1
|
Wells LM, Roberts HC, Luyten FP, Roberts SJ. Identifying Fibroblast Growth Factor Receptor 3 as a Mediator of Periosteal Osteochondral Differentiation through the Construction of microRNA-Based Interaction Networks. BIOLOGY 2023; 12:1381. [PMID: 37997980 PMCID: PMC10669632 DOI: 10.3390/biology12111381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023]
Abstract
Human periosteum-derived progenitor cells (hPDCs) have the ability to differentiate towards both the chondrogenic and osteogenic lineages. This coordinated and complex osteochondrogenic differentiation process permits endochondral ossification and is essential in bone development and repair. We have previously shown that humanised cultures of hPDCs enhance their osteochondrogenic potentials in vitro and in vivo; however, the underlying mechanisms are largely unknown. This study aimed to identify novel regulators of hPDC osteochondrogenic differentiation through the construction of miRNA-mRNA regulatory networks derived from hPDCs cultured in human serum or foetal bovine serum as an alternative in silico strategy to serum characterisation. Sixteen differentially expressed miRNAs (DEMis) were identified in the humanised culture. In silico analysis of the DEMis with TargetScan allowed for the identification of 1503 potential miRNA target genes. Upon comparison with a paired RNAseq dataset, a 4.5% overlap was observed (122 genes). A protein-protein interaction network created with STRING interestingly identified FGFR3 as a key network node, which was further predicted using multiple pathway analyses. Functional analysis revealed that hPDCs with the activating mutation FGFR3N540K displayed increased expressions of chondrogenic gene markers when cultured under chondrogenic conditions in vitro and displayed enhanced endochondral bone formation in vivo. A further histological analysis uncovered known downstream mediators involved in FGFR3 signalling and endochondral ossification to be upregulated in hPDC FGFR3N540K-seeded implants. This combinational approach of miRNA-mRNA-protein network analysis with in vitro and in vivo characterisation has permitted the identification of FGFR3 as a novel mediator of hPDC biology. Furthermore, this miRNA-based workflow may also allow for the identification of drug targets, which may be of relevance in instances of delayed fracture repair.
Collapse
Affiliation(s)
- Leah M. Wells
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London NW1 0TU, UK;
| | - Helen C. Roberts
- Department of Natural Sciences, Middlesex University, London NW4 4BT, UK;
| | - Frank P. Luyten
- Skeletal Biology and Engineering Research Centre (SBE), KU Leuven, 3000 Leuven, Belgium;
| | - Scott J. Roberts
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London NW1 0TU, UK;
| |
Collapse
|
2
|
Mangiavini L, Peretti GM, Canciani B, Maffulli N. Epidermal growth factor signalling pathway in endochondral ossification: an evidence-based narrative review. Ann Med 2022; 54:37-50. [PMID: 34955078 PMCID: PMC8725985 DOI: 10.1080/07853890.2021.2015798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
During endochondral bone development, a complex process that leads to the formation of the majority of skeletal elements, mesenchymal cells condense, differentiating into chondrocytes and producing the foetal growth plate. Chondrocytes progressively hypertrophy, induce angiogenesis and are then gradually replaced by bone. Epidermal Growth Factor (EGF), one of many growth factors, is the prototype of the EGF-ligand family, which comprises several proteins involved in cell proliferation, migration and survival. In bone, EGF pathway signalling finely tunes the first steps of chondrogenesis by maintaining mesenchymal cells in an undifferentiated stage, and by promoting hypertrophic cartilage replacement. Moreover, EGF signalling modulates bone homeostasis by stimulating osteoblast and osteoclast proliferation, and by regulating osteoblast differentiation under specific spatial and temporal conditions. This evidence-based narrative review describes the EGF pathway in bone metabolism and endochondral bone development. This comprehensive description may be useful in light of possible clinical applications in orthopaedic practice. A deeper knowledge of the role of EGF in bone may be useful in musculoskeletal conditions which may benefit from the modulation of this signalling pathway.Key messagesThe EGF pathway is involved in bone metabolism.EGF signalling is essential in the very early stages of limb development by maintaining cells in an undifferentiated stage.EGF pathway positively regulates chondrocyte proliferation, negatively modulates hypertrophy, and favours cartilage replacement by bone.EGF and EGF-like proteins finely tune the proliferation and differentiation of bone tissue cells, and they also regulate the initial phases of endochondral ossification.
Collapse
Affiliation(s)
- L Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - G M Peretti
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - B Canciani
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - N Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy.,Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK.,School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent, UK
| |
Collapse
|
3
|
Daneshmandi L, Holt BD, Arnold AM, Laurencin CT, Sydlik SA. Ultra-low binder content 3D printed calcium phosphate graphene scaffolds as resorbable, osteoinductive matrices that support bone formation in vivo. Sci Rep 2022; 12:6960. [PMID: 35484292 PMCID: PMC9050648 DOI: 10.1038/s41598-022-10603-3] [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] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/02/2022] [Indexed: 12/18/2022] Open
Abstract
Bone regenerative engineering could replace autografts; however, no synthetic material fulfills all design criteria. Nanocarbons incorporated into three-dimensional printed (3DP) matrices can improve properties, but incorporation is constrained to low wt%. Further, unmodified nanocarbons have limited osteogenic potential. Functionalization to calcium phosphate graphene (CaPG) imparts osteoinductivity and osteoconductivity, but loading into matrices remained limited. This work presents ultra-high content (90%), 3DP-CaPG matrices. 3DP-CaPG matrices are highly porous (95%), moderately stiff (3 MPa), and mechanically robust. In vitro, they are cytocompatible and induce osteogenic differentiation of human mesenchymal stem cells (hMSCs), indicated by alkaline phosphatase, mineralization, and COL1α1 expression. In vivo, bone regeneration was studied using a transgenic fluorescent-reporter mouse non-union calvarial defect model. 3DP-CaPG stimulates cellular ingrowth, retains donor cells, and induces osteogenic differentiation. Histology shows TRAP staining around struts, suggesting potential osteoclast activity. Apparent resorption of 3DP-CaPG was observed and presented no toxicity. 3DP-CaPG represents an advancement towards a synthetic bone regeneration matrix.
Collapse
Affiliation(s)
- Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
| | - Brian D Holt
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Anne M Arnold
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA
- National Security Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, 06030, USA.
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA.
- Department of Material Science and Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Stefanie A Sydlik
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
4
|
Basu B, Gowtham N, Xiao Y, Kalidindi SR, Leong KW. Biomaterialomics: Data science-driven pathways to develop fourth-generation biomaterials. Acta Biomater 2022; 143:1-25. [PMID: 35202854 DOI: 10.1016/j.actbio.2022.02.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
Abstract
Conventional approaches to developing biomaterials and implants require intuitive tailoring of manufacturing protocols and biocompatibility assessment. This leads to longer development cycles, and high costs. To meet existing and unmet clinical needs, it is critical to accelerate the production of implantable biomaterials, implants and biomedical devices. Building on the Materials Genome Initiative, we define the concept 'biomaterialomics' as the integration of multi-omics data and high-dimensional analysis with artificial intelligence (AI) tools throughout the entire pipeline of biomaterials development. The Data Science-driven approach is envisioned to bring together on a single platform, the computational tools, databases, experimental methods, machine learning, and advanced manufacturing (e.g., 3D printing) to develop the fourth-generation biomaterials and implants, whose clinical performance will be predicted using 'digital twins'. While analysing the key elements of the concept of 'biomaterialomics', significant emphasis has been put forward to effectively utilize high-throughput biocompatibility data together with multiscale physics-based models, E-platform/online databases of clinical studies, data science approaches, including metadata management, AI/ Machine Learning (ML) algorithms and uncertainty predictions. Such integrated formulation will allow one to adopt cross-disciplinary approaches to establish processing-structure-property (PSP) linkages. A few published studies from the lead author's research group serve as representative examples to illustrate the formulation and relevance of the 'Biomaterialomics' approaches for three emerging research themes, i.e. patient-specific implants, additive manufacturing, and bioelectronic medicine. The increased adaptability of AI/ML tools in biomaterials science along with the training of the next generation researchers in data science are strongly recommended. STATEMENT OF SIGNIFICANCE: This leading opinion review paper emphasizes the need to integrate the concepts and algorithms of the data science with biomaterials science. Also, this paper emphasizes the need to establish a mathematically rigorous cross-disciplinary framework that will allow a systematic quantitative exploration and curation of critical biomaterials knowledge needed to drive objectively the innovation efforts within a suitable uncertainty quantification framework, as embodied in 'biomaterialomics' concept, which integrates multi-omics data and high-dimensional analysis with artificial intelligence (AI) tools, like machine learning. The formulation of this approach has been demonstrated for patient-specific implants, additive manufacturing, and bioelectronic medicine.
