1
|
Shang X, Chen C, Zhou J, Yang Y, Qu J. Muscle loading and endochondral ossification are involved in the regeneration of a fibrocartilaginous enthesis during tendon to bone healing in rabbits. BMC Musculoskelet Disord 2025; 26:277. [PMID: 40108557 PMCID: PMC11921697 DOI: 10.1186/s12891-025-08508-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE The purposes of the present study are to investigate the effects of reduced muscle loading by prolonged immobilization on the regeneration of fibrocartilaginous enthesis through endochondral ossification in rabbits. METHODS Forty-eight rabbits underwent standard partial patellectomy were randomly divided into the control group and the prolonged immobilization (PIM) group. The immobilized cast was only maintained for the first 4 weeks in the control group, while for the first 12 weeks or until euthanization in the PIM group. The Patella-patella tendon complexes were harvested for Micro-CT and histology at week 6, 12 and 18. RESULTS There was significantly lower bone volume in the PIM group than the control group at week 12, but not at week 6 and 18. At week 6, new bone was formed at the osteotomy site of the residual patella through endochondral ossification. At week 12, the chondrocytes in the tendon to bone interface were ordered and arranged in longitudinal rows separated by collagen fibres in the control group. While there were no visible fibers running continuously from tendon into bone in the PIM group. At week 18, a nearly normal fibrocartilaginous enthesis were regenerated in the control group. A similar fibrocartilaginous enthesis were also formed at the tendon to bone interface in the PIM group, but the four zones were not as distinct as that in the control group. CONCLUSION Muscle loading and endochondral ossification are involved in the regeneration of a fibrocartilaginous enthesis during tendon to bone healing in this partial patellectomy model.
Collapse
Affiliation(s)
- Xiaoke Shang
- Department of Orthopedic Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Can Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Jiefu Zhou
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Yang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Jin Qu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Takahashi M, Shibata S, Shibui T, Takechi M, Irie K. Comparison of the structural features between chicken quadratomandibular joint and rat temporomandibular joint with reference to ligament and articular meniscus/disc. J Oral Biosci 2025; 67:100623. [PMID: 39894251 DOI: 10.1016/j.job.2025.100623] [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: 08/18/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVES The new jaw joint of mammals evolved between the squamosal and the dentary. We investigated the structural features of the chicken quadratomandibular joint (QMJ) (primary jaw joint) and the rat temporomandibular joint (TMJ) (new jaw joint) to determine whether these structures reflect their putative movements from macroscopic and histological perspectives. METHODS Chicken QMJ and rat TMJ were dissected, and their joint apparatuses, including ligaments and meniscus/disc, were analyzed macroscopically. Microscopic analysis, incorporating immunohistochemistry and in situ hybridization, was conducted to investigate protein localization and gene expression within the extracellular matrix of cartilage and bone. RESULTS The chicken QMJ contained the jugomandibular ligament, running laterally and posteriorly to the QMJ, tightly bound to the articular meniscus. These features suggest a role in supporting and controlling quadrate bone movements. The rat TMJ lacked a lateral ligament, and the mandibular head had an elongated oval shape along the anterior-posterior axis. This morphology indicates that lateral mandibular movements are rare, with jaw motion predominantly occurring in the anterior-posterior direction. The chicken QMJ meniscus contained distinct cartilaginous tissues, whereas the rat TMJ articular disc acquired cartilaginous characteristics after occlusion was established (5 weeks postnatally), suggesting that the meniscus/disc properties are closely associated with mastication. CONCLUSIONS The structural features of chicken and rat jaw joints, including their joint apparatuses, appropriately reflect their respective functions, such as movement dynamics and resistance to mastication pressure.
Collapse
Affiliation(s)
- Masami Takahashi
- Department of Anatomy, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Shunichi Shibata
- Department of Anatomy, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan.
| | - Toru Shibui
- Department of Anatomy, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Masaki Takechi
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuharu Irie
- Department of Anatomy, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| |
Collapse
|
3
|
DiStefano MS, Weiss SN, Nuss CA, Betts RL, Han B, Kuntz AF, Soslowsky LJ. Mature murine supraspinatus tendons demonstrate regional differences in multiscale structure, function and gene expression. PLoS One 2025; 20:e0318809. [PMID: 39977400 PMCID: PMC11841869 DOI: 10.1371/journal.pone.0318809] [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: 11/07/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
The hierarchical structure of tendon dictates its ability to effectively transmit loads from muscle to bone. Tendon- and site-specific differences in mechanical loading result in the establishment and remodeling of structure, as well as associated changes in composition throughout development and healing. Previous work has demonstrated region-specific differences in the response of collagen fibrils to mechanical loading within the insertion region and midsubstance regions of mouse supraspinatus tendons using atomic force microscopy. However, multiscale structure, function, and gene expression differences between the insertion and midsubstance of the supraspinatus tendon have not yet been linked together in a comprehensive study. Therefore, the purpose of this study was to elucidate site-specific hierarchical structure, function, and gene expression differences in mouse supraspinatus tendons. Supraspinatus tendons from day 150 wild-type C57BL/6 mice were harvested for regional mechanics, histology, transmission electron microscopy (TEM), and quantitative polymerase chain reaction (qPCR). Mechanical testing revealed that the midsubstance region demonstrated a greater modulus and increased collagen fiber realignment compared to the insertion region. Histological scoring demonstrated greater cellularity and more rounded cells in the insertion region. TEM analysis showed differences in collagen fibril diameter distributions between the two regions, with a shift towards smaller diameters observed at the insertion region. Gene expression analysis identified several genes that were differentially expressed between regions, with principal component analysis revealing distinct clustering based on region. These findings provide insight into the regional heterogeneity of the supraspinatus tendon and underscore the importance of considering these differences in the context of tendon injury and repair, contributing to a better understanding of tendon structure-function and guiding future studies aimed at elucidating the mechanisms underlying tendon pathology.
Collapse
Affiliation(s)
- Michael S. DiStefano
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Stephanie N. Weiss
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Courtney A. Nuss
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rebecca L. Betts
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Biao Han
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrew F. Kuntz
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Louis J. Soslowsky
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
4
|
Garau Paganella L, Badolato A, Labouesse C, Fischer G, Sänger CS, Kourouklis A, Giampietro C, Werner S, Mazza E, Tibbitt MW. Variations in fluid chemical potential induce fibroblast mechano-response in 3D hydrogels. BIOMATERIALS ADVANCES 2024; 163:213933. [PMID: 38972277 DOI: 10.1016/j.bioadv.2024.213933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Mechanical deformation of skin creates variations in fluid chemical potential, leading to local changes in hydrostatic and osmotic pressure, whose effects on mechanobiology remain poorly understood. To study these effects, we investigate the specific influences of hydrostatic and osmotic pressure on primary human dermal fibroblasts in three-dimensional hydrogel culture models. Cyclic hydrostatic pressure and hyperosmotic stress enhanced the percentage of cells expressing the proliferation marker Ki67 in both collagen and PEG-based hydrogels. Osmotic pressure also activated the p38 MAPK stress response pathway and increased the expression of the osmoresponsive genes PRSS35 and NFAT5. When cells were cultured in two-dimension (2D), no change in proliferation was observed with either hydrostatic or osmotic pressure. Furthermore, basal, and osmotic pressure-induced expression of osmoresponsive genes differed in 2D culture versus 3D hydrogels, highlighting the role of dimensionality in skin cell mechanotransduction and stressing the importance of 3D tissue-like models that better replicate in vivo conditions. Overall, these results indicate that fluid chemical potential changes affect dermal fibroblast mechanobiology, which has implications for skin function and for tissue regeneration strategies.
Collapse
Affiliation(s)
- Lorenza Garau Paganella
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland; Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Asia Badolato
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Céline Labouesse
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Gabriel Fischer
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Catharina S Sänger
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Andreas Kourouklis
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Costanza Giampietro
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland; EMPA, Swiss Federal Laboratories for Material Science and Technologies, Dubendorf, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Edoardo Mazza
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland; EMPA, Swiss Federal Laboratories for Material Science and Technologies, Dubendorf, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Villeneuve C, Hashmi A, Ylivinkka I, Lawson-Keister E, Miroshnikova YA, Pérez-González C, Myllymäki SM, Bertillot F, Yadav B, Zhang T, Matic Vignjevic D, Mikkola ML, Manning ML, Wickström SA. Mechanical forces across compartments coordinate cell shape and fate transitions to generate tissue architecture. Nat Cell Biol 2024; 26:207-218. [PMID: 38302719 PMCID: PMC10866703 DOI: 10.1038/s41556-023-01332-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/08/2023] [Indexed: 02/03/2024]
Abstract
Morphogenesis and cell state transitions must be coordinated in time and space to produce a functional tissue. An excellent paradigm to understand the coupling of these processes is mammalian hair follicle development, which is initiated by the formation of an epithelial invagination-termed placode-that coincides with the emergence of a designated hair follicle stem cell population. The mechanisms directing the deformation of the epithelium, cell state transitions and physical compartmentalization of the placode are unknown. Here we identify a key role for coordinated mechanical forces stemming from contractile, proliferative and proteolytic activities across the epithelial and mesenchymal compartments in generating the placode structure. A ring of fibroblast cells gradually wraps around the placode cells to generate centripetal contractile forces, which, in collaboration with polarized epithelial myosin activity, promote elongation and local tissue thickening. These mechanical stresses further enhance compartmentalization of Sox9 expression to promote stem cell positioning. Subsequently, proteolytic remodelling locally softens the basement membrane to facilitate a release of pressure on the placode, enabling localized cell divisions, tissue fluidification and epithelial invagination into the underlying mesenchyme. Together, our experiments and modelling identify dynamic cell shape transformations and tissue-scale mechanical cooperation as key factors for orchestrating organ formation.
