healing after restoration surgery.
Collapse
|
45
|
Bautista CA, Srikumar A, Tichy ED, Qian G, Jiang X, Qin L, Mourkioti F, Dyment NA. CD206+ tendon resident macrophages and their potential crosstalk with fibroblasts and the ECM during tendon growth and maturation. Front Physiol 2023; 14:1122348. [PMID: 36909235 PMCID: PMC9992419 DOI: 10.3389/fphys.2023.1122348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023] Open
Abstract
Resident macrophages exist in a variety of tissues, including tendon, and play context-specific roles in their tissue of residence. In this study, we define the spatiotemporal distribution and phenotypic profile of tendon resident macrophages and their crosstalk with neighboring tendon fibroblasts and the extracellular matrix (ECM) during murine tendon development, growth, and homeostasis. Fluorescent imaging of cryosections revealed that F4/80+ tendon resident macrophages reside adjacent to Col1a1-CFP+ Scx-GFP+ fibroblasts within the tendon fascicle from embryonic development (E15.5) into adulthood (P56). Through flow cytometry and qPCR, we found that these tendon resident macrophages express several well-known macrophage markers, including Adgre1 (F4/80), Mrc1 (CD206), Lyve1, and Folr2, but not Ly-6C, and express the Csf1r-EGFP ("MacGreen") reporter. The proportion of Csf1r-EGFP+ resident macrophages in relation to the total cell number increases markedly during early postnatal growth, while the density of macrophages per mm2 remains constant during this same time frame. Interestingly, proliferation of resident macrophages is higher than adjacent fibroblasts, which likely contributes to this increase in macrophage proportion. The expression profile of tendon resident macrophages also changes with age, with increased pro-inflammatory and anti-inflammatory cytokine expression in P56 compared to P14 macrophages. In addition, the expression profile of limb tendon resident macrophages diverges from that of tail tendon resident macrophages, suggesting differential phenotypes across anatomically and functionally different tendons. As macrophages are known to communicate with adjacent fibroblasts in other tissues, we conducted ligand-receptor analysis and found potential two-way signaling between tendon fibroblasts and resident macrophages. Tendon fibroblasts express high levels of Csf1, which encodes macrophage colony stimulating factor (M-CSF) that acts on the CSF1 receptor (CSF1R) on macrophages. Importantly, Csf1r-expressing resident macrophages preferentially localize to Csf1-expressing fibroblasts, supporting the "nurturing scaffold" model for tendon macrophage patterning. Lastly, we found that tendon resident macrophages express high levels of ECM-related genes, including Mrc1 (mannose receptor), Lyve1 (hyaluronan receptor), Lair1 (type I collagen receptor), Ctss (elastase), and Mmp13 (collagenase), and internalize DQ Collagen in explant cultures. Overall, our study provides insights into the potential roles of tendon resident macrophages in regulating fibroblast phenotype and the ECM during tendon growth.