Collapse
|
5
|
Xie C, Ye J, Liang R, Yao X, Wu X, Koh Y, Wei W, Zhang X, Ouyang H. Advanced Strategies of Biomimetic Tissue-Engineered Grafts for Bone Regeneration. Adv Healthc Mater 2021; 10:e2100408. [PMID: 33949147 DOI: 10.1002/adhm.202100408] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/16/2021] [Indexed: 12/21/2022]
Abstract
The failure to repair critical-sized bone defects often leads to incomplete regeneration or fracture non-union. Tissue-engineered grafts have been recognized as an alternative strategy for bone regeneration due to their potential to repair defects. To design a successful tissue-engineered graft requires the understanding of physicochemical optimization to mimic the composition and structure of native bone, as well as the biological strategies of mimicking the key biological elements during bone regeneration process. This review provides an overview of engineered graft-based strategies focusing on physicochemical properties of materials and graft structure optimization from macroscale to nanoscale to further boost bone regeneration, and it summarizes biological strategies which mainly focus on growth factors following bone regeneration pattern and stem cell-based strategies for more efficient repair. Finally, it discusses the current limitations of existing strategies upon bone repair and highlights a promising strategy for rapid bone regeneration.
Collapse
Affiliation(s)
- Chang Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
| | - Jinchun Ye
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Xudong Yao
- The Fourth Affiliated Hospital Zhejiang University School of Medicine Yiwu 322000 China
| | - Xinyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Yiwen Koh
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Wei Wei
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| | - Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| |
Collapse
|
6
|
Bone Morphogenetic Proteins, Carriers, and Animal Models in the Development of Novel Bone Regenerative Therapies. MATERIALS 2021; 14:ma14133513. [PMID: 34202501 PMCID: PMC8269575 DOI: 10.3390/ma14133513] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Abstract
Bone morphogenetic proteins (BMPs) possess a unique ability to induce new bone formation. Numerous preclinical studies have been conducted to develop novel, BMP-based osteoinductive devices for the management of segmental bone defects and posterolateral spinal fusion (PLF). In these studies, BMPs were combined with a broad range of carriers (natural and synthetic polymers, inorganic materials, and their combinations) and tested in various models in mice, rats, rabbits, dogs, sheep, and non-human primates. In this review, we summarized bone regeneration strategies and animal models used for the initial, intermediate, and advanced evaluation of promising therapeutical solutions for new bone formation and repair. Moreover, in this review, we discuss basic aspects to be considered when planning animal experiments, including anatomical characteristics of the species used, appropriate BMP dosing, duration of the observation period, and sample size.
Collapse
|
7
|
Shah M, Maroof A, Gikas P, Mittal G, Keen R, Baeten D, Shaw S, Roberts SJ. Dual neutralisation of IL-17F and IL-17A with bimekizumab blocks inflammation-driven osteogenic differentiation of human periosteal cells. RMD Open 2021; 6:rmdopen-2020-001306. [PMID: 32723833 PMCID: PMC7722278 DOI: 10.1136/rmdopen-2020-001306] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/25/2020] [Accepted: 06/25/2020] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES Interleukin (IL)-17 signalling has been shown to be a key regulator of disease in ankylosing spondylitis (AS) with several IL-17 blockers currently clinically approved. Despite this, the role of IL-17 in bone pathology is poorly understood. This study aimed to investigate IL-17 signalling in the context of pathological bone formation. METHODS A biomimetic human periosteum-derived cell (hPDC) model of osteogenic differentiation was used in combination with recombinant IL-17 cytokines, T-cell supernatants or serum from patients with AS. IL-17A, IL-17F and bimekizumab monoclonal antibodies were used to block IL-17 cytokine action. RESULTS Recombinant IL-17A and IL-17F are pro-osteogenic with respect to hPDC differentiation. T helper 17 or γδ-T cell supernatants also potently stimulated in vitro bone formation, which was blocked deeper by dual inhibition of IL-17A and IL-17F than by neutralisation of IL-17A or IL-17F individually. Osteogenic blockade may be due to an increase in expression of the Wnt antagonist DKK1. Interestingly, osteocommitment was also induced by serum obtained from patients with AS, which was also abrogated by dual neutralisation of IL-17A and IL-17F. CONCLUSIONS These data show for the first time that IL-17A and IL-17F enhance in vitro osteogenic differentiation and bone formation from hPDCs, inhibition of which may offer an attractive therapeutic strategy to prevent pathological bone formation.
Collapse
Affiliation(s)
- Mittal Shah
- UCB Pharma, Slough, UK.,Division of Surgery and Interventional Science, University College London, London, UK
| | | | - Panos Gikas
- Royal National Orthopaedic Hospital Stanmore, Stanmore, UK
| | - Gayatri Mittal
- Royal National Orthopaedic Hospital Stanmore, Stanmore, UK
| | - Richard Keen
- Royal National Orthopaedic Hospital Stanmore, Stanmore, UK
| | | | | | - Scott J Roberts
- UCB Pharma, Slough, UK .,Division of Surgery and Interventional Science, University College London, London, UK.,Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
8
|
A Review of Recent Developments in the Molecular Mechanisms of Bone Healing. Int J Mol Sci 2021; 22:ijms22020767. [PMID: 33466612 PMCID: PMC7828700 DOI: 10.3390/ijms22020767] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Between 5 and 10 percent of fractures do not heal, a condition known as nonunion. In clinical practice, stable fracture fixation associated with autologous iliac crest bone graft placement is the gold standard for treatment. However, some recalcitrant nonunions do not resolve satisfactorily with this technique. For these cases, biological alternatives are sought based on the molecular mechanisms of bone healing, whose most recent findings are reviewed in this article. The pro-osteogenic efficacy of morin (a pale yellow crystalline flavonoid pigment found in old fustic and osage orange trees) has recently been reported, and the combined use of bone morphogenetic protein-9 (BMP9) and leptin might improve fracture healing. Inhibition with methyl-piperidino-pyrazole of estrogen receptor alpha signaling delays bone regeneration. Smoking causes a chondrogenic disorder, aberrant activity of the skeleton’s stem and progenitor cells, and an intense initial inflammatory response. Smoking cessation 4 weeks before surgery is therefore highly recommended. The delay in fracture consolidation in diabetic animals is related to BMP6 deficiency (35 kDa). The combination of bioceramics and expanded autologous human mesenchymal stem cells from bone marrow is a new and encouraging alternative for treating recalcitrant nonunions.
Collapse
|
9
|
Al Hosni R, Shah M, Cheema U, Roberts HC, Luyten FP, Roberts SJ. Mapping human serum-induced gene networks as a basis for the creation of biomimetic periosteum for bone repair. Cytotherapy 2020; 22:424-435. [PMID: 32522398 DOI: 10.1016/j.jcyt.2020.03.434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/21/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The periosteum is a highly vascularized, collagen-rich tissue that plays a crucial role in directing bone repair. This is orchestrated primarily by its resident progenitor cell population. Indeed, preservation of periosteum integrity is critical for bone healing. Cells extracted from the periosteum retain their osteochondrogenic properties and as such are a promising basis for tissue engineering strategies for the repair of bone defects. However, the culture expansion conditions and the way in which the cells are reintroduced to the defect site are critical aspects of successful translation. Indeed, expansion in human serum and implantation on biomimetic materials has previously been shown to improve in vivo bone formation. AIM This study aimed to develop a protocol to allow for the expansion of human periosteum derived cells (hPDCs) in a biomimetic periosteal-like environment. METHODS The expansion conditions were defined through the investigation of the bioactive cues involved in augmenting hPDC proliferative and multipotency characteristics, based on transcriptomic analysis of cells cultured in human serum. RESULTS Master regulators of transcriptional networks were identified, and an optimized periosteum-derived growth factor cocktail (PD-GFC; containing β-estradiol, FGF2, TNFα, TGFβ, IGF-1 and PDGF-BB) was generated. Expansion of hPDCs in PD-GFC resulted in serum mimicry with regard to the cell morphology, proliferative capacity and chondrogenic differentiation. When incorporated into a three-dimensional collagen type 1 matrix and cultured in PD-GFC, the hPDCs migrated to the surface that represented the matrix topography of the periosteum cambium layer. Furthermore, gene expression analysis revealed a down-regulated WNT and TGFβ signature and an up-regulation of CREB, which may indicate the hPDCs are recreating their progenitor cell signature. CONCLUSION This study highlights the first stage in the development of a biomimetic periosteum, which may have applications in bone repair.