Collapse
Affiliation(s)
- Clémentine Villeneuve
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Ali Hashmi
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Irene Ylivinkka
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Yekaterina A Miroshnikova
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Carlos Pérez-González
- Cell Biology and Cancer Unit, Institut Curie, PSL Research University, CNRS, Paris, France
| | - Satu-Marja Myllymäki
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Fabien Bertillot
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Bhagwan Yadav
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tao Zhang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | | | - Marja L Mikkola
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - M Lisa Manning
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY, USA.
| | - Sara A Wickström
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Vágó J, Takács R, Kovács P, Hajdú T, van der Veen DR, Matta C. Combining biomechanical stimulation and chronobiology: a novel approach for augmented chondrogenesis? Front Bioeng Biotechnol 2023; 11:1232465. [PMID: 37456723 PMCID: PMC10349586 DOI: 10.3389/fbioe.2023.1232465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The unique structure and composition of articular cartilage is critical for its physiological function. However, this architecture may get disrupted by degeneration or trauma. Due to the low intrinsic regeneration properties of the tissue, the healing response is generally poor. Low-grade inflammation in patients with osteoarthritis advances cartilage degradation, resulting in pain, immobility, and reduced quality of life. Generating neocartilage using advanced tissue engineering approaches may address these limitations. The biocompatible microenvironment that is suitable for cartilage regeneration may not only rely on cells and scaffolds, but also on the spatial and temporal features of biomechanics. Cell-autonomous biological clocks that generate circadian rhythms in chondrocytes are generally accepted to be indispensable for normal cartilage homeostasis. While the molecular details of the circadian clockwork are increasingly well understood at the cellular level, the mechanisms that enable clock entrainment by biomechanical signals, which are highly relevant in cartilage, are still largely unknown. This narrative review outlines the role of the biomechanical microenvironment to advance cartilage tissue engineering via entraining the molecular circadian clockwork, and highlights how application of this concept may enhance the development and successful translation of biomechanically relevant tissue engineering interventions.
Collapse
Affiliation(s)
- Judit Vágó
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Roland Takács
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Tibor Hajdú
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Daan R. van der Veen
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Csaba Matta
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
7
|
Zhou Z, Wang W, Wang J, Wang H, Xia Y, Zhang W, Lai Y, Lin X, Huang Y, Zou X, Stoddart MJ, Li Z, Tian W, Liu S, Wu X, Gao M, Li J, Yang L, Chen D. Function-oriented design: A novel strategy for advanced biomedical materials. JOURNAL OF MATERIALS SCIENCE & TECHNOLOGY 2023; 145:197-209. [DOI: 10.1016/j.jmst.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
|
8
|
Chondromodulin is necessary for cartilage callus distraction in mice. PLoS One 2023; 18:e0280634. [PMID: 36795722 PMCID: PMC9934371 DOI: 10.1371/journal.pone.0280634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/05/2023] [Indexed: 02/17/2023] Open
Abstract
Chondromodulin (Cnmd) is a glycoprotein known to stimulate chondrocyte growth. We examined in this study the expression and functional role of Cnmd during distraction osteogenesis that is modulated by mechanical forces. The right tibiae of the mice were separated by osteotomy and subjected to slow progressive distraction using an external fixator. In situ hybridization and immunohistochemical analyses of the lengthened segment revealed that Cnmd mRNA and its protein in wild-type mice were localized in the cartilage callus, which was initially generated in the lag phase and was lengthened gradually during the distraction phase. In Cnmd null (Cnmd-/-) mice, less cartilage callus was observed, and the distraction gap was filled by fibrous tissues. Additionally, radiological and histological investigations demonstrated delayed bone consolidation and remodeling of the lengthened segment in Cnmd-/- mice. Eventually, Cnmd deficiency caused a one-week delay in the peak expression of VEGF, MMP2, and MMP9 genes and the subsequent angiogenesis and osteoclastogenesis. We conclude that Cnmd is necessary for cartilage callus distraction.
Collapse
|
9
|
Putra VDL, Kilian KA, Knothe Tate ML. Biomechanical, biophysical and biochemical modulators of cytoskeletal remodelling and emergent stem cell lineage commitment. Commun Biol 2023; 6:75. [PMID: 36658332 PMCID: PMC9852586 DOI: 10.1038/s42003-022-04320-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 11/30/2022] [Indexed: 01/20/2023] Open
Abstract
Across complex, multi-time and -length scale biological systems, redundancy confers robustness and resilience, enabling adaptation and increasing survival under dynamic environmental conditions; this review addresses ubiquitous effects of cytoskeletal remodelling, triggered by biomechanical, biophysical and biochemical cues, on stem cell mechanoadaptation and emergent lineage commitment. The cytoskeleton provides an adaptive structural scaffold to the cell, regulating the emergence of stem cell structure-function relationships during tissue neogenesis, both in prenatal development as well as postnatal healing. Identification and mapping of the mechanical cues conducive to cytoskeletal remodelling and cell adaptation may help to establish environmental contexts that can be used prospectively as translational design specifications to target tissue neogenesis for regenerative medicine. In this review, we summarize findings on cytoskeletal remodelling in the context of tissue neogenesis during early development and postnatal healing, and its relevance in guiding lineage commitment for targeted tissue regeneration. We highlight how cytoskeleton-targeting chemical agents modulate stem cell differentiation and govern responses to mechanical cues in stem cells' emerging form and function. We further review methods for spatiotemporal visualization and measurement of cytoskeletal remodelling, as well as its effects on the mechanical properties of cells, as a function of adaptation. Research in these areas may facilitate translation of stem cells' own healing potential and improve the design of materials, therapies, and devices for regenerative medicine.
Collapse
Affiliation(s)
- Vina D L Putra
- School of Chemistry and School of Materials Science & Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Kristopher A Kilian
- School of Chemistry and School of Materials Science & Engineering, University of New South Wales, Sydney, NSW, Australia.
| | - Melissa L Knothe Tate
- Blue Mountains World Interdisciplinary Innovation Institute (bmwi³), Blue Mountains, NSW, Australia.
| |
Collapse
|
10
|
Li DX, Ma Z, Szojka ARA, Lan X, Kunze M, Mulet-Sierra A, Westover L, Adesida AB. Non-hypertrophic chondrogenesis of mesenchymal stem cells through mechano-hypoxia programing. J Tissue Eng 2023; 14:20417314231172574. [PMID: 37216035 PMCID: PMC10192798 DOI: 10.1177/20417314231172574] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/09/2023] [Indexed: 05/24/2023] Open
Abstract
Cartilage tissue engineering aims to generate functional replacements to treat cartilage defects from damage and osteoarthritis. Human bone marrow-derived mesenchymal stem cells (hBM-MSC) are a promising cell source for making cartilage, but current differentiation protocols require the supplementation of growth factors like TGF-β1 or -β3. This can lead to undesirable hypertrophic differentiation of hBM-MSC that progress to bone. We have found previously that exposing engineered human meniscus tissues to physiologically relevant conditions of the knee (mechanical loading and hypoxia; hence, mechano-hypoxia conditioning) increased the gene expression of hyaline cartilage markers, SOX9 and COL2A1, inhibited hypertrophic marker COL10A1, and promoted bulk mechanical property development. Adding further to this protocol, we hypothesize that combined mechano-hypoxia conditioning with TGF-β3 growth factor withdrawal will promote stable, non-hypertrophic chondrogenesis of hBM-MSC embedded in an HA-hydrogel. We found that the combined treatment upregulated many cartilage matrix- and development-related markers while suppressing many hypertrophic- and bone development-related markers. Tissue level assessments with biochemical assays, immunofluorescence, and histochemical staining confirmed the gene expression data. Further, mechanical property development in the dynamic compression treatment shows promise toward generating functional engineered cartilage through more optimized and longer culture conditions. In summary, this study introduced a novel protocol to differentiate hBM-MSC into stable, cartilage-forming cells.
Collapse
Affiliation(s)
- David Xinzheyang Li
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Civil and Environmental
Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, AB,
Canada
| | - Zhiyao Ma
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexander RA Szojka
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Xiaoyi Lan
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Civil and Environmental
Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, AB,
Canada
| | - Melanie Kunze
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lindsey Westover
- Department of Mechanical Engineering,
Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
11
|
Vágó J, Katona É, Takács R, Dócs K, Hajdú T, Kovács P, Zákány R, van der Veen DR, Matta C. Cyclic uniaxial mechanical load enhances chondrogenesis through entraining the molecular circadian clock. J Pineal Res 2022; 73:e12827. [PMID: 36030553 PMCID: PMC9786663 DOI: 10.1111/jpi.12827] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/28/2022] [Accepted: 08/20/2022] [Indexed: 12/30/2022]
Abstract
The biomechanical environment plays a key role in regulating cartilage formation, but the current understanding of mechanotransduction pathways in chondrogenic cells is incomplete. Among the combination of external factors that control chondrogenesis are temporal cues that are governed by the cell-autonomous circadian clock. However, mechanical stimulation has not yet directly been proven to modulate chondrogenesis via entraining the circadian clock in chondroprogenitor cells. The purpose of this study was to establish whether mechanical stimuli entrain the core clock in chondrogenic cells, and whether augmented chondrogenesis caused by mechanical loading was at least partially mediated by the synchronised, rhythmic expression of the core circadian clock genes, chondrogenic transcription factors, and cartilage matrix constituents at both transcript and protein levels. We report here, for the first time, that cyclic uniaxial mechanical load applied for 1 h for a period of 6 days entrains the molecular clockwork in chondroprogenitor cells during chondrogenesis in limb bud-derived micromass cultures. In addition to the several core clock genes and proteins, the chondrogenic markers SOX9 and ACAN also followed a robust sinusoidal rhythmic expression pattern. These rhythmic conditions significantly enhanced cartilage matrix production and upregulated marker gene expression. The observed chondrogenesis-promoting effect of the mechanical environment was at least partially attributable to its entraining effect on the molecular clockwork, as co-application of the small molecule clock modulator longdaysin attenuated the stimulatory effects of mechanical load. This study suggests that an optimal biomechanical environment enhances tissue homoeostasis and histogenesis during chondrogenesis at least partially through entraining the molecular clockwork.