Collapse
Affiliation(s)
- Catherine A. Bautista
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of PA, Philadelphia, PA, United States
| | - Anjana Srikumar
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Elisia D. Tichy
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Grace Qian
- Department of Bioengineering, School of Engineering and Applied Science, University of PA, Philadelphia, PA, United States
| | - Xi Jiang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Ling Qin
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Foteini Mourkioti
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PA, Philadelphia, PA, United States
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of PA, Philadelphia, PA, United States
| | - Nathaniel A. Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of PA, Philadelphia, PA, United States
| |
Collapse
|
46
|
Hasan MR, Koskenranta A, Alakurtti K, Takatalo M, Rice DP. RAB23 regulates musculoskeletal development and patterning. Front Cell Dev Biol 2023; 11:1049131. [PMID: 36910145 PMCID: PMC9995984 DOI: 10.3389/fcell.2023.1049131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
RAB23 is a small GTPase which functions at the plasma membrane to regulate growth factor signaling. Mutations in RAB23 cause Carpenter syndrome, a condition that affects normal organogenesis and patterning. In this study, we investigate the role of RAB23 in musculoskeletal development and show that it is required for patella bone formation and for the maintenance of tendon progenitors. The patella is the largest sesamoid bone in mammals and plays a critical role during movement by providing structural and mechanical support to the knee. Rab23 -/- mice fail to form a patella and normal knee joint. The patella is formed from Sox9 and scleraxis (Scx) double-positive chondroprogenitor cells. We show that RAB23 is required for the specification of SOX9 and scleraxis double-positive patella chondroprogenitors during the formation of patella anlagen and the subsequent establishment of patellofemoral joint. We find that scleraxis and SOX9 expression are disrupted in Rab23 -/- mice, and as a result, development of the quadriceps tendons, cruciate ligaments, patella tendons, and entheses is either abnormal or lost. TGFβ-BMP signaling is known to regulate patella initiation and patella progenitor differentiation and growth. We find that the expression of TGFβR2, BMPR1, BMP4, and pSmad are barely detectable in the future patella site and in the rudimentary tendons and ligaments around the patellofemoral joint in Rab23 -/- mice. Also, we show that GLI1, SOX9, and scleraxis, which regulate entheses establishment and maturation, are weakly expressed in Rab23 -/- mice. Further analysis of the skeletal phenotype of Rab23 -/- mice showed a close resemblance to that of Tgfβ2 -/- mice, highlighting a possible role for RAB23 in regulating TGFβ superfamily signaling.
Collapse
Affiliation(s)
- Md Rakibul Hasan
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Anna Koskenranta
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Kirsi Alakurtti
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Maarit Takatalo
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - David P Rice
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
47
|
Anthwal N, Tucker AS. Evolution and development of the mammalian jaw joint: Making a novel structure. Evol Dev 2023; 25:3-14. [PMID: 36504442 PMCID: PMC10078425 DOI: 10.1111/ede.12426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/08/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022]
Abstract
A jaw joint between the squamosal and dentary is a defining feature of mammals and is referred to as the temporomandibular joint (TMJ) in humans. Driven by changes in dentition and jaw musculature, this new joint evolved early in the mammalian ancestral lineage and permitted the transference of the ancestral jaw joint into the middle ear. The fossil record demonstrates the steps in the cynodont lineage that led to the acquisition of the TMJ, including the expansion of the dentary bone, formation of the coronoid process, and initial contact between the dentary and squamosal. From a developmental perspective, the components of the TMJ form through tissue interactions of muscle and skeletal elements, as well as through interaction between the jaw and the cranial base, with the signals involved in these interactions being both biomechanical and biochemical. In this review, we discuss the development of the TMJ in an evolutionary context. We describe the evolution of the TMJ in the fossil record and the development of the TMJ in embryonic development. We address the formation of key elements of the TMJ and how knowledge from developmental biology can inform our understanding of TMJ evolution.
Collapse
Affiliation(s)
- Neal Anthwal
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentisry, Oral and Craniofacial Sciences, London, UK
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentisry, Oral and Craniofacial Sciences, London, UK
| |
Collapse
|
48
|
Istiaq A, Ohta K. A review on Tsukushi: mammalian development, disorders, and therapy. J Cell Commun Signal 2022; 16:505-513. [PMID: 35233735 PMCID: PMC9733752 DOI: 10.1007/s12079-022-00669-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Tsukushi (TSK), a leucine-rich peptidoglycan in the extracellular compartment, mediates multiple signaling pathways that are critical for development and metabolism. TSK regulates signaling pathways that eventually control cellular communication, proliferation, and cell fate determination. Research on TSK has become more sophisticated in recent years, illustrating its involvement in the physiology and pathophysiology of neural, genetic, and metabolic diseases. In a recent study, we showed that TSK therapy reversed the pathophysiological abnormalities of the hydrocephalic (a neurological disorder) brain in mice. This review summarizes the roles of TSK in key signaling processes in the mammalian development, disorders, and evaluating its possible therapeutic and diagnostic potential.