Collapse
Affiliation(s)
- Rawiya Al Hosni
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Mittal Shah
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Umber Cheema
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Helen C Roberts
- Department of Natural Sciences, Faculty of Science & Technology, Middlesex University, London, UK
| | - Frank P Luyten
- Skeletal Biology and Tissue Engineering Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium and
| | - Scott J Roberts
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK; Skeletal Biology and Tissue Engineering Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium and; Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK.
| |
Collapse
|
10
|
Gandolfi MG, Gardin C, Zamparini F, Ferroni L, Esposti MD, Parchi G, Ercan B, Manzoli L, Fava F, Fabbri P, Prati C, Zavan B. Mineral-Doped Poly(L-lactide) Acid Scaffolds Enriched with Exosomes Improve Osteogenic Commitment of Human Adipose-Derived Mesenchymal Stem Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E432. [PMID: 32121340 PMCID: PMC7153699 DOI: 10.3390/nano10030432] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/05/2020] [Accepted: 02/22/2020] [Indexed: 12/18/2022]
Abstract
Exosomes derived from mesenchymal stem cells are extracellular vesicles released to facilitate cell communication and function. Recently, polylactic acid (PLA), calcium silicates (CaSi), and dicalcium phosphate dihydrate (DCPD) have been used to produce bioresorbable functional mineral-doped porous scaffolds-through thermally induced phase separation technique, as materials for bone regeneration. The aim of this study was to investigate the effect of mineral-doped PLA-based porous scaffolds enriched with exosome vesicles (EVs) on osteogenic commitment of human adipose mesenchymal stem cells (hAD-MSCs). Two different mineral-doped scaffolds were produced: PLA-10CaSi-10DCPD and PLA-5CaSi-5DCPD. Scaffolds surface micromorphology was investigated by ESEM-EDX before and after 28 days immersion in simulated body fluid (HBSS). Exosomes were deposited on the surface of the scaffolds and the effect of exosome-enriched scaffolds on osteogenic commitment of hAD-MSCs cultured in proximity of the scaffolds has been evaluated by real time PCR. In addition, the biocompatibility was evaluated by direct-contact seeding hAD-MSCs on scaffolds surface-using MTT viability test. In both formulations, ESEM showed pores similar in shape (circular and elliptic) and size (from 10-30 µm diameter). The porosity of the scaffolds decreased after 28 days immersion in simulated body fluid. Mineral-doped scaffolds showed a dynamic surface and created a suitable bone-forming microenvironment. The presence of the mineral fillers increased the osteogenic commitment of hAD-MSCs. Exosomes were easily entrapped on the surface of the scaffolds and their presence improved gene expression of major markers of osteogenesis such as collagen type I, osteopontin, osteonectin, osteocalcin. The experimental scaffolds enriched with exosomes, in particular PLA-10CaSi-10DCPD, increased the osteogenic commitment of MSCs. In conclusion, the enrichment of bioresorbable functional scaffolds with exosomes is confirmed as a potential strategy to improve bone regeneration procedures.
Collapse
Affiliation(s)
- Maria Giovanna Gandolfi
- Laboratory of Biomaterials and Oral Pathology, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40125 Bologna, Italy
| | - Chiara Gardin
- Medical Sciences Department, University of Ferrara, 44100 Ferrara, Italy
| | - Fausto Zamparini
- Laboratory of Biomaterials and Oral Pathology, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40125 Bologna, Italy
| | - Letizia Ferroni
- Medical Sciences Department, University of Ferrara, 44100 Ferrara, Italy
| | - Micaela Degli Esposti
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, 40136 Bologna, Italy
| | - Greta Parchi
- Laboratory of Biomaterials and Oral Pathology, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40125 Bologna, Italy
| | - Batur Ercan
- Department of Metallurgical and Materials Engineering, 06800 Ankara, Turkey
| | - Lucia Manzoli
- Cellular Signaling Laboratory, Institute of Human Anatomy, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Fabio Fava
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, 40136 Bologna, Italy
| | - Paola Fabbri
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, 40136 Bologna, Italy
| | - Carlo Prati
- Endodontic Clinical Section, School of Dentistry, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40125 Bologna, Italy
| | - Barbara Zavan
- Medical Sciences Department, University of Ferrara, 44100 Ferrara, Italy
| |
Collapse
|
11
|
Bolander J, Herpelinck T, Chaklader M, Gklava C, Geris L, Luyten FP. Single-cell characterization and metabolic profiling of in vitro cultured human skeletal progenitors with enhanced in vivo bone forming capacity. Stem Cells Transl Med 2019; 9:389-402. [PMID: 31738481 PMCID: PMC7031650 DOI: 10.1002/sctm.19-0151] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/19/2019] [Accepted: 10/05/2019] [Indexed: 12/15/2022] Open
Abstract
Cell populations and their interplay provide the basis of a cell‐based regenerative construct. Serum‐free preconditioning can overcome the less predictable behavior of serum expanded progenitor cells, but the underlying mechanism and how this is reflected in vivo remains unknown. Herein, the cellular and molecular changes associated with a cellular phenotype shift induced by serum‐free preconditioning of human periosteum‐derived cells were investigated. Following BMP‐2 stimulation, preconditioned cells displayed enhanced in vivo bone forming capacity, associated with an adapted cellular metabolism together with an elevated expression of BMPR2. Single‐cell RNA sequencing confirmed the activation of pathways and transcriptional regulators involved in bone development and fracture healing, providing support for the augmentation of specified skeletal progenitor cell populations. The reported findings illustrate the importance of appropriate in vitro conditions for the in vivo outcome. In addition, BMPR2 represents a promising biomarker for the enrichment of skeletal progenitor cells for in vivo bone regeneration.
Collapse
Affiliation(s)
- Johanna Bolander
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Tim Herpelinck
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Malay Chaklader
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Charikleia Gklava
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico medicine, University of Liege, Liège, Belgium
| | - Frank P Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Gupta P, Hall GN, Geris L, Luyten FP, Papantoniou I. Human Platelet Lysate Improves Bone Forming Potential of Human Progenitor Cells Expanded in Microcarrier-Based Dynamic Culture. Stem Cells Transl Med 2019; 8:810-821. [PMID: 31038850 PMCID: PMC6646698 DOI: 10.1002/sctm.18-0216] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/19/2019] [Indexed: 12/22/2022] Open
Abstract
Xenogeneic‐free media are required for translating advanced therapeutic medicinal products to the clinics. In addition, process efficiency is crucial for ensuring cost efficiency, especially when considering large‐scale production of mesenchymal stem cells (MSCs). Human platelet lysate (HPL) has been increasingly adopted as an alternative for fetal bovine serum (FBS) for MSCs. However, its therapeutic and regenerative potential in vivo is largely unexplored. Herein, we compare the effects of FBS and HPL supplementation for a scalable, microcarrier‐based dynamic expansion of human periosteum‐derived cells (hPDCs) while assessing their bone forming capacity by subcutaneous implantation in small animal model. We observed that HPL resulted in faster cell proliferation with a total fold increase of 5.2 ± 0.61 in comparison to 2.7 ± 02.22‐fold in FBS. Cell viability and trilineage differentiation capability were maintained by HPL, although a suppression of adipogenic differentiation potential was observed. Differences in mRNA expression profiles were also observed between the two on several markers. When implanted, we observed a significant difference between the bone forming capacity of cells expanded in FBS and HPL, with HPL supplementation resulting in almost three times more mineralized tissue within calcium phosphate scaffolds. FBS‐expanded cells resulted in a fibrous tissue structure, whereas HPL resulted in mineralized tissue formation, which can be classified as newly formed bone, verified by μCT and histological analysis. We also observed the presence of blood vessels in our explants. In conclusion, we suggest that replacing FBS with HPL in bioreactor‐based expansion of hPDCs is an optimal solution that increases expansion efficiency along with promoting bone forming capacity of these cells. stem cells translational medicine2019;8:810&821
Collapse
Affiliation(s)
- Priyanka Gupta
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Gabriella Nilsson Hall
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA-R In Silico Medicine, Université de Liege, Liège, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Frank P Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Ebrahimi M, Botelho M, Lu W, Monmaturapoj N. Synthesis and characterization of biomimetic bioceramic nanoparticles with optimized physicochemical properties for bone tissue engineering. J Biomed Mater Res A 2019; 107:1654-1666. [PMID: 30916848 DOI: 10.1002/jbm.a.36681] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/10/2019] [Accepted: 03/15/2019] [Indexed: 02/06/2023]
Abstract
Calcium phosphate bioceramics nanoparticles such as nano-hydroxyapatite (nHA) and nano-tricalcium phosphate (nTCP) are the main focus of basic and applied research for bone tissue regeneration. In particular, a combination of these two phases (nHA + nTCP) which refers to as "nano-biphasic calcium phosphates (nBCP)" is of interest due to the preferred biodegradation nature compared to single-phase bioceramics. However, the available synthesis processes are challenging and the biomaterials properties are yet to be optimized to mimic the physiochemical properties of the natural nanoscale bone apatite. In this study, a new approach was developed for the production of optimized bioceramic nanoparticles aiming to improve their biomimecity for better biological performances. Nanoparticles were synthesized through a carefully controlled and modified wet mechano-chemical method combined with a controlled solid-state synthesis. Different processing variables have been analyzed including; milling parameters, post-synthesis treatment, and calcination phase. Detailed physicochemical characterizations of nanoparticles revealed higher crystallinity (∼100%), lower crystallite/particle size (58 nm), higher homogeneity, reduced particle agglomeration size (6 μm), and a closer molar ratio (1.8) to biological apatite compared to control and standard samples. Furthermore, the study group was confirmed as calcium-deficient carbonate-substituted BCP nanoparticles (nHA/nβ-TCP: 92/8%). As such, the introduced method can afford an easier and accurate control over nanoparticle physiochemical properties including the composition phase which can be used for better customization of biomaterials for clinical applications. The findings of this article will also help researchers in the further advancement of production strategies of biomaterials. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1654-1666, 2019.