Collapse
Affiliation(s)
- Judit Vágó
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Éva Katona
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Klaudia Dócs
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Tibor Hajdú
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Daan R. van der Veen
- Chronobiology Section, Faculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUnited Kingdom
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| |
Collapse
|
12
|
Parada C, Banavar SP, Khalilian P, Rigaud S, Michaut A, Liu Y, Joshy DM, Campàs O, Gros J. Mechanical feedback defines organizing centers to drive digit emergence. Dev Cell 2022; 57:854-866.e6. [PMID: 35413235 DOI: 10.1016/j.devcel.2022.03.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/25/2022] [Accepted: 03/10/2022] [Indexed: 11/03/2022]
Abstract
During embryonic development, digits gradually emerge in a periodic pattern. Although genetic evidence indicates that digit formation results from a self-organizing process, the underlying mechanisms are still unclear. Here, we find that convergent-extension tissue flows driven by active stresses underlie digit formation. These active stresses simultaneously shape cartilage condensations and lead to the emergence of a compressive stress region that promotes high activin/p-SMAD/SOX9 expression, thereby defining digit-organizing centers via a mechanical feedback. In Wnt5a mutants, such mechanical feedback is disrupted due to the loss of active stresses, organizing centers do not emerge, and digit formation is precluded. Thus, digit emergence does not result solely from molecular interactions, as was previously thought, but requires a mechanical feedback that ensures continuous coupling between phalanx specification and elongation. Our work, which links mechanical and molecular signals, provides a mechanistic context for the emergence of organizing centers that may underlie various developmental processes.
Collapse
Affiliation(s)
- Carolina Parada
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75724 Paris Cedex 15, France; CNRS UMR 3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Samhita P Banavar
- Department of Physics, University of California, Santa Barbara, CA 93106-5070, USA
| | - Parisa Khalilian
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75724 Paris Cedex 15, France; CNRS UMR 3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Stephane Rigaud
- Image Analysis Hub, C2RT, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Arthur Michaut
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75724 Paris Cedex 15, France; CNRS UMR 3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Yucen Liu
- Department of Mechanical Engineering, University of California, Santa Barbara, CA 93106-5070, USA
| | - Dennis Manjaly Joshy
- Department of Mechanical Engineering, University of California, Santa Barbara, CA 93106-5070, USA
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA 93106-5070, USA; Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, CA, USA; Cluster of Excellence Physics of Life, TU Dresden, 01062 Dresden, Germany.
| | - Jerome Gros
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75724 Paris Cedex 15, France; CNRS UMR 3738, 25 rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
13
|
Sermeus Y, Vangheel J, Geris L, Smeets B, Tylzanowski P. Mechanical Regulation of Limb Bud Formation. Cells 2022; 11:420. [PMID: 35159230 PMCID: PMC8834596 DOI: 10.3390/cells11030420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/20/2022] [Accepted: 01/23/2022] [Indexed: 12/28/2022] Open
Abstract
Early limb bud development has been of considerable interest for the study of embryological development and especially morphogenesis. The focus has long been on biochemical signalling and less on cell biomechanics and mechanobiology. However, their importance cannot be understated since tissue shape changes are ultimately controlled by active forces and bulk tissue rheological properties that in turn depend on cell-cell interactions as well as extracellular matrix composition. Moreover, the feedback between gene regulation and the biomechanical environment is still poorly understood. In recent years, novel experimental techniques and computational models have reinvigorated research on this biomechanical and mechanobiological side of embryological development. In this review, we consider three stages of early limb development, namely: outgrowth, elongation, and condensation. For each of these stages, we summarize basic biological regulation and examine the role of cellular and tissue mechanics in the morphogenetic process.
Collapse
Affiliation(s)
- Yvenn Sermeus
- MeBioS, KU Leuven, 3000 Leuven, Belgium; (Y.S.); (J.V.); (B.S.)
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
| | - Jef Vangheel
- MeBioS, KU Leuven, 3000 Leuven, Belgium; (Y.S.); (J.V.); (B.S.)
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
- GIGA In Silico Medicine, Université de Liège, 4000 Liège, Belgium
- SBE, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Bart Smeets
- MeBioS, KU Leuven, 3000 Leuven, Belgium; (Y.S.); (J.V.); (B.S.)
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
| | - Przemko Tylzanowski
- SBE, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Molecular Genetics, Department of Biomedical Sciences, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| |
Collapse
|
14
|
Campos Y, Almirall A, Fuentes G, Bloem HL, Kaijzel EL, Cruz LJ. Tissue Engineering: An Alternative to Repair Cartilage. TISSUE ENGINEERING PART B-REVIEWS 2020; 25:357-373. [PMID: 30913997 DOI: 10.1089/ten.teb.2018.0330] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Herein we review the state-of-the-art in tissue engineering for repair of articular cartilage. First, we describe the molecular, cellular, and histologic structure and function of endogenous cartilage, focusing on chondrocytes, collagens, extracellular matrix, and proteoglycans. We then explore in vitro cell culture on scaffolds, discussing the difficulties involved in maintaining or obtaining a chondrocytic phenotype. Next, we discuss the diverse compounds and designs used for these scaffolds, including natural and synthetic biomaterials and porous, fibrous, and multilayer architectures. We then report on the mechanical properties of different cell-loaded scaffolds, and the success of these scaffolds following in vivo implantation in small animals, in terms of generating tissue that structurally and functionally resembles native tissue. Last, we highlight future trends in this field. We conclude that despite major technical advances made over the past 15 years, and continually improving results in cartilage repair experiments in animals, the development of clinically useful implants for regeneration of articular cartilage remains a challenge
Collapse
Affiliation(s)
- Yaima Campos
- 1Biomaterials Center, Havana University, LA Habana, Cuba.,2Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Gastón Fuentes
- 1Biomaterials Center, Havana University, LA Habana, Cuba.,2Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans L Bloem
- 2Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric L Kaijzel
- 2Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Luis J Cruz
- 2Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
15
|
Xiang W, Jiang T, Hao X, Wang R, Yao X, Sun K, Guo F, Xu T. Primary cilia and autophagy interaction is involved in mechanical stress mediated cartilage development via ERK/mTOR axis. Life Sci 2019; 218:308-313. [PMID: 30610869 DOI: 10.1016/j.lfs.2019.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 12/14/2022]
Abstract
AIMS Biomechanical reactivity is a special property of chondrocytes and mechanical stress can affect the development of cartilage. Primary cilia have been proved a cellular sensory which can detect physical and chemical stimuli extracellular and initiate multiple signaling transduction. Autophagy is an important environmental adaptive mechanism for cells maintenance of homeostasis. The aims of this study were to detect whether there is an interaction between primary cilia and autophagy in the regulation of mechanical stress-mediated cartilage development and to explore the underlying mechanism. MAIN METHODS In this study, chondrocytes were treated with cyclic tensile strain (CTS) by the four-point bending system. Chondrocytes viability, proliferation and differentiation capacities were analyzed by western blot and live/dead assays after CTS of different intensities. Meanwhile, primary cilia incidence and length changes, and autophagy expression were detected by immunofluorescence staining. The primary cilia and autophagy interaction regulation and the underlying mechanism were detected by immunofluorescence double staining and western blot. KEY FINDINGS Mechanical stress could affect chondrocytes proliferation, phenotype and viability in an intensity dependent manner. The incidence and length of primary cilia as well as autophagy expression could be regulated by CTS. The integrity of primary cilia structure is vital for mechanical stress regulated ERK/mTOR signaling transduction and autophagy expression in chondrocyte. SIGNIFICANCE These findings indicate that mechanical stress could affect the interaction between primary cilia and autophagy and help to reveal the underlying mechanism of stress regulated cartilage development.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Ting Jiang
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xudong Yao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
16
|
Zamanlui S, Amirabad LM, Soleimani M, Faghihi S. Influence of hydrodynamic pressure on chondrogenic differentiation of human bone marrow mesenchymal stem cells cultured in perfusion system. Biologicals 2018; 56:1-8. [DOI: 10.1016/j.biologicals.2018.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 04/08/2018] [Accepted: 04/20/2018] [Indexed: 11/19/2022] Open
|
17
|
Li X, Shen P, Su W, Zhao S, Zhao J. Into-Tunnel Repair Versus Onto-Surface Repair for Rotator Cuff Tears in a Rabbit Model. Am J Sports Med 2018; 46:1711-1719. [PMID: 29620913 DOI: 10.1177/0363546518764685] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Clinically, onto-surface repair is commonly used for rotator cuff tears. The retear rate after rotator cuff repair (RCR) is relatively high, with failure occurring mostly at the tendon-bone connection site. For anterior cruciate ligament (ACL) reconstruction, into-tunnel reconstruction is commonly employed. The retear rate after ACL reconstruction is relatively low, with retears seldom occurring at the tendon-bone interface. No study on into-tunnel RCR has been conducted. HYPOTHESIS Into-tunnel RCR could promote fibrocartilage regeneration at the tendon-bone interface and has biomechanical advantage over onto-surface repair in a rabbit rotator cuff tear model. STUDY DESIGN Controlled laboratory study. METHODS Thirty-six New Zealand White rabbits were used in this study. The supraspinatus tendons were cut from the footprint to create a rotator cuff tear on both shoulders. On one side, the supraspinatus was cut longitudinally into 2 halves, sutured, and pulled into 2 tunnels through the greater tuberosity (into-tunnel repair). On the other side, the tendon was reattached to the surface of the footprint with transosseous sutures (onto-surface repair). Twelve animals were sacrificed, of which 6 were used for a histological examination and the other 6 for biomechanical testing, at 4, 8, and 12 weeks, respectively. RESULTS The tendon-bone interface in the into-tunnel group showed a different healing pattern from that in the onto-surface group. In the former, most of the tendon tissue in the tunnel was replaced with newly generated fibrocartilage; the rest of the tendon fibers appeared in large bundles with direct connection to the bone. In the latter, fibrocartilage regeneration was seldom found at the tendon-bone interface; the tendon near the bone surface appeared as small fibrils. The biomechanical evaluation revealed a higher ultimate load ( P < .001) and stiffness ( P < .001) at the tendon-bone junction in the into-tunnel group than those in the onto-surface group at 12 weeks. CONCLUSION In a rabbit rotator cuff tear model, into-tunnel RCR could result in a different tendon-bone healing pattern, with obvious fibrocartilage regeneration at the interface and higher tendon-bone healing strength than that in onto-surface repair. CLINICAL RELEVANCE New RCR patterns may be developed to improve the tendon-bone healing pattern and obtain better tendon-bone healing strength.