Collapse
Affiliation(s)
- Arif Istiaq
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, 819-0395 Fukuoka, Japan ,Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, 860-8555 Kumamoto, Japan ,HIGO Program, Kumamoto University, 860-8555 Kumamoto, Japan
| | - Kunimasa Ohta
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, 819-0395 Fukuoka, Japan
| |
Collapse
|
49
|
Fang F, Xiao Y, Zelzer E, Leong KW, Thomopoulos S. A mineralizing pool of Gli1-expressing progenitors builds the tendon enthesis and demonstrates therapeutic potential. Cell Stem Cell 2022; 29:1669-1684.e6. [PMID: 36459968 PMCID: PMC10422080 DOI: 10.1016/j.stem.2022.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/27/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
The enthesis, a fibrocartilaginous transition between tendon and bone, is necessary for force transfer from muscle to bone to produce joint motion. The enthesis is prone to injury due to mechanical demands, and it cannot regenerate. A better understanding of how the enthesis develops will lead to more effective therapies to prevent pathology and promote regeneration. Here, we used single-cell RNA sequencing to define the developmental transcriptome of the mouse entheses over postnatal stages. Six resident cell types, including enthesis progenitors and mineralizing chondrocytes, were identified along with their transcription factor regulons and temporal regulation. Following the prior discovery of the necessity of Gli1-lineage cells for mouse enthesis development and healing, we then examined their transcriptomes at single-cell resolution and demonstrated clonogenicity and multipotency of the Gli1-expressing progenitors. Transplantation of Gli1-lineage cells to mouse enthesis injuries improved healing, demonstrating their therapeutic potential for enthesis regeneration.
Collapse
Affiliation(s)
- Fei Fang
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA; Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
50
|
Liu C, Zhou N, Li N, Xu T, Chen X, Zhou H, Xie A, Liu H, Zhu L, Wang S, Xiao J. Disrupted tenogenesis in masseter as a potential cause of micrognathia. Int J Oral Sci 2022; 14:50. [PMID: 36257937 PMCID: PMC9579150 DOI: 10.1038/s41368-022-00196-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/09/2022] Open
Abstract
Micrognathia is a severe craniofacial deformity affecting appearance and survival. Previous studies revealed that multiple factors involved in the osteogenesis of mandibular bone have contributed to micrognathia, but concerned little on factors other than osteogenesis. In the current study, we found that ectopic activation of Fgf8 by Osr2-cre in the presumptive mesenchyme for masseter tendon in mice led to micrognathia, masseter regression, and the disrupted patterning and differentiation of masseter tendon. Since Myf5-cre;Rosa26R-Fgf8 mice exhibited the normal masseter and mandibular bone, the possibility that the micrognathia and masseter regression resulted directly from the over-expressed Fgf8 was excluded. Further investigation disclosed that a series of chondrogenic markers were ectopically activated in the developing Osr2-cre;Rosa26R-Fgf8 masseter tendon, while the mechanical sensing in the masseter and mandibular bone was obviously reduced. Thus, it suggested that the micrognathia in Osr2-cre;Rosa26R-Fgf8 mice resulted secondarily from the reduced mechanical force transmitted to mandibular bone. Consistently, when tenogenic or myogenic components were deleted from the developing mandibles, both the micrognathia and masseter degeneration took place with the decreased mechanical sensing in mandibular bone, which verified that the loss of mechanical force transmitted by masseter tendon could result in micrognathia. Furthermore, it appeared that the micrognathia resulting from the disrupted tenogenesis was attributed to the impaired osteogenic specification, instead of the differentiation in the periosteal progenitors. Our findings disclose a novel mechanism for mandibular morphogenesis, and shed light on the prevention and treatment for micrognathia.
Collapse
Affiliation(s)
- Chao Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Nan Zhou
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Nan Li
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Tian Xu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Xiaoyan Chen
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Hailing Zhou
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Ailun Xie
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Han Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Lei Zhu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Songlin Wang
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China. .,Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
| | - Jing Xiao
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China. .,Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
| |
Collapse
|