Collapse
Affiliation(s)
- Mehdi Ebrahimi
- Prosthodontics, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong
| | - Michael Botelho
- Prosthodontics, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong
| | - William Lu
- Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Naruporn Monmaturapoj
- Polymer Unit, National Metal and Materials Technology Center (MTEC), NSTDA, Pathumthani, Thailand
| |
Collapse
|
14
|
Katagiri H, Mendes LF, Luyten FP. Reduction of BMP6‐induced bone formation by calcium phosphate in wild‐type compared with nude mice. J Tissue Eng Regen Med 2019; 13:846-856. [DOI: 10.1002/term.2837] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 12/01/2018] [Accepted: 02/13/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Hiroki Katagiri
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research CenterKatholieke Universiteit Leuven Leuven Belgium
- Prometheus, Division of Skeletal Tissue EngineeringKatholieke Universiteit Leuven Leuven Belgium
| | - Luis Filipe Mendes
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research CenterKatholieke Universiteit Leuven Leuven Belgium
- Prometheus, Division of Skeletal Tissue EngineeringKatholieke Universiteit Leuven Leuven Belgium
| | - Frank P. Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research CenterKatholieke Universiteit Leuven Leuven Belgium
- Prometheus, Division of Skeletal Tissue EngineeringKatholieke Universiteit Leuven Leuven Belgium
| |
Collapse
|
15
|
Ho-Shui-Ling A, Bolander J, Rustom LE, Johnson AW, Luyten FP, Picart C. Bone regeneration strategies: Engineered scaffolds, bioactive molecules and stem cells current stage and future perspectives. Biomaterials 2018; 180:143-162. [PMID: 30036727 PMCID: PMC6710094 DOI: 10.1016/j.biomaterials.2018.07.017] [Citation(s) in RCA: 540] [Impact Index Per Article: 77.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 12/25/2022]
Abstract
Bone fractures are the most common traumatic injuries in humans. The repair of bone fractures is a regenerative process that recapitulates many of the biological events of embryonic skeletal development. Most of the time it leads to successful healing and the recovery of the damaged bone. Unfortunately, about 5-10% of fractures will lead to delayed healing or non-union, more so in the case of co-morbidities such as diabetes. In this article, we review the different strategies to heal bone defects using synthetic bone graft substitutes, biologically active substances and stem cells. The majority of currently available reviews focus on strategies that are still at the early stages of development and use mostly in vitro experiments with cell lines or stem cells. Here, we focus on what is already implemented in the clinics, what is currently in clinical trials, and what has been tested in animal models. Treatment approaches can be classified in three major categories: i) synthetic bone graft substitutes (BGS) whose architecture and surface can be optimized; ii) BGS combined with bioactive molecules such as growth factors, peptides or small molecules targeting bone precursor cells, bone formation and metabolism; iii) cell-based strategies with progenitor cells combined or not with active molecules that can be injected or seeded on BGS for improved delivery. We review the major types of adult stromal cells (bone marrow, adipose and periosteum derived) that have been used and compare their properties. Finally, we discuss the remaining challenges that need to be addressed to significantly improve the healing of bone defects.
Collapse
Affiliation(s)
- Antalya Ho-Shui-Ling
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France; CNRS, LMGP, 3 Parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Johanna Bolander
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
| | - Laurence E Rustom
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 1304 West Springfield Avenue, Urbana, IL 61801, USA
| | - Amy Wagoner Johnson
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, 1206 West Green Street, Urbana, IL 61081, USA; Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, IL 61801, USA
| | - Frank P Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium.
| | - Catherine Picart
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France; CNRS, LMGP, 3 Parvis Louis Néel, 38031 Grenoble Cedex 01, France.
| |
Collapse
|
16
|
Tang Z, Li X, Tan Y, Fan H, Zhang X. The material and biological characteristics of osteoinductive calcium phosphate ceramics. Regen Biomater 2018; 5:43-59. [PMID: 29423267 PMCID: PMC5798025 DOI: 10.1093/rb/rbx024] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/16/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
The discovery of osteoinductivity of calcium phosphate (Ca-P) ceramics has set an enduring paradigm of conferring biological regenerative activity to materials with carefully designed structural characteristics. The unique phase composition and porous structural features of osteoinductive Ca-P ceramics allow it to interact with signaling molecules and extracellular matrices in the host system, creating a local environment conducive to new bone formation. Mounting evidence now indicate that the osteoinductive activity of Ca-P ceramics is linked to their physicochemical and three-dimensional structural properties. Inspired by this conceptual breakthrough, many laboratories have shown that other materials can be also enticed to join the rank of tissue-inducing biomaterials, and besides the bones, other tissues such as cartilage, nerves and blood vessels were also regenerated with the assistance of biomaterials. Here, we give a brief historical recount about the discovery of the osteoinductivity of Ca-P ceramics, summarize the underlying material factors and biological characteristics, and discuss the mechanism of osteoinduction concerning protein adsorption, and the interaction with different types of cells, and the involvement of the vascular and immune systems.
Collapse
Affiliation(s)
- Zhurong Tang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Yanfei Tan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| |
Collapse
|
17
|
Magnesium phosphate ceramics incorporating a novel indene compound promote osteoblast differentiation in vitro and bone regeneration in vivo. Biomaterials 2017; 157:51-61. [PMID: 29245051 DOI: 10.1016/j.biomaterials.2017.11.032] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/15/2017] [Accepted: 11/21/2017] [Indexed: 02/04/2023]
Abstract
Incorporating bioactive molecules into synthetic ceramic scaffolds is challenging. In this study, to enhance bone regeneration, a magnesium phosphate (MgP) ceramic scaffold was incorporated with a novel indene compound, KR-34893. KR-34893 induced the deposition of minerals and expression of osteoblast marker genes in primary human bone marrow mesenchymal stem cells (BMSCs) and a mouse osteoblastic MC3T3-E1 cell line. Analysis of the mode of action showed that KR-34893 induced the phosphorylation of MAPK/extracellular signal-regulated kinase and extracellular signal-regulated kinase, and subsequently the expression of bone morphogenetic protein 7, accompanied by SMAD1/5/8 phosphorylation. Accordingly, KR-34893 was incorporated into an MgP scaffold prepared by 3D printing at room temperature, followed by cement reaction. KR-34893-incorporated MgP (KR-MgP) induced the expression of osteoblast differentiation marker genes in vitro. In a rat calvaria defect model, KR-MgP scaffolds enhanced bone regeneration and increased bone volume compared with MgP scaffolds, as assessed by micro-computed tomography and histological analyses. In conclusion, we developed a method for producing osteoinductive MgP scaffolds incorporating a bioactive organic compound, without high temperature sintering. The KR-MgP scaffolds enhanced osteoblast activation in vitro and bone regeneration in vivo.