Collapse
Affiliation(s)
- Xiaoxi Li
- Department of Sports Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Shen
- Department of Sports Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Su
- Department of Sports Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Song Zhao
- Department of Sports Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Castelucci BG, Consonni SR, Rosa VS, Sensiate LA, Delatti PCR, Alvares LE, Joazeiro PP. Time-dependent regulation of morphological changes and cartilage differentiation markers in the mouse pubic symphysis during pregnancy and postpartum recovery. PLoS One 2018; 13:e0195304. [PMID: 29621303 PMCID: PMC5886480 DOI: 10.1371/journal.pone.0195304] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 03/20/2018] [Indexed: 02/02/2023] Open
Abstract
Animal models commonly serve as a bridge between in vitro experiments and clinical applications; however, few physiological processes in adult animals are sufficient to serve as proof-of-concept models for cartilage regeneration. Intriguingly, some rodents, such as young adult mice, undergo physiological connective tissue modifications to birth canal elements such as the pubic symphysis during pregnancy; therefore, we investigated whether the differential expression of cartilage differentiation markers is associated with cartilaginous tissue morphological modifications during these changes. Our results showed that osteochondral progenitor cells expressing Runx2, Sox9, Col2a1 and Dcx at the non-pregnant pubic symphysis proliferated and differentiated throughout pregnancy, giving rise to a complex osteoligamentous junction that attached the interpubic ligament to the pubic bones until labour occurred. After delivery, the recovery of pubic symphysis cartilaginous tissues was improved by the time-dependent expression of these chondrocytic lineage markers at the osteoligamentous junction. This process potentially recapitulates embryologic chondrocytic differentiation to successfully recover hyaline cartilaginous pads at 10 days postpartum. Therefore, we propose that this physiological phenomenon represents a proof-of-concept model for investigating the mechanisms involved in cartilage restoration in adult animals.
Collapse
Affiliation(s)
- Bianca Gazieri Castelucci
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
- * E-mail: (BGC); (SRC); (PPJ)
| | - Sílvio Roberto Consonni
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
- * E-mail: (BGC); (SRC); (PPJ)
| | - Viviane Souza Rosa
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Lucimara Aparecida Sensiate
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Paula Cristina Rugno Delatti
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Lúcia Elvira Alvares
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Paulo Pinto Joazeiro
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
- * E-mail: (BGC); (SRC); (PPJ)
| |
Collapse
|
19
|
Carelli S, Colli M, Vinci V, Caviggioli F, Klinger M, Gorio A. Mechanical Activation of Adipose Tissue and Derived Mesenchymal Stem Cells: Novel Anti-Inflammatory Properties. Int J Mol Sci 2018; 19:ijms19010267. [PMID: 29337886 PMCID: PMC5796213 DOI: 10.3390/ijms19010267] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 12/29/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
The adipose tissue is a source of inflammatory proteins, such as TNF, IL-6, and CXCL8. Most of their production occurs in macrophages that act as scavengers of dying adipocytes. The application of an orbital mechanical force for 6-10 min at 97 g to the adipose tissue, lipoaspirated and treated according to Coleman procedures, abolishes the expression of TNF-α and stimulates the expression of the anti-inflammatory protein TNF-stimulated gene-6 (TSG-6). This protein had protective and anti-inflammatory effects when applied to animal models of rheumatic diseases. We examined biopsy, lipoaspirate, and mechanically activated fat and observed that in addition to the increased TSG-6, Sox2, Nanog, and Oct4 were also strongly augmented by mechanical activation, suggesting an effect on stromal cell stemness. Human adipose tissue-derived mesenchymal stem cells (hADSCs), produced from activated fat, grow and differentiate normally with proper cell surface markers and chromosomal integrity, but their anti-inflammatory action is far superior compared to those mesenchymal stem cells (MSCs) obtained from lipoaspirate. The expression and release of inflammatory cytokines from THP-1 cells was totally abolished in mechanically activated adipose tissue-derived hADSCs. In conclusion, we report that the orbital shaking of adipose tissue enhances its anti-inflammatory properties, and derived MSCs maintain such enhanced activity.
Collapse
Affiliation(s)
- Stephana Carelli
- Pediatric Clinical Research Center "Fondazione Romeo e Enrica Invernizzi", University of Milan, 20142 Milan, Italy.
| | - Mattia Colli
- Pediatric Clinical Research Center "Fondazione Romeo e Enrica Invernizzi", University of Milan, 20142 Milan, Italy.
| | - Valeriano Vinci
- Humanitas Research Hospital, Plastic Surgery Unit, Via Manzoni 56, 20089 Rozzano, Italy.
| | - Fabio Caviggioli
- Multimedica San Giuseppe Hospital, Plastic Surgery Unit, Via San Vittore 12, 20123 Milan, Italy.
| | - Marco Klinger
- Humanitas Research Hospital, Plastic Surgery Unit, Via Manzoni 56, 20089 Rozzano, Italy.
| | - Alfredo Gorio
- Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Via A. di Rudinì 8, 20142 Milan, Italy.
| |
Collapse
|
20
|
Fahy N, Alini M, Stoddart MJ. Mechanical stimulation of mesenchymal stem cells: Implications for cartilage tissue engineering. J Orthop Res 2018; 36:52-63. [PMID: 28763118 DOI: 10.1002/jor.23670] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/24/2017] [Indexed: 02/04/2023]
Abstract
Articular cartilage is a load-bearing tissue playing a crucial mechanical role in diarthrodial joints, facilitating joint articulation, and minimizing wear. The significance of biomechanical stimuli in the development of cartilage and maintenance of chondrocyte phenotype in adult tissues has been well documented. Furthermore, dysregulated loading is associated with cartilage pathology highlighting the importance of mechanical cues in cartilage homeostasis. The repair of damaged articular cartilage resulting from trauma or degenerative joint disease poses a major challenge due to a low intrinsic capacity of cartilage for self-renewal, attributable to its avascular nature. Bone marrow-derived mesenchymal stem cells (MSCs) are considered a promising cell type for cartilage replacement strategies due to their chondrogenic differentiation potential. Chondrogenesis of MSCs is influenced not only by biological factors but also by the environment itself, and various efforts to date have focused on harnessing biomechanics to enhance chondrogenic differentiation of MSCs. Furthermore, recapitulating mechanical cues associated with cartilage development and homeostasis in vivo, may facilitate the development of a cellular phenotype resembling native articular cartilage. The goal of this review is to summarize current literature examining the effect of mechanical cues on cartilage homeostasis, disease, and MSC chondrogenesis. The role of biological factors produced by MSCs in response to mechanical loading will also be examined. An in-depth understanding of the impact of mechanical stimulation on the chondrogenic differentiation of MSCs in terms of endogenous bioactive factor production and signaling pathways involved, may identify therapeutic targets and facilitate the development of more robust strategies for cartilage replacement using MSCs. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:52-63, 2018.
Collapse
Affiliation(s)
- Niamh Fahy
- AO Research Institute Davos, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | |
Collapse
|
21
|
Vargas AO, Ruiz-Flores M, Soto-Acuña S, Haidr N, Acosta-Hospitaleche C, Ossa-Fuentes L, Muñoz-Walther V. The Origin and Evolutionary Consequences of Skeletal Traits Shaped by Embryonic Muscular Activity, from Basal Theropods to Modern Birds. Integr Comp Biol 2017; 57:1281-1292. [DOI: 10.1093/icb/icx074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
22
|
Paracrine Potential of the Human Adipose Tissue-Derived Stem Cells to Modulate Balance between Matrix Metalloproteinases and Their Inhibitors in the Osteoarthritic Cartilage In Vitro. Stem Cells Int 2017; 2017:9542702. [PMID: 28819366 PMCID: PMC5551534 DOI: 10.1155/2017/9542702] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/10/2017] [Accepted: 05/15/2017] [Indexed: 01/01/2023] Open
Abstract
Adipose tissue represents an abundant source of stem cells. Along with anti-inflammatory effects, ASC secrete various factors that may modulate metabolism of extracellular matrix in osteoarthritic (OA) cartilage, suggesting that the presence of ASC could be advantageous for OA cartilage due to the recovery of homeostasis between matrix metalloproteinases (MMPs) and their tissue inhibitors of metalloproteinases (TIMPs). To evaluate these effects, cartilage explants (CE) were cocultured with ASC for 3 and 7 days under stimulation with or without IL-1β. The pattern of gene expression in CE was modified by ASC, including the upregulation of COL1A1 and COL3A1 and the downregulation of MMP13 and COL10A1. The production of MMP-1, MMP-3, and MMP-13 by ASC was not significant; moreover, cocultures with ASC reduced MMP-13 production in CE. In conclusion, active production of TIMP-1, TIMP-2, TIMP-3, IL-6, IL-8, and gelatinases MMP-2 and MMP-9 by ASC may be involved in the extracellular matrix remodelling, as indicated by the altered expression of collagens, the downregulated production of MMP-13, and the reduced chondrocyte apoptosis in the cocultured CE. These data suggest that ASC modulated homeostasis of MMPs/TIMPs in degenerated OA cartilage in vitro and might be favourable in case of the intra-articular application of ASC therapy for the treatment of OA.
Collapse
|
23
|
Shen G, Darendeliler MA. The Adaptive Remodeling of Condylar Cartilage— A Transition from Chondrogenesis to Osteogenesis. J Dent Res 2016; 84:691-9. [PMID: 16040724 DOI: 10.1177/154405910508400802] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mandibular condylar cartilage is categorized as articular cartilage but markedly distinguishes itself in many biological aspects, such as its embryonic origin, ontogenetic development, post-natal growth mode, and histological structures. The most marked uniqueness of condylar cartilage lies in its capability of adaptive remodeling in response to external stimuli during or after natural growth. The adaptation of condylar cartilage to mandibular forward positioning constitutes the fundamental rationale for orthodontic functional therapy, which partially contributes to the correction of jaw discrepancies by achieving mandibular growth modification. The adaptive remodeling of condylar cartilage proceeds with the biomolecular pathway initiating from chondrogenesis and finalizing with osteogenesis. During condylar adaptation, chondrogenesis is activated when the external stimuli, e.g., condylar repositioning, generate the differentiation of mesenchymal cells in the articular layer of cartilage into chondrocytes, which proliferate and then progressively mature into hypertrophic cells. The expression of regulatory growth factors, which govern and control phenotypic conversions of chondrocytes during chondrogenesis, increases during adaptive remodeling to enhance the transition from chondrogenesis into osteogenesis, a process in which hypertrophic chondrocytes and matrices degrade and are replaced by bone. The transition is also sustained by increased neovascularization, which brings in osteoblasts that finally result in new bone formation beneath the degraded cartilage.
Collapse
Affiliation(s)
- G Shen
- Discipline of Orthodontics, Faculty of Dentistry, Sydney Dental Hospital, The University of Sydney, 2 Chalmers Street, Surry Hills, NSW 2010, Australia.
| | | |
Collapse
|
24
|
Luciani N, Du V, Gazeau F, Richert A, Letourneur D, Le Visage C, Wilhelm C. Successful chondrogenesis within scaffolds, using magnetic stem cell confinement and bioreactor maturation. Acta Biomater 2016; 37:101-10. [PMID: 27063490 DOI: 10.1016/j.actbio.2016.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 01/24/2023]
Abstract
UNLABELLED Tissue engineering strategies, such as cellularized scaffolds approaches, have been explored for cartilage replacement. The challenge, however, remains to produce a cartilaginous tissue incorporating functional chondrocytes and being large and thick enough to be compatible with the replacement of articular defects. Here, we achieved unprecedented cartilage tissue production into a porous polysaccharide scaffold by combining of efficient magnetic condensation of mesenchymal stem cells, and dynamic maturation in a bioreactor. In optimal conditions, all the hallmarks of chondrogenesis were enhanced with a 50-fold increase in collagen II expression compared to negative control, an overexpression of aggrecan and collagen XI, and a very low expression of collagen I and RUNX2. Histological staining showed a large number of cellular aggregates, as well as an increased proteoglycan synthesis by chondrocytes. Interestingly, electron microscopy showed larger chondrocytes and a more abundant extracellular matrix. In addition, the periodicity of the neosynthesized collagen fibers matched that of collagen II. These results represent a major step forward in replacement tissue for cartilage defects. STATEMENT OF SIGNIFICANCE A combination of several innovative technologies (magnetic cell seeding, polysaccharide porous scaffolds, and dynamic maturation in bioreactor) enabled unprecedented successful chondrogenesis within scaffolds.
Collapse
Affiliation(s)
- Nathalie Luciani
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France.
| | - Vicard Du
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| | - Alain Richert
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| | - Didier Letourneur
- Laboratoire de recherche vasculaire translationnelle, INSERM UMR 1148 & University Paris Diderot, Paris, France
| | | | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| |
Collapse
|
25
|
Peterson T, Müller GB. Phenotypic Novelty in EvoDevo: The Distinction Between Continuous and Discontinuous Variation and Its Importance in Evolutionary Theory. Evol Biol 2016; 43:314-335. [PMID: 27512237 PMCID: PMC4960286 DOI: 10.1007/s11692-016-9372-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/29/2016] [Indexed: 10/25/2022]
Abstract
The introduction of novel phenotypic structures is one of the most significant aspects of organismal evolution. Yet the concept of evolutionary novelty is used with drastically different connotations in various fields of research, and debate exists about whether novelties represent features that are distinct from standard forms of phenotypic variation. This article contrasts four separate uses for novelty in genetics, population genetics, morphology, and behavioral science, before establishing how novelties are used in evolutionary developmental biology (EvoDevo). In particular, it is detailed how an EvoDevo-specific research approach to novelty produces insight distinct from other fields, gives the concept explanatory power with predictive capacities, and brings new consequences to evolutionary theory. This includes the outlining of research strategies that draw attention to productive areas of inquiry, such as threshold dynamics in development. It is argued that an EvoDevo-based approach to novelty is inherently mechanistic, treats the phenotype as an agent with generative potential, and prompts a distinction between continuous and discontinuous variation in evolutionary theory.
Collapse
Affiliation(s)
- Tim Peterson
- Department of Theoretical Biology, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Gerd B. Müller
- Department of Theoretical Biology, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- The KLI Institute, Martinstrasse 12, 3400 Klosterneuburg, Austria
| |
Collapse
|
26
|
Chen C, Wei X, Wang S, Jiao Q, Zhang Y, Du G, Wang X, Wei F, Zhang J, Wei L. Compression regulates gene expression of chondrocytes through HDAC4 nuclear relocation via PP2A-dependent HDAC4 dephosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1633-42. [PMID: 27106144 DOI: 10.1016/j.bbamcr.2016.04.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 04/13/2016] [Accepted: 04/17/2016] [Indexed: 11/15/2022]
Abstract
Biomechanics plays a critical role in the modulation of chondrocyte function. The mechanisms by which mechanical loading is transduced into intracellular signals that regulate chondrocyte gene expression remain largely unknown. Histone deacetylase 4 (HDAC4) is specifically expressed in chondrocytes. Mice lacking HDAC4 display chondrocyte hypertrophy, ectopic and premature ossification, and die early during the perinatal period. HDAC4 has a remarkable ability to translocate between the cell's cytoplasm and nucleus. It has been established that subcellular relocation of HDAC4 plays a critical role in chondrocyte differentiation and proliferation. However, it remains unclear whether subcellular relocation of HDAC4 in chondrocytes can be induced by mechanical loading. In this study, we first report that compressive loading induces HDAC4 relocation from the cytoplasm to the nucleus of chondrocytes via stimulation of Ser/Thr-phosphoprotein phosphatases 2A (PP2A) activity, which results in dephosphorylation of HDAC4. Dephosphorylated HDAC4 relocates to the nucleus to achieve transcriptional repression of Runx2 and regulates chondrocyte gene expression in response to compression. Our results elucidate the mechanism by which mechanical compression regulates chondrocyte gene expression through HDAC4 relocation from the cell's cytoplasm to the nucleus via PP2A-dependent HDAC4 dephosphorylation.
Collapse
Affiliation(s)
- Chongwei Chen
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Shaowei Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Qiang Jiao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Yang Zhang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Guoqing Du
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Xiaohu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Fangyuan Wei
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jianzhong Zhang
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Lei Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA.
| |
Collapse
|
27
|
Green JD, Tollemar V, Dougherty M, Yan Z, Yin L, Ye J, Collier Z, Mohammed MK, Haydon RC, Luu HH, Kang R, Lee MJ, Ho SH, He TC, Shi LL, Athiviraham A. Multifaceted signaling regulators of chondrogenesis: Implications in cartilage regeneration and tissue engineering. Genes Dis 2015; 2:307-327. [PMID: 26835506 PMCID: PMC4730920 DOI: 10.1016/j.gendis.2015.09.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/16/2015] [Indexed: 01/08/2023] Open
Abstract
Defects of articular cartilage present a unique clinical challenge due to its poor self-healing capacity and avascular nature. Current surgical treatment options do not ensure consistent regeneration of hyaline cartilage in favor of fibrous tissue. Here, we review the current understanding of the most important biological regulators of chondrogenesis and their interactions, to provide insight into potential applications for cartilage tissue engineering. These include various signaling pathways, including: fibroblast growth factors (FGFs), transforming growth factor β (TGF-β)/bone morphogenic proteins (BMPs), Wnt/β-catenin, Hedgehog, Notch, hypoxia, and angiogenic signaling pathways. Transcriptional and epigenetic regulation of chondrogenesis will also be discussed. Advances in our understanding of these signaling pathways have led to promising advances in cartilage regeneration and tissue engineering.
Collapse
Affiliation(s)
- Jordan D. Green
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Viktor Tollemar
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Mark Dougherty
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhengjian Yan
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Liangjun Yin
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jixing Ye
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Bioengineering, Chongqing University, Chongqing, China
| | - Zachary Collier
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Maryam K. Mohammed
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Richard Kang
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin H. Ho
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
28
|
Guo T, Yu L, Lim CG, Goodley AS, Xiao X, Placone JK, Ferlin KM, Nguyen BNB, Hsieh AH, Fisher JP. Effect of Dynamic Culture and Periodic Compression on Human Mesenchymal Stem Cell Proliferation and Chondrogenesis. Ann Biomed Eng 2015; 44:2103-13. [PMID: 26577256 DOI: 10.1007/s10439-015-1510-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/11/2015] [Indexed: 12/27/2022]
Abstract
We have recently developed a bioreactor that can apply both shear and compressive forces to engineered tissues in dynamic culture. In our system, alginate hydrogel beads with encapsulated human mesenchymal stem cells (hMSCs) were cultured under different dynamic conditions while subjected to periodic, compressive force. A customized pressure sensor was developed to track the pressure fluctuations when shear forces and compressive forces were applied. Compared to static culture, dynamic culture can maintain a higher cell population throughout the study. With the application of only shear stress, qRT-PCR and immunohistochemistry revealed that hMSCs experienced less chondrogenic differentiation than the static group. The second study showed that chondrogenic differentiation was enhanced by additional mechanical compression. After 14 days, alcian blue staining showed more extracellular matrix formed in the compression group. The upregulation of the positive chondrogenic markers such as Sox 9, aggrecan, and type II collagen were demonstrated by qPCR. Our bioreactor provides a novel approach to apply mechanical forces to engineered cartilage. Results suggest that a combination of dynamic culture with proper mechanical stimulation may promote efficient progenitor cell expansion in vitro, thereby allowing the culture of clinically relevant articular chondrocytes for the treatment of articular cartilage defects.
Collapse
Affiliation(s)
- Ting Guo
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Li Yu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Casey G Lim
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Addison S Goodley
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Xuan Xiao
- Department of Ophthalmology, Renming Hospital of Wuhan University, Wuhan, China
| | - Jesse K Placone
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Kimberly M Ferlin
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Bao-Ngoc B Nguyen
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Adam H Hsieh
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA.
| |
Collapse
|
29
|
Font Tellado S, Balmayor ER, Van Griensven M. Strategies to engineer tendon/ligament-to-bone interface: Biomaterials, cells and growth factors. Adv Drug Deliv Rev 2015; 94:126-40. [PMID: 25777059 DOI: 10.1016/j.addr.2015.03.004] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/27/2015] [Accepted: 03/07/2015] [Indexed: 02/06/2023]
Abstract
Integration between tendon/ligament and bone occurs through a specialized tissue interface called enthesis. The complex and heterogeneous structure of the enthesis is essential to ensure smooth mechanical stress transfer between bone and soft tissues. Following injury, the interface is not regenerated, resulting in high rupture recurrence rates. Tissue engineering is a promising strategy for the regeneration of a functional enthesis. However, the complex structural and cellular composition of the native interface makes enthesis tissue engineering particularly challenging. Thus, it is likely that a combination of biomaterials and cells stimulated with appropriate biochemical and mechanical cues will be needed. The objective of this review is to describe the current state-of-the-art, challenges and future directions in the field of enthesis tissue engineering focusing on four key parameters: (1) scaffold and biomaterials, (2) cells, (3) growth factors and (4) mechanical stimuli.