Collapse
|
18
|
Lv L, Tang Y, Zhang P, Liu Y, Bai X, Zhou Y. Biomaterial Cues Regulate Epigenetic State and Cell Functions-A Systematic Review. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:112-132. [PMID: 28903618 DOI: 10.1089/ten.teb.2017.0287] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Biomaterial cues can act as potent regulators of cell niche and microenvironment. Epigenetic regulation plays an important role in cell functions, including proliferation, differentiation, and reprogramming. It is now well appreciated that biomaterials can alter epigenetic states of cells. In this study, we systematically reviewed the underlying epigenetic mechanisms of how different biomaterial cues, including material chemistry, topography, elasticity, and mechanical stimulus, influence cell functions, such as nuclear deformation, cell proliferation, differentiation, and reprogramming, to summarize the differences and similarities among each biomaterial cues and their mechanisms, and to find common and unique properties of different biomaterial cues. Moreover, this work aims to establish a mechanogenomic map facilitating highly functionalized biomaterial design, and renders new thoughts of epigenetic regulation in controlling cell fates in disease treatment and regenerative medicine.
Collapse
Affiliation(s)
- Longwei Lv
- 1 Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing, People's Republic of China
| | - Yiman Tang
- 1 Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing, People's Republic of China
| | - Ping Zhang
- 1 Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing, People's Republic of China
| | - Yunsong Liu
- 1 Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing, People's Republic of China
| | - Xiangsong Bai
- 1 Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing, People's Republic of China
| | - Yongsheng Zhou
- 1 Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing, People's Republic of China
- 2 National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology , Beijing, People's Republic of China
| |
Collapse
|
19
|
Fernando WA, Papantoniou I, Mendes LF, Hall GN, Bosmans K, Tam WL, Teixeira LM, Moos M, Geris L, Luyten FP. Limb derived cells as a paradigm for engineering self-assembling skeletal tissues. J Tissue Eng Regen Med 2017; 12:794-807. [PMID: 28603948 DOI: 10.1002/term.2498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/21/2022]
Abstract
Mimicking developmental events has been proposed as a strategy to engineer tissue constructs for regenerative medicine. However, this approach has not yet been investigated for skeletal tissues. Here, it is demonstrated that ectopic implantation of day-14.5 mouse embryonic long bone anlagen, dissociated into single cells and randomly incorporated in a bioengineered construct, gives rise to epiphyseal growth plate-like structures, bone and marrow, which share many morphological and molecular similarities to epiphyseal units that form after transplanting intact long bone anlage, demonstrating substantial robustness and autonomy of complex tissue self-assembly and the overall organogenesis process. In vitro studies confirm the self-aggregation and patterning capacity of anlage cells and demonstrate that the model can be used to evaluate the effects of large and small molecules on biological behaviour. These results reveal the preservation of self-organizing and self-patterning capacity of anlage cells even when disconnected from their developmental niche and subjected to system perturbations such as cellular dissociation. These inherent features make long bone anlage cells attractive as a model system for tissue engineering technologies aimed at creating constructs that have the potential to self-assemble and self-pattern complex architectural structures.
Collapse
Affiliation(s)
- Warnakulasuriya A Fernando
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Ioannis Papantoniou
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Luis F Mendes
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Gabriella Nilsson Hall
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Kathleen Bosmans
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Wai L Tam
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Liliana M Teixeira
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| | - Malcolm Moos
- Division of Cellular, Tissue and Gene Therapies, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Liesbet Geris
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium.,Biomechanics Research Unit, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, Belgium.,Prometheus Division of Skeletal Tissue Engineering, Belgium
| |
Collapse
|
20
|
Vasilevich AS, Carlier A, de Boer J, Singh S. How Not To Drown in Data: A Guide for Biomaterial Engineers. Trends Biotechnol 2017; 35:743-755. [PMID: 28693857 DOI: 10.1016/j.tibtech.2017.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/27/2017] [Accepted: 05/30/2017] [Indexed: 01/20/2023]
Abstract
High-throughput assays that produce hundreds of measurements per sample are powerful tools for quantifying cell-material interactions. With advances in automation and miniaturization in material fabrication, hundreds of biomaterial samples can be rapidly produced, which can then be characterized using these assays. However, the resulting deluge of data can be overwhelming. To the rescue are computational methods that are well suited to these problems. Machine learning techniques provide a vast array of tools to make predictions about cell-material interactions and to find patterns in cellular responses. Computational simulations allow researchers to pose and test hypotheses and perform experiments in silico. This review describes approaches from these two domains that can be brought to bear on the problem of analyzing biomaterial screening data.
Collapse
Affiliation(s)
- Aliaksei S Vasilevich
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Aurélie Carlier
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jan de Boer
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Shantanu Singh
- Imaging Platform, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
21
|
Bolander J, Ji W, Leijten J, Teixeira LM, Bloemen V, Lambrechts D, Chaklader M, Luyten FP. Healing of a Large Long-Bone Defect through Serum-Free In Vitro Priming of Human Periosteum-Derived Cells. Stem Cell Reports 2017; 8:758-772. [PMID: 28196691 PMCID: PMC5355567 DOI: 10.1016/j.stemcr.2017.01.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/25/2022] Open
Abstract
Clinical translation of cell-based strategies for regenerative medicine demands predictable in vivo performance where the use of sera during in vitro preparation inherently limits the efficacy and reproducibility. Here, we present a bioinspired approach by serum-free pre-conditioning of human periosteum-derived cells, followed by their assembly into microaggregates simultaneously primed with bone morphogenetic protein 2 (BMP-2). Pre-conditioning resulted in a more potent progenitor cell population, while aggregation induced osteochondrogenic differentiation, further enhanced by BMP-2 stimulation. Ectopic implantation displayed a cascade of events that closely resembled the natural endochondral process resulting in bone ossicle formation. Assessment in a critical size long-bone defect in immunodeficient mice demonstrated successful bridging of the defect within 4 weeks, with active contribution of the implanted cells. In short, the presented serum-free process represents a biomimetic strategy, resulting in a cartilage tissue intermediate that, upon implantation, robustly leads to the healing of a large long-bone defect. Serum-free pre-conditioning affects the identity of periosteal progenitor cells A reduced CD105+, elevated CD34+, and upregulated BMP receptor expression was seen Priming by aggregation and BMP stimulation induced endochondral bone formation Validation in a critical size fracture model confirmed endochondral healing
Collapse
Affiliation(s)
- Johanna Bolander
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium
| | - Wei Ji
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium
| | - Jeroen Leijten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Drienerlolaan 5, 7522NB Enschede, the Netherlands
| | - Liliana Moreira Teixeira
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium
| | - Veerle Bloemen
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Materials Technology TC, Campus Group T, KU Leuven, Andreas Vesaliusstraat 13, 3000 Leuven, Belgium
| | - Dennis Lambrechts
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium
| | - Malay Chaklader
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium
| | - Frank P Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Box 813 13, 3000 Leuven, Belgium.
| |
Collapse
|
22
|
Ning B, Zhao Y, Buza JA, Li W, Wang W, Jia T. Surgically‑induced mouse models in the study of bone regeneration: Current models and future directions (Review). Mol Med Rep 2017; 15:1017-1023. [PMID: 28138711 PMCID: PMC5367352 DOI: 10.3892/mmr.2017.6155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 12/13/2016] [Indexed: 01/17/2023] Open
Abstract
Bone regeneration has been extensively studied over the past several decades. The surgically‑induced mouse model is the key animal model for studying bone regeneration, of the various research strategies used. These mouse models mimic the trauma and recovery processes in vivo and serve as carriers for tissue engineering and gene modification to test various therapies or associated genes in bone regeneration. The present review introduces a classification of surgically induced mouse models in bone regeneration, evaluates the application and value of these models and discusses the potential development of further innovations in this field in the future.
Collapse
Affiliation(s)
- Bin Ning
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Yunpeng Zhao
- Department of Orthopedic Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - John A Buza
- Department of Orthopedic Surgery, New York University Medical Center, New York, NY 10003, USA
| | - Wei Li
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Wenzhao Wang
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Tanghong Jia
- Department of Orthopedic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
23
|
Vas WJ, Shah M, Al Hosni R, Owen HC, Roberts SJ. Biomimetic strategies for fracture repair: Engineering the cell microenvironment for directed tissue formation. J Tissue Eng 2017; 8:2041731417704791. [PMID: 28491274 PMCID: PMC5406151 DOI: 10.1177/2041731417704791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
Complications resulting from impaired fracture healing have major clinical implications on fracture management strategies. Novel concepts taken from developmental biology have driven research strategies towards the elaboration of regenerative approaches that can truly harness the complex cellular events involved in tissue formation and repair. Advances in polymer technology and a better understanding of naturally derived scaffolds have given rise to novel biomaterials with an increasing ability to recapitulate native tissue environments. This coupled with advances in the understanding of stem cell biology and technology has opened new avenues for regenerative strategies with true clinical translatability. These advances have provided the impetus to develop alternative approaches to enhance the fracture repair process. We provide an update on these advances, with a focus on the development of novel biomimetic approaches for bone regeneration and their translational potential.
Collapse
Affiliation(s)
- Wollis J Vas
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| | - Mittal Shah
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| | - Rawiya Al Hosni
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| | - Helen C Owen
- Department of Natural Sciences, School of Science & Technology, Middlesex University, London, UK
| | - Scott J Roberts
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| |
Collapse
|
24
|
Tang Z, Tan Y, Ni Y, Wang J, Zhu X, Fan Y, Chen X, Yang X, Zhang X. Comparison of ectopic bone formation process induced by four calcium phosphate ceramics in mice. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:1000-1010. [DOI: 10.1016/j.msec.2016.06.097] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/15/2016] [Accepted: 06/29/2016] [Indexed: 12/11/2022]
|
25
|
Langhans MT, Yu S, Tuan RS. Stem Cells in Skeletal Tissue Engineering: Technologies and Models. Curr Stem Cell Res Ther 2016; 11:453-474. [PMID: 26423296 DOI: 10.2174/1574888x10666151001115248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
This review surveys the use of pluripotent and multipotent stem cells in skeletal tissue engineering. Specific emphasis is focused on evaluating the function and activities of these cells in the context of development in vivo, and how technologies and methods of stem cell-based tissue engineering for stem cells must draw inspiration from developmental biology. Information on the embryonic origin and in vivo differentiation of skeletal tissues is first reviewed, to shed light on the persistence and activities of adult stem cells that remain in skeletal tissues after embryogenesis. Next, the development and differentiation of pluripotent stem cells is discussed, and some of their advantages and disadvantages in the context of tissue engineering are presented. The final section highlights current use of multipotent adult mesenchymal stem cells, reviewing their origin, differentiation capacity, and potential applications to tissue engineering.
Collapse
Affiliation(s)
| | | | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA 15219, USA.
| |
Collapse
|
26
|
Groen N, Guvendiren M, Rabitz H, Welsh WJ, Kohn J, de Boer J. Stepping into the omics era: Opportunities and challenges for biomaterials science and engineering. Acta Biomater 2016; 34:133-142. [PMID: 26876875 DOI: 10.1016/j.actbio.2016.02.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/22/2016] [Accepted: 02/10/2016] [Indexed: 12/11/2022]
Abstract
The research paradigm in biomaterials science and engineering is evolving from using low-throughput and iterative experimental designs towards high-throughput experimental designs for materials optimization and the evaluation of materials properties. Computational science plays an important role in this transition. With the emergence of the omics approach in the biomaterials field, referred to as materiomics, high-throughput approaches hold the promise of tackling the complexity of materials and understanding correlations between material properties and their effects on complex biological systems. The intrinsic complexity of biological systems is an important factor that is often oversimplified when characterizing biological responses to materials and establishing property-activity relationships. Indeed, in vitro tests designed to predict in vivo performance of a given biomaterial are largely lacking as we are not able to capture the biological complexity of whole tissues in an in vitro model. In this opinion paper, we explain how we reached our opinion that converging genomics and materiomics into a new field would enable a significant acceleration of the development of new and improved medical devices. The use of computational modeling to correlate high-throughput gene expression profiling with high throughput combinatorial material design strategies would add power to the analysis of biological effects induced by material properties. We believe that this extra layer of complexity on top of high-throughput material experimentation is necessary to tackle the biological complexity and further advance the biomaterials field. STATEMENT OF SIGNIFICANCE In this opinion paper, we postulate that converging genomics and materiomics into a new field would enable a significant acceleration of the development of new and improved medical devices. The use of computational modeling to correlate high-throughput gene expression profiling with high throughput combinatorial material design strategies would add power to the analysis of biological effects induced by material properties. We believe that this extra layer of complexity on top of high-throughput material experimentation is necessary to tackle the biological complexity and further advance the biomaterials field.
Collapse
Affiliation(s)
- Nathalie Groen
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Murat Guvendiren
- New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ, USA
| | - Herschel Rabitz
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - William J Welsh
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ, USA
- Department of Chemistry and Chemical Biology, New Jersey Center for Biomaterials, Rutgers University, Piscataway, NJ, USA
| | - Jan de Boer
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- cBITE Lab, Merln Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
27
|
Bolander J, Chai YC, Geris L, Schrooten J, Lambrechts D, Roberts SJ, Luyten FP. Early BMP, Wnt and Ca(2+)/PKC pathway activation predicts the bone forming capacity of periosteal cells in combination with calcium phosphates. Biomaterials 2016; 86:106-18. [PMID: 26901484 DOI: 10.1016/j.biomaterials.2016.01.059] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 02/08/2023]
Abstract
The development of osteoinductive calcium phosphate- (CaP) based biomaterials has, and continues to be, a major focus in the field of bone tissue engineering. However, limited insight into the spatiotemporal activation of signalling pathways has hampered the optimisation of in vivo bone formation and subsequent clinical translation. To gain further knowledge regarding the early molecular events governing bone tissue formation, we combined human periosteum derived progenitor cells with three types of clinically used CaP-scaffolds, to obtain constructs with a distinct range of bone forming capacity in vivo. Protein phosphorylation together with gene expression for key ligands and target genes were investigated 24 hours after cell seeding in vitro, and 3 and 12 days post ectopic implantation in nude mice. A computational modelling approach was used to deduce critical factors for bone formation 8 weeks post implantation. The combined Ca(2+)-mediated activation of BMP-, Wnt- and PKC signalling pathways 3 days post implantation were able to discriminate the bone forming from the non-bone forming constructs. Subsequently, a mathematical model able to predict in vivo bone formation with 96% accuracy was developed. This study illustrates the importance of defining and understanding CaP-activated signalling pathways that are required and sufficient for in vivo bone formation. Furthermore, we demonstrate the reliability of mathematical modelling as a tool to analyse and deduce key factors within an empirical data set and highlight its relevance to the translation of regenerative medicine strategies.
Collapse
Affiliation(s)
- Johanna Bolander
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium
| | - Yoke Chin Chai
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Biomechanics Research Unit, University of Liege, Chemin des Chevreuils 1, BAT 52/3, 4000 Liege 1, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C, Bus 2419, 3001 Leuven, Belgium
| | - Jan Schrooten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44, Bus 2450, 3001 Heverlee, Belgium
| | - Dennis Lambrechts
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium
| | - Scott J Roberts
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, The Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, United Kingdom
| | - Frank P Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium.
| |
Collapse
|
28
|
Chen X, Wang J, Chen Y, Cai H, Yang X, Zhu X, Fan Y, Zhang X. Roles of calcium phosphate-mediated integrin expression and MAPK signaling pathways in the osteoblastic differentiation of mesenchymal stem cells. J Mater Chem B 2016; 4:2280-2289. [DOI: 10.1039/c6tb00349d] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BCP ceramics mediated MSC's integrin expression to realize “outside-in signaling” transduction and then activated MAPK signaling to induce osteogenesis.
Collapse
Affiliation(s)
- Xuening Chen
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Jing Wang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Ying Chen
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Hanxu Cai
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| |
Collapse
|
29
|
Xu SJ, Qiu ZY, Wu JJ, Kong XD, Weng XS, Cui FZ, Wang XM. Osteogenic Differentiation Gene Expression Profiling of hMSCs on Hydroxyapatite and Mineralized Collagen. Tissue Eng Part A 2015; 22:170-81. [PMID: 26529501 DOI: 10.1089/ten.tea.2015.0237] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this study, human mesenchymal stem cells (hMSCs) were cultured on the hydroxyapatite (HA) and mineralized collagen (MC), and their proliferation, adhesion, and differentiation, especially the molecular mechanisms on gene level, were investigated. Proliferation and morphological responses of hMSCs and their osteogenic differentiation were detected by quantitative detection of alkaline phosphatase. Gene expression profilings were examined by microarrays, and the gene expression data were studied through gene ontology terms and pathway analyses. The results showed that MC promoted cell proliferation and osteogenic differentiation of hMSCs. Microarray analysis showed that MC was conducive to express osteogenesis-related genes, such as BMP-2, COL1A1, and CTSK, and stimulate osteogenic differentiation, such as osteoblast differentiation pathway and skeletal system development pathway.