Collapse
Affiliation(s)
- Sonia Font Tellado
- Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany.
| | - Elizabeth R Balmayor
- Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Martijn Van Griensven
- Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| |
Collapse
|
30
|
Uekita H, Takahashi S, Domon T, Yamaguchi T. Changes in collagens and chondrocytes in the temporomandibular joint cartilage in growing rats fed a liquid diet. Ann Anat 2015; 202:78-87. [PMID: 26434755 DOI: 10.1016/j.aanat.2015.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/27/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022]
Abstract
The temporomandibular joint (TMJ) of growing rats fed a soft diet is reported to be smaller in size and to have thinner condyle and glenoid fossa cartilage than rats fed a solid diet. The aim of this study was to determine the effect of a soft diet on the collagens and chondrocytes in the growing TMJ cartilage. Forty-eight male Wistar rats were divided into a control group fed a solid diet and an experimental group fed a liquid diet for 1-8 weeks. After the experimental period, the TMJs were harvested and examined histologically, immunohistochemically for collagen types I, II, and X, and with transmission electron microscopy. The condylar cartilage in the experimental rats showed weak immunoreactions for three types of collagens compared with the controls. The ultrastructure had fewer fine collagen fibrils in the experimental rats compared with that of the controls. The glenoid fossa cartilage in the experimental rats showed narrower Alcian blue-positive areas than the control staining. The immunoreactions for three types of collagen in the experimental rats were also weaker than those of the controls. The chondrocytes in the experimental rats appeared dark, had extended thin cytoplasmic processes, and had formed gap junctions, as assessed by transmission electron microscopy. Fewer fine collagen fibrils, but thick bands of collagen fibrils were observed in the glenoid fossa of the experimental cartilage. The results of the present study showed that a liquid diet had deleterious effects on the quality and quantity of collagens and chondrocytes in the TMJ cartilage in growing rats.
Collapse
Affiliation(s)
- Hiroki Uekita
- Division of Crown and Bridge Prosthodontics, Department of Oral Functional Science, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan; Division of Oral Functional Anatomy, Department of Oral Functional Science, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.
| | - Shigeru Takahashi
- Division of Oral Functional Anatomy, Department of Oral Functional Science, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Takanori Domon
- Division of Oral Functional Anatomy, Department of Oral Functional Science, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Taihiko Yamaguchi
- Division of Crown and Bridge Prosthodontics, Department of Oral Functional Science, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| |
Collapse
|
31
|
Akhmanova M, Osidak E, Domogatsky S, Rodin S, Domogatskaya A. Physical, Spatial, and Molecular Aspects of Extracellular Matrix of In Vivo Niches and Artificial Scaffolds Relevant to Stem Cells Research. Stem Cells Int 2015; 2015:167025. [PMID: 26351461 PMCID: PMC4553184 DOI: 10.1155/2015/167025] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/07/2015] [Accepted: 06/24/2015] [Indexed: 12/27/2022] Open
Abstract
Extracellular matrix can influence stem cell choices, such as self-renewal, quiescence, migration, proliferation, phenotype maintenance, differentiation, or apoptosis. Three aspects of extracellular matrix were extensively studied during the last decade: physical properties, spatial presentation of adhesive epitopes, and molecular complexity. Over 15 different parameters have been shown to influence stem cell choices. Physical aspects include stiffness (or elasticity), viscoelasticity, pore size, porosity, amplitude and frequency of static and dynamic deformations applied to the matrix. Spatial aspects include scaffold dimensionality (2D or 3D) and thickness; cell polarity; area, shape, and microscale topography of cell adhesion surface; epitope concentration, epitope clustering characteristics (number of epitopes per cluster, spacing between epitopes within cluster, spacing between separate clusters, cluster patterns, and level of disorder in epitope arrangement), and nanotopography. Biochemical characteristics of natural extracellular matrix molecules regard diversity and structural complexity of matrix molecules, affinity and specificity of epitope interaction with cell receptors, role of non-affinity domains, complexity of supramolecular organization, and co-signaling by growth factors or matrix epitopes. Synergy between several matrix aspects enables stem cells to retain their function in vivo and may be a key to generation of long-term, robust, and effective in vitro stem cell culture systems.
Collapse
Affiliation(s)
| | - Egor Osidak
- Imtek Limited, 3 Cherepkovskaya 15, Moscow 21552, Russia
- Gamaleya Research Institute of Epidemiology and Microbiology Federal State Budgetary Institution, Ministry of Health of the Russian Federation, Gamalei 18, Moscow 123098, Russia
| | - Sergey Domogatsky
- Imtek Limited, 3 Cherepkovskaya 15, Moscow 21552, Russia
- Russian Cardiology Research and Production Center Federal State Budgetary Institution, Ministry of Health of the Russian Federation, 3 Cherepkovskaya 15, Moscow 21552, Russia
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Anna Domogatskaya
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| |
Collapse
|
32
|
Yang L, Yang Y, Wang S, Li Y, Zhao Z. In vitro mechanical loading models for periodontal ligament cells: From two-dimensional to three-dimensional models. Arch Oral Biol 2015; 60:416-24. [DOI: 10.1016/j.archoralbio.2014.11.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 02/08/2023]
|
33
|
Steward AJ, Kelly DJ. Mechanical regulation of mesenchymal stem cell differentiation. J Anat 2014; 227:717-31. [PMID: 25382217 DOI: 10.1111/joa.12243] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2014] [Indexed: 12/18/2022] Open
Abstract
Biophysical cues play a key role in directing the lineage commitment of mesenchymal stem cells or multipotent stromal cells (MSCs), but the mechanotransductive mechanisms at play are still not fully understood. This review article first describes the roles of both substrate mechanics (e.g. stiffness and topography) and extrinsic mechanical cues (e.g. fluid flow, compression, hydrostatic pressure, tension) on the differentiation of MSCs. A specific focus is placed on the role of such factors in regulating the osteogenic, chondrogenic, myogenic and adipogenic differentiation of MSCs. Next, the article focuses on the cellular components, specifically integrins, ion channels, focal adhesions and the cytoskeleton, hypothesized to be involved in MSC mechanotransduction. This review aims to illustrate the strides that have been made in elucidating how MSCs sense and respond to their mechanical environment, and also to identify areas where further research is needed.
Collapse
Affiliation(s)
- Andrew J Steward
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
| | - Daniel J Kelly
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Shimomura K, Kanamoto T, Kita K, Akamine Y, Nakamura N, Mae T, Yoshikawa H, Nakata K. Cyclic compressive loading on 3D tissue of human synovial fibroblasts upregulates prostaglandin E2 via COX-2 production without IL-1β and TNF-α. Bone Joint Res 2014; 3:280-8. [PMID: 25237168 PMCID: PMC4178306 DOI: 10.1302/2046-3758.39.2000287] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Objective Excessive mechanical stress on synovial joints causes osteoarthritis
(OA) and results in the production of prostaglandin E2 (PGE2), a
key molecule in arthritis, by synovial fibroblasts. However, the
relationship between arthritis-related molecules and mechanical
stress is still unclear. The purpose of this study was to examine
the synovial fibroblast response to cyclic mechanical stress using
an in vitro osteoarthritis model. Method Human synovial fibroblasts were cultured on collagen scaffolds
to produce three-dimensional constructs. A cyclic compressive loading
of 40 kPa at 0.5 Hz was applied to the constructs, with or without
the administration of a cyclooxygenase-2 (COX-2) selective inhibitor
or dexamethasone, and then the concentrations of PGE2, interleukin-1β (IL-1β),
tumour necrosis factor-α (TNF-α), IL-6, IL-8 and COX-2 were measured. Results The concentrations of PGE2, IL-6 and IL-8 in the loaded samples
were significantly higher than those of unloaded samples; however,
the concentrations of IL-1β and TNF-α were the same as the unloaded
samples. After the administration of a COX-2 selective inhibitor,
the increased concentration of PGE2 by cyclic compressive loading
was impeded, but the concentrations of IL-6 and IL-8 remained high.
With dexamethasone, upregulation of PGE2, IL-6 and IL-8 was suppressed. Conclusion These results could be useful in revealing the molecular mechanism
of mechanical stress in vivo for a better understanding
of the pathology and therapy of OA. Cite this article: Bone Joint Res 2014;3:280–8.
Collapse
Affiliation(s)
- K Shimomura
- Osaka University Graduate School of Medicine, Department of Orthopaedics, 2-2 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - T Kanamoto
- Osaka University Graduate School of Medicine, Department of Orthopaedics, 2-2 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - K Kita
- Osaka University Graduate School of Medicine, Department of Orthopaedics, 2-2 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Y Akamine
- Osaka University Graduate School of Medicine, Department of Orthopaedics, 2-2 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - N Nakamura
- Osaka Health Science University, Department of Rehabilitation Science, 1-9-27 Tenma, Kita-ku, Osaka City, Osaka 530-0043, Japan
| | - T Mae
- Osaka University Graduate School of Medicine, Department of Orthopaedics, 2-2 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - H Yoshikawa
- Osaka University Graduate School of Medicine, Department of Orthopaedics, 2-2 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - K Nakata
- Osaka University Graduate School of Medicine, Department of Health and Sport Sciences, 1-17 Machikaneyamacho, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
35
|
SAFSHEKAN FARZANEH, SHADPOUR MOHAMMADTAFAZZOLI, SHOKRGOZAR MOHAMMADALI, HAGHIGHIPOUR NOOSHIN, ALAVI SEYEDHAMED. EFFECTS OF SHORT-TERM CYCLIC HYDROSTATIC PRESSURE ON INITIATING AND ENHANCING THE EXPRESSION OF CHONDROGENIC GENES IN HUMAN ADIPOSE-DERIVED MESENCHYMAL STEM CELLS. J MECH MED BIOL 2014; 14:1450054. [DOI: 10.1142/s0219519414500547] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Cartilage tissue engineering is a promising treatment for damaged or diseased cartilage that requires thorough understanding of influential parameters involved in chondrogenic differentiation. This study examined how 4-h application of cyclic hydrostatic pressure (CHP) of 5 MPa at 0.5 Hz could modulate chondroinduction of human adipose-derived mesenchymal stem cells (hAMSCs) in vitro. Four groups were examined including a negative control group, a chemical group treated by growth factor for 10 days, a mechanical group exposed to 4-h loading on the 10th day of pellet culture without any chondrogenic stimulator, and finally a chemical-mechanical group subjected to both growth factor and loading. Application of cyclic hydrostatic pressure increased the expression of chondrogenic genes, including sox9 and aggrecan to higher levels than those of the chemical group. This study indicates that cyclic hydrostatic pressure initiates and enhances the chondrogenic differentiation of mesenchymal stem cells with or without growth factors in vitro and confirms the important role of hydrostatic pressure during chondrogenesis in vivo.