Collapse
Affiliation(s)
- Su-Ju Xu
- 1 State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University , Beijing, China .,2 College of Life Sciences, Zhejiang Sci-Tech University , Hangzhou, China
| | - Zhi-Ye Qiu
- 1 State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University , Beijing, China
| | - Jing-Jing Wu
- 1 State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University , Beijing, China .,3 School of Engineering and Technology, China University of Geosciences , Beijing, China
| | - Xiang-Dong Kong
- 2 College of Life Sciences, Zhejiang Sci-Tech University , Hangzhou, China
| | - Xi-Sheng Weng
- 4 Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing, China
| | - Fu-Zhai Cui
- 1 State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University , Beijing, China
| | - Xiu-Mei Wang
- 1 State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University , Beijing, China
| |
Collapse
|
30
|
Rao V, Shih YRV, Kang H, Kabra H, Varghese S. Adenosine Signaling Mediates Osteogenic Differentiation of Human Embryonic Stem Cells on Mineralized Matrices. Front Bioeng Biotechnol 2015; 3:185. [PMID: 26618155 PMCID: PMC4639610 DOI: 10.3389/fbioe.2015.00185] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/28/2015] [Indexed: 12/20/2022] Open
Abstract
Human embryonic stem cells (hESCs) are attractive cell sources for tissue engineering and regenerative medicine due to their self-renewal and differentiation ability. Design of biomaterials with an intrinsic ability that promotes hESC differentiation to the targeted cell type boasts significant advantages for tissue regeneration. We have previously developed biomineralized calcium phosphate (CaP) matrices that inherently direct osteogenic differentiation of hESCs without the need of osteogenic-inducing chemicals or growth factors. Here, we show that CaP matrix-driven osteogenic differentiation of hESCs occurs through A2b adenosine receptor (A2bR). The inhibition of the receptor with an A2bR-specific antagonist attenuated mineralized matrix-mediated osteogenic differentiation of hESCs. In addition, when cultured on matrices in an environment deficient of CaP minerals, exogenous adenosine promoted osteogenic differentiation of hESCs, but was attenuated by the inhibition of A2bR. Such synthetic matrices that intrinsically support osteogenic commitment of hESCs are not only beneficial for bone tissue engineering but can also be used as a platform to study the effect of the physical and chemical cues to the extracellular milieu on stem cell commitment. Insights into the cell signaling during matrix-induced differentiation of stem cells will also help define the key processes and enable discovery of new targets that promote differentiation of pluripotent stem cells for bone tissue engineering.
Collapse
Affiliation(s)
- Vikram Rao
- Department of Bioengineering, University of California San Diego , La Jolla, CA , USA
| | - Yu-Ru V Shih
- Department of Bioengineering, University of California San Diego , La Jolla, CA , USA
| | - Heemin Kang
- Materials Science and Engineering Program, University of California San Diego , La Jolla, CA , USA
| | - Harsha Kabra
- Department of Bioengineering, University of California San Diego , La Jolla, CA , USA
| | - Shyni Varghese
- Department of Bioengineering, University of California San Diego , La Jolla, CA , USA
| |
Collapse
|
31
|
Groen N, Tahmasebi N, Shimizu F, Sano Y, Kanda T, Barbieri D, Yuan H, Habibovic P, van Blitterswijk CA, de Boer J. Exploring the Material-Induced Transcriptional Landscape of Osteoblasts on Bone Graft Materials. Adv Healthc Mater 2015; 4:1691-700. [PMID: 26046651 DOI: 10.1002/adhm.201500171] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/30/2015] [Indexed: 01/14/2023]
Abstract
During the past decades, there have been major advances in the field of biomaterials, thereby generating a vast variety of materials for a broad range of tissue engineering and regeneration applications. Although gene expression profiling has been used occasionally in biomaterial research, its usefulness for understanding cell-biomaterial interactions should be further explored for it to fulfill its promise as a tool to assess and improve material properties. Here, the transcriptional landscape induced by 23 materials is explored with a variety of properties within the scope of bone regeneration. An osteoblast cell line is used to identify the gene expression profiles that can be adopted in response to biophysical and chemical cues. It is shown that TGF-β and WNT signaling may be involved in the cellular response to osteoinductive materials along with differential cell adhesion kinetics via attenuated FAK signaling. The previously reported effect of calcium and phosphate on BMP2 and TGF-β signaling is confirmed and the biological effect of the addition of nanohydroxyapatite in poly (d,l-lactic acid) polymer particles is studied. Together with future applications, this approach will help researchers understand cellular responses in relation to material properties, which will promote the development of more effective biomaterials for applications in tissue regeneration.
Collapse
Affiliation(s)
- Nathalie Groen
- Department of Tissue Regeneration; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede 7500 AE The Netherlands
| | - Niloofar Tahmasebi
- Department of Tissue Regeneration; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede 7500 AE The Netherlands
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi 755-8505 Japan
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi 755-8505 Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi 755-8505 Japan
| | - Davide Barbieri
- Department of Tissue Regeneration; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede 7500 AE The Netherlands
- Xpand Biotechnology BV; Prof. Bronkhorstlaan 10-D 3723 MB Bilthoven The Netherlands
| | - Huipin Yuan
- Department of Tissue Regeneration; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede 7500 AE The Netherlands
- Xpand Biotechnology BV; Prof. Bronkhorstlaan 10-D 3723 MB Bilthoven The Netherlands
| | - Pamela Habibovic
- Department of Tissue Regeneration; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede 7500 AE The Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine; Maastricht University; 6200 MD Maastricht The Netherlands
| | - Clemens A van Blitterswijk
- Department of Tissue Regeneration; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede 7500 AE The Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine; Maastricht University; 6200 MD Maastricht The Netherlands
| | - Jan de Boer
- Department of Tissue Regeneration; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Enschede 7500 AE The Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine; Maastricht University; 6200 MD Maastricht The Netherlands
| |
Collapse
|
32
|
Brennan O, Stenson B, Widaa A, O Gorman DM, O Brien FJ. Incorporation of the natural marine multi-mineral dietary supplement Aquamin enhances osteogenesis and improves the mechanical properties of a collagen-based bone graft substitute. J Mech Behav Biomed Mater 2015; 47:114-123. [PMID: 25884141 DOI: 10.1016/j.jmbbm.2015.03.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/22/2015] [Indexed: 12/12/2022]
Abstract
Aquamin is a commercially-available supplement derived from the algae species Lithothamnion, which has proven osteogenic potential. By harnessing this potential and combining Aquamin with a collagen scaffold, with architecture and composition optimised for bone repair, the aim of this study was to develop a natural osteo-stimulative bone graft substitute. A fabrication process was developed to incorporate Aquamin into scaffolds to produce collagen-Aquamin (CollAqua) scaffolds at two different Aquamin concentrations, 100F or 500F (equivalent weight% of collagen or five times the weight of collagen respectively). CollAqua constructs had improved mechanical properties which were achieved without reducing the scaffold׳s permeability or porosity below the minimum level required for successful bone tissue engineering. The fabrication process produced a homogenous Aquamin distribution throughout the scaffold. Release kinetics revealed that in the first 12h, the entire Aquamin content was released from the 100F however, less than half of Aquamin in the 500F was released with the remainder released approximately 21 days later giving an initial burst release followed by a delayed release. Osteoblasts cultured on the CollAqua scaffolds showed improved osteogenesis as measured by alkaline phosphatase, osteopontin and osteocalcin expression. This was confirmed by increased mineralisation as determined by von Kossa and Alizarin red staining. In conclusion, a cell and growth factor free collagen-based bone graft substitute with enhanced mechanical properties has been developed. The addition of Aquamin to the collagen biomaterial significantly improved mineralisation by osteoblasts and results in a new product which may be capable of enhancing osteogenesis to facilitate bone repair in vivo.
Collapse
Affiliation(s)
- Orlaith Brennan
- Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| | - Barry Stenson
- Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland
| | - Amro Widaa
- Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland, Dublin 2, Ireland; Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Denise M O Gorman
- Marigot Ltd., Strand Farm, Currabinny, Carrigaline, Co. Cork, Ireland
| | - Fergal J O Brien
- Tissue Engineering Research Group (TERG), Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
33
|
Gandolfi MG, Spagnuolo G, Siboni F, Procino A, Rivieccio V, Pelliccioni GA, Prati C, Rengo S. Calcium silicate/calcium phosphate biphasic cements for vital pulp therapy: chemical-physical properties and human pulp cells response. Clin Oral Investig 2015; 19:2075-89. [DOI: 10.1007/s00784-015-1443-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/25/2015] [Indexed: 01/19/2023]
|
34
|
Roberts SJ, van Gastel N, Carmeliet G, Luyten FP. Uncovering the periosteum for skeletal regeneration: the stem cell that lies beneath. Bone 2015; 70:10-8. [PMID: 25193160 DOI: 10.1016/j.bone.2014.08.007] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 08/14/2014] [Accepted: 08/16/2014] [Indexed: 12/20/2022]
Abstract
The cartilage- and bone-forming properties of the periosteum have long since been recognized. As one of the major sources of skeletal progenitor cells, the periosteum plays a crucial role not only in bone development and growth, but also during bone fracture healing. Aided by the continuous expansion of tools and techniques, we are now starting to acquire more insight into the specific role and regulation of periosteal cells. From a therapeutic point of view, the periosteum has attracted much attention as a cell source for bone tissue engineering purposes. This interest derives not only from the physiological role of the periosteum during bone repair, but is also supported by the unique properties and marked bone-forming potential of expanded periosteum-derived cells. We provide an overview of the current knowledge of periosteal cell biology, focusing on the cellular composition and molecular regulation of this remarkable tissue, as well as the application of periosteum-derived cells in regenerative medicine approaches. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Scott J Roberts
- Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium; Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, The Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, UK
| | - Nick van Gastel
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium; Clinical and Experimental Endocrinology, KU Leuven, O&N 1 Herestraat 49 bus 902, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium; Clinical and Experimental Endocrinology, KU Leuven, O&N 1 Herestraat 49 bus 902, 3000 Leuven, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium.
| |
Collapse
|
35
|
Bassi G, Guilloton F, Menard C, Di Trapani M, Deschaseaux F, Sensebé L, Schrezenmeier H, Giordano R, Bourin P, Dominici M, Tarte K, Krampera M. Effects of a ceramic biomaterial on immune modulatory properties and differentiation potential of human mesenchymal stromal cells of different origin. Tissue Eng Part A 2014; 21:767-81. [PMID: 25322665 DOI: 10.1089/ten.tea.2014.0269] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to assess the immune modulatory properties of human mesenchymal stromal cells obtained from bone marrow (BM-MSCs), fat (ASCs), and cord blood (CB-MSCs) in the presence of a hydroxyapatite and tricalcium-phosphate (HA/TCP) biomaterial as a scaffold for MSC delivery. In resting conditions, a short-term culture with HA/TCP did not modulate the anti-apoptotic and suppressive features of the various MSC types toward T, B, and NK cells; in addition, when primed with inflammatory cytokines, MSCs similarly increased their suppressive capacities in the presence or absence of HA/TCP. The long-term culture of BM-MSCs with HA/TCP induced an osteoblast-like phenotype with upregulation of OSTERIX and OSTEOCALCIN, similar to what was obtained with dexamethasone and, to a higher extent, with bone morphogenetic protein 4 (BMP-4) treatment. MSC-derived osteoblasts did not trigger immune cell activation, but were less efficient than undifferentiated MSCs in inhibiting stimulated T and NK cells. Interestingly, their suppressive machinery included not only the activation of indoleamine-2,3 dioxygenase (IDO), which plays a central role in T-cell inhibition, but also cyclooxygenase-2 (COX-2) that was not significantly involved in the immune modulatory effect of human undifferentiated MSCs. Since COX-2 is significantly involved in bone healing, its induction by HA/TCP could also contribute to the therapeutic activity of MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Giulio Bassi
- 1 Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona , Verona, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ectopic osteoid and bone formation by three calcium-phosphate ceramics in rats, rabbits and dogs. PLoS One 2014; 9:e107044. [PMID: 25229501 PMCID: PMC4167699 DOI: 10.1371/journal.pone.0107044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 08/14/2014] [Indexed: 02/05/2023] Open
Abstract
Calcium phosphate ceramics with specific physicochemical properties have been shown to induce de novo bone formation upon ectopic implantation in a number of animal models. In this study we explored the influence of physicochemical properties as well as the animal species on material-induced ectopic bone formation. Three bioceramics were used for the study: phase-pure hydroxyapatite (HA) sintered at 1200°C and two biphasic calcium phosphate (BCP) ceramics, consisting of 60 wt.% HA and 40 wt.% TCP (β-Tricalcium phosphate), sintered at either 1100°C or 1200°C. 108 samples of each ceramic were intramuscularly implanted in dogs, rabbits, and rats for 6, 12, and 24 weeks respectively. Histological and histomorphometrical analyses illustrated that ectopic bone and/or osteoid tissue formation was most pronounced in BCP sintered at 1100°C and most limited in HA, independent of the animal model. Concerning the effect of animal species, ectopic bone formation reproducibly occurred in dogs, while in rabbits and rats, new tissue formation was mainly limited to osteoid. The results of this study confirmed that the incidence and the extent of material-induced bone formation are related to both the physicochemical properties of calcium phosphate ceramics and the animal model.
Collapse
|
37
|
McArdle A, Chung MT, Paik KJ, Duldulao C, Chan C, Rennert R, Walmsley GG, Senarath-Yapa K, Hu M, Seo E, Lee M, Wan DC, Longaker MT. Positive selection for bone morphogenetic protein receptor type-IB promotes differentiation and specification of human adipose-derived stromal cells toward an osteogenic lineage. Tissue Eng Part A 2014; 20:3031-40. [PMID: 24854876 DOI: 10.1089/ten.tea.2014.0101] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Adipose tissue represents an abundant and easily accessible source of multipotent cells that may serve as an excellent building block for tissue engineering. However, adipose-derived stromal cells (ASCs) are a heterogeneous group and subpopulations may be identified with enhanced osteogenic potential. METHODS Human ASC subpopulations were prospectively isolated based on expression of bone morphogenetic protein receptor type-IB (BMPR-IB). Unsorted, BMPR-IB(+), and BMPR-IB(-) cells were analyzed for their osteogenic capacity through histological staining and gene expression. To evaluate their in vivo osteogenic potential, critical-sized calvarial defects were created in immunocompromised mice and treated with unsorted, BMPR-IB(+), or BMPR-IB(-) cells. Healing was assessed using microcomputed tomography and pentachrome staining of specimens at 8 weeks. RESULTS Increased osteogenic differentiation was noted in the BMPR-IB(+) subpopulation, as demonstrated by alkaline phosphatase staining at day 7 and extracellular matrix mineralization with Alizarin red staining at day 14. This was also associated with increased expression for osteocalcin, a late marker of osteogenesis. Radiographic analysis demonstrated significantly enhanced healing of critical-sized calvarial defects treated with BMPR-IB(+) ASCs compared with unsorted or BMPR-IB(-) cells. This was confirmed through pentachrome staining, which revealed more robust bone regeneration in the BMPR-IB(+) group. CONCLUSION BMPR-IB(+) human ASCs have an enhanced ability to form bone both in vitro and in vivo. These data suggest that positive selection for BMPR-IB(+) and manipulation of the BMP pathway in these cells may yield a highly osteogenic subpopulation of cells for bone tissue engineering.
Collapse
Affiliation(s)
- Adrian McArdle
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW This review outlines the concept of cell-based therapy to restore tissue function, and addresses four key points to consider in cell transplantation: source, surveillance, safety, and site. Whereas each point is essential, additional attention should be given to transplantation sites if cell therapy is going to be successful in the clinic. Various ectopic locations are discussed, and the strengths and weaknesses of each are compared as suitable candidates for cell therapy. RECENT FINDINGS Studies in rodents often demonstrate cell transplantation and engraftment in ectopic sites, with little evidence to suggest why it may also work in humans. For example, transplantation to the subcapsular space of the kidney is often performed in rodents, but has not been a good predictor of clinical success. Recent work has shown that the lymph node may be a good site for transplantation of multiple tissue types, and several reasons are highlighted as to why it should be considered for future studies. SUMMARY The use of cell-based therapy in