Collapse
Affiliation(s)
- FARZANEH SAFSHEKAN
- Faculty of Biomedical Engineering, Amirkabir University of Technology, 424, Hafez Ave, Tehran, Iran
| | | | - MOHAMMAD ALI SHOKRGOZAR
- National Cell Bank of Iran, Pasteur Institute of Iran, 69, Pasteur Ave, P. O. Box 1316943551, Tehran, Iran
| | - NOOSHIN HAGHIGHIPOUR
- National Cell Bank of Iran, Pasteur Institute of Iran, 69, Pasteur Ave, P. O. Box 1316943551, Tehran, Iran
| | - SEYED HAMED ALAVI
- Faculty of Biomedical Engineering, Amirkabir University of Technology, 424, Hafez Ave, Tehran, Iran
| |
Collapse
|
36
|
Wang Y, Yuan M, Guo QY, Lu SB, Peng J. Mesenchymal Stem Cells for Treating Articular Cartilage Defects and Osteoarthritis. Cell Transplant 2014; 24:1661-78. [PMID: 25197793 DOI: 10.3727/096368914x683485] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Articular cartilage damage and osteoarthritis are the most common joint diseases. Joints are prone to damage caused by sports injuries or aging, and such damage regularly progresses to more serious joint disorders, including osteoarthritis, which is a degenerative disease characterized by the thinning and eventual wearing out of articular cartilage, ultimately leading to joint destruction. Osteoarthritis affects millions of people worldwide. Current approaches to repair of articular cartilage damage include mosaicplasty, microfracture, and injection of autologous chondrocytes. These treatments relieve pain and improve joint function, but the long-term results are unsatisfactory. The long-term success of cartilage repair depends on development of regenerative methodologies that restore articular cartilage to a near-native state. Two promising approaches are (i) implantation of engineered constructs of mesenchymal stem cell (MSC)-seeded scaffolds, and (ii) delivery of an appropriate population of MSCs by direct intra-articular injection. MSCs may be used as trophic producers of bioactive factors initiating regenerative activities in a defective joint. Current challenges in MSC therapy are the need to overcome current limitations in cartilage cell purity and to in vitro engineer tissue structures exhibiting the required biomechanical properties. This review outlines the current status of MSCs used in cartilage tissue engineering and in cell therapy seeking to repair articular cartilage defects and related problems. MSC-based technologies show promise when used to repair cartilage defects in joints.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | |
Collapse
|
37
|
Burke DP, Khayyeri H, Kelly DJ. Substrate stiffness and oxygen availability as regulators of mesenchymal stem cell differentiation within a mechanically loaded bone chamber. Biomech Model Mechanobiol 2014; 14:93-105. [DOI: 10.1007/s10237-014-0591-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 04/24/2014] [Indexed: 10/25/2022]
|
38
|
Zhang S, Dutton JR, Su L, Zhang J, Ye L. The influence of a spatiotemporal 3D environment on endothelial cell differentiation of human induced pluripotent stem cells. Biomaterials 2014; 35:3786-93. [PMID: 24485793 DOI: 10.1016/j.biomaterials.2014.01.037] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/13/2014] [Indexed: 12/01/2022]
Abstract
Current EC differentiation protocols are inefficient, and the phenotypes of the differentiated ECs are only briefly stable, which significantly inhibits their utility for basic science research. Here, a remarkably more efficient hiPSC-EC differentiation protocol that incorporates a three-dimensional (3D) fibrin scaffold is presented. With this protocol, up to 45% of the differentiated hiPSCs assumed an EC phenotype, and after purification, greater than 95% of the cells displayed the EC phenotype (based on CD31 expression). The hiPSC-ECs continued to display EC characteristics for 4 weeks in vitro. Gene and protein expression levels of CD31, CD144 and von Willebrand factor-8 (vWF-8) were significantly up-regulated in differentiated hiPSC-ECs. hiPSC-ECs also have biological function to up-take Dil-conjugated acetylated LDL (Dil-ac-LDL) and form tubular structures on Matrigel. Collectively, these data demonstrate that a 3D differentiation protocol can efficiently generate ECs from hiPSCs and, furthermore, the differentiated hiPSC-ECs are functional and can maintain EC fate up to 4 weeks in vitro.
Collapse
Affiliation(s)
- Sophia Zhang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - James R Dutton
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Liping Su
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jianyi Zhang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lei Ye
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
39
|
Takahashi I, Matsuzaki T, Yoshida S, Kitade I, Hoso M. Differences in Cartilage Repair between Loading and Unloading Environments in the Rat Knee. JOURNAL OF THE JAPANESE PHYSICAL THERAPY ASSOCIATION 2014; 17:22-30. [PMID: 25792905 DOI: 10.1298/jjpta.vol17_004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 05/07/2014] [Indexed: 12/26/2022]
Abstract
We investigated the histopathological and immunohistochemical effects of loading on cartilage repair in rat full-thickness articular cartilage defects. A total of 40 male 9-week-old Wistar rats were studied. Full-thickness articular cartilage defects were created over the capsule at the loading portion in the medial condyle of the femur. Twenty rats were randomly allocated into each of the 2 groups: a loading group and a unloading group. Twenty rats from these 2 groups were later randomly allocated to each of the 2 groups for evaluation at 1 and 2 weeks after surgery. At the end of each period, knee joints were examined histopathologically and immunohistochemically. In both groups at 1 and 2 weeks, the defects were filled with a mixture of granulation tissue and some remnants of hyaline cartilage. The repair tissue was not stained with toluidine blue in both groups. Strong staining of type I collagen was observed in the repair tissue of both groups. The area stained with type I collagen was smaller in the unloading group than in the loading groups, and the stained area was smaller at 2 weeks than at 1 week. In the staining for type II collagen, apparent staining of type II collagen was observed in the repair tissue of both groups at 1 week. At 2 weeks, there was a tendency toward a higher degree of apparent staining in the loading group than in the unloading group. Accordingly, these results indicated that loading and unloading in the early phase of cartilage repair have both merits and demerits.
Collapse
Affiliation(s)
- Ikufumi Takahashi
- Department of Rehabilitation, Houju Memorial Hospital.,School of Health Sciences, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Taro Matsuzaki
- School of Health Sciences, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Shinya Yoshida
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University.,Department of Rehabilitation Medicine, Kanazawa University Hospital
| | - Ippei Kitade
- School of Health Sciences, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University.,Department of Rehabilitation Medicine, University of Fukui Hospital
| | - Masahiro Hoso
- School of Health Sciences, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| |
Collapse
|
40
|
Cell sources for nucleus pulposus regeneration. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2013; 23 Suppl 3:S364-74. [PMID: 24297331 DOI: 10.1007/s00586-013-3106-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 11/07/2013] [Accepted: 11/07/2013] [Indexed: 12/12/2022]
Abstract
PURPOSE There is increasing interest in the development of cell therapy as a possible approach for the treatment of degenerative disc disease. To regenerate nucleus pulposus tissue, the cells must produce an appropriate proteoglycan-rich matrix, as this is essential for the functioning of the intervertebral disc. The natural environment within the disc is very challenging to implanted cells, particularly if they have been subcultured in normal laboratory conditions. The purpose of this work is to discuss parameters relevant to translating different proposed cell therapies of IVD into clinical use. RESULTS Several sources of cells have been proposed, including nucleus pulposus cells, chondrocytes and mesenchymal stem cells derived from bone marrow or adipose tissue. There are some clinical trials and reports of attempts to regenerate nucleus pulposus utilising either autologous or allogenic cells. While the published results of clinical applications of these cell therapies do not indicate any safety issues, additional evidence will be needed to prove their long-term efficacy. CONCLUSION This article discusses parameters relevant for successful translation of research on different cell sources into clinically applicable cell therapies: the influence of the intervertebral disc microenvironment on the cell phenotype, issues associated with cell culture and technical preparation of cell products, as well as discussing current regulatory requirements. There are advantages and disadvantages of each proposed cell type, but no strong evidence to favour any one particular cell source at the moment.
Collapse
|
41
|
Wang YK, Chen CS. Cell adhesion and mechanical stimulation in the regulation of mesenchymal stem cell differentiation. J Cell Mol Med 2013; 17:823-32. [PMID: 23672518 PMCID: PMC3741348 DOI: 10.1111/jcmm.12061] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 03/01/2013] [Indexed: 12/15/2022] Open
Abstract
Stem cells have been shown to have the potential to provide a source of cells for applications to tissue engineering and organ repair. The mechanisms that regulate stem cell fate, however, mostly remain unclear. Mesenchymal stem cells (MSCs) are multipotent progenitor cells that are isolated from bone marrow and other adult tissues, and can be differentiated into multiple cell lineages, such as bone, cartilage, fat, muscles and neurons. Although previous studies have focused intensively on the effects of chemical signals that regulate MSC commitment, the effects of physical/mechanical cues of the microenvironment on MSC fate determination have long been neglected. However, several studies provided evidence that mechanical signals, both direct and indirect, played important roles in regulating a stem cell fate. In this review, we summarize a number of recent studies on how cell adhesion and mechanical cues influence the differentiation of MSCs into specific lineages. Understanding how chemical and mechanical cues in the microenvironment orchestrate stem cell differentiation may provide new insights into ways to improve our techniques in cell therapy and organ repair.
Collapse
Affiliation(s)
- Yang-Kao Wang
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan.
| | | |
Collapse
|
42
|
Wang Y, Wang J, Bai D, Song J, Ye R, Zhao Z, Lei L, Hao J, Jiang C, Fang S, An S, Cheng Q, Li J. Cell proliferation is promoted by compressive stress during early stage of chondrogenic differentiation of rat BMSCs. J Cell Physiol 2013; 228:1935-42. [DOI: 10.1002/jcp.24359] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 03/11/2013] [Indexed: 12/25/2022]
|
43
|
Ko CL, Tien YC, Wang JC, Chen WC. Characterization of controlled highly porous hyaluronan/gelatin cross-linking sponges for tissue engineering. J Mech Behav Biomed Mater 2012; 14:227-38. [DOI: 10.1016/j.jmbbm.2012.06.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/01/2012] [Accepted: 06/05/2012] [Indexed: 12/28/2022]
|
44
|
Nishimura R, Wakabayashi M, Hata K, Matsubara T, Honma S, Wakisaka S, Kiyonari H, Shioi G, Yamaguchi A, Tsumaki N, Akiyama H, Yoneda T. Osterix regulates calcification and degradation of chondrogenic matrices through matrix metalloproteinase 13 (MMP13) expression in association with transcription factor Runx2 during endochondral ossification. J Biol Chem 2012; 287:33179-90. [PMID: 22869368 PMCID: PMC3460424 DOI: 10.1074/jbc.m111.337063] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 07/27/2012] [Indexed: 11/06/2022] Open
Abstract
Endochondral ossification is temporally and spatially regulated by several critical transcription factors, including Sox9, Runx2, and Runx3. Although the molecular mechanisms that control the late stages of endochondral ossification (e.g. calcification) are physiologically and pathologically important, these precise regulatory mechanisms remain unclear. Here, we demonstrate that Osterix is an essential transcription factor for endochondral ossification that functions downstream of Runx2. The global and conditional Osterix-deficient mice studied here exhibited a defect of cartilage-matrix ossification and matrix vesicle formation. Importantly, Osterix deficiencies caused the arrest of endochondral ossification at the hypertrophic stage. Microarray analysis revealed that matrix metallopeptidase 13 (MMP13) is an important target of Osterix. We also showed that there exists a physical interaction between Osterix and Runx2 and that these proteins function cooperatively to induce MMP13 during chondrocyte differentiation. Most interestingly, the introduction of MMP13 stimulated the calcification of matrices in Osterix-deficient mouse limb bud cells. Our results demonstrated that Osterix was essential to endochondral ossification and revealed that the physical and functional interaction between Osterix and Runx2 were necessary for the induction of MMP13 during endochondral ossification.
Collapse
Affiliation(s)
- Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rutgers M, Saris DB, Vonk LA, van Rijen MH, Akrum V, Langeveld D, van Boxtel A, Dhert WJ, Creemers LB. Effect of collagen type I or type II on chondrogenesis by cultured human articular chondrocytes. Tissue Eng Part A 2012; 19:59-65. [PMID: 22861168 DOI: 10.1089/ten.tea.2011.0416] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTION Current cartilage repair procedures using autologous chondrocytes rely on a variety of carriers for implantation. Collagen types I and II are frequently used and valuable properties of both were shown earlier in vitro, although a preference for either was not demonstrated. Recently, however, fibrillar collagens were shown to promote cartilage degradation. The goal of this study was to evaluate the effects of collagen type I and type II coating on chondrogenic properties of in vitro cultured human chondrocytes, and to investigate if collagen-mediated cartilage degradation occurs. METHODS Human chondrocytes of eight healthy cartilage donors were isolated, expanded, and cultured on culture well inserts coated with either collagen type I, type II, or no coating (control). After 28 days of redifferentiation culture, safranin O and immunohistochemical staining for collagen types I, II, X, and Runx2/Cbfa1 were performed and glycosaminoglycan (GAG) and DNA content and release were examined. Further, expression of collagen type I, type II, type X, MMP13, Runx2/Cbfa1, DDR2, α2 and β1 integrin were examined by reverse transcriptase-polymerase chain reaction. RESULTS The matrix, created by chondrocytes grown on collagen type I- and II-coated membranes, resembled cartilage more than when grown on noncoated membranes as reflected by histological scoring. Immunohistochemical staining did not differ between the conditions. GAG content as well as GAG/DNA were higher for collagen type II-coated cartilage constructs than control. GAG release was also higher on collagen type I- and II-coated constructs. Expression of collagen type X was higher of chondrocytes grown on collagen type II compared to controls, but no collagen X protein could be demonstrated by immunohistochemistry. No effects of collagen coating on DDR2 nor MMP-13 gene expression were found. No differences were observed between collagen types I and II. CONCLUSION Chondrocyte culture on collagen type I or II promotes more active matrix production and turnover. No significant differences between collagen types I and II were observed, nor were hypertrophic changes more evident in either condition. The use of collagen type I or II coating for in vitro models, thus, seems a sound basis for in vivo repair procedures.
Collapse
Affiliation(s)
- Marijn Rutgers
- Orthopaedics Department, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Grünbaum T, Cloutier R, Vincent B. Dynamic skeletogenesis in fishes: Insight of exercise training on developmental plasticity. Dev Dyn 2012; 241:1507-24. [DOI: 10.1002/dvdy.23837] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2012] [Indexed: 11/10/2022] Open
|
47
|
Responte DJ, Lee JK, Hu JC, Athanasiou KA. Biomechanics-driven chondrogenesis: from embryo to adult. FASEB J 2012; 26:3614-24. [PMID: 22673579 DOI: 10.1096/fj.12-207241] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Biomechanics plays a pivotal role in articular cartilage development, pathophysiology, and regeneration. During embryogenesis and cartilage maturation, mechanical stimuli promote chondrogenesis and limb formation. Mechanical loading, which has been characterized using computer modeling and in vivo studies, is crucial for maintaining the phenotype of cartilage. However, excessive or insufficient loading has deleterious effects and promotes the onset of cartilage degeneration. Informed by the prominent role of biomechanics, mechanical stimuli have been harnessed to enhance redifferentiation of chondrocytes and chondroinduction of other cell types, thus providing new chondrocyte cell sources. Biomechanical stimuli, such as hydrostatic pressure or compression, have been used to enhance the functional properties of neocartilage. By identifying pathways involved in mechanical stimulation, chemical equivalents that mimic mechanical signaling are beginning to offer exciting new methods for improving neocartilage. Harnessing biomechanics to improve differentiation, maintenance, and regeneration is emerging as pivotal toward producing functional neocartilage that could eventually be used to treat cartilage degeneration.
Collapse
Affiliation(s)
- Donald J Responte
- Department of Biomedical Engineering, University of California-Davis, Davis, California 95616, USA
| | | | | | | |
Collapse
|
48
|
Ge Z, Li C, Heng BC, Cao G, Yang Z. Functional biomaterials for cartilage regeneration. J Biomed Mater Res A 2012; 100:2526-36. [PMID: 22492677 DOI: 10.1002/jbm.a.34147] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 01/30/2012] [Accepted: 02/19/2012] [Indexed: 12/22/2022]
Abstract
The injury and degeneration of articular cartilage and associated arthritis are leading causes of disability worldwide. Cartilage tissue engineering as a treatment modality for cartilage defects has been investigated for over 20 years. Various scaffold materials have been developed for this purpose, but has yet to achieve feasibility and effectiveness for widespread clinical use. Currently, the regeneration of articular cartilage remains a formidable challenge, due to the complex physiology of cartilage tissue and its poor healing capacity. Although intensive research has been focused on the developmental biology and regeneration of cartilage tissue and a diverse plethora of biomaterials have been developed for this purpose, cartilage regeneration is still suboptimal, such as lacking a layered structure, mechanical mismatch with native cartilage and inadequate integration between native tissue and implanted scaffold. The ideal scaffold material should have versatile properties that actively contribute to cartilage regeneration. Functional scaffold materials may overcome the various challenges faced in cartilage tissue engineering by providing essential biological, mechanical, and physical/chemical signaling cues through innovative design. This review thus focuses on the complex structure of native articular cartilage, the critical properties of scaffolds required for cartilage regeneration, present strategies for scaffold design, and future directions for cartilage regeneration with functional scaffold materials.
Collapse
Affiliation(s)
- Zigang Ge
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, People's Republic of China.
| | | | | | | | | |
Collapse
|
49
|
Takahashi I, Hoso M, Matsuzaki T. Histopathological Effects of Loading on Cartilage Repair in a Rat Full-thickness Articular Cartilage Defect Model. J Phys Ther Sci 2012. [DOI: 10.1589/jpts.24.1187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Ikufumi Takahashi
- Department of Rehabilitation, Houju Memorial Hospital
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University
| | - Masahiro Hoso
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University
| | - Taro Matsuzaki
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University
| |
Collapse
|
50
|
Michalopoulos E, Knight RL, Korossis S, Kearney JN, Fisher J, Ingham E. Development of methods for studying the differentiation of human mesenchymal stem cells under cyclic compressive strain. Tissue Eng Part C Methods 2011; 18:252-62. [PMID: 22047076 DOI: 10.1089/ten.tec.2011.0347] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human mesenchymal stem cells (hMSC) have numerous potential advantages over terminally differentiated cells and embryonic stem cells for use in tissue engineering applications. The aims of this study were to develop methods to test the hypothesis that hMSC could be differentiated using cyclic compressive strain alone. hMSC were successfully isolated, purified using D7-FIB antibody, cloned, and characterized. The cells were subsequently analyzed using fluorescence-activated cell sorting using a panel of antibodies and differentiation into multiple cell lineages. D7FIB-positive cells were then seeded into collagen-alginate scaffolds and subjected to 10% or 15% cyclic compressive strain for 4 out of 24 hours for up to 21 days in a bespoke servo-assisted displacement-controlled device. Cells were analyzed using adenosine triphosphate assay to determine cell number, live-dead cell assay, and quantitative real-time polymerase chain reaction at 7 and 21 days. Cloned D7-FIB-positive hMSCs showed evidence of differentiation to an osteogenic lineage under 10% cyclic compressive strain alone (core binding factor alpha 1 (CBFA-1) was significantly upregulated at 7 and 21 days by a factor of 18.3 and 32.2, respectively) and to an osteo-chondrogenic lineage under 15% cyclic compressive strain alone (increased expression of CBFA-1, Sox9, and aggrecan). A combination of a composite viscoelastic scaffold and controlled cyclic compressive strain may be useful for study of the differentiation of MSC.
Collapse
Affiliation(s)
- Efstathios Michalopoulos
- Faculty of Biological Sciences, Institute of Medical & Biological Engineering, University of Leeds, Leeds, United Kingdom
| | | | | | | | | | | |
Collapse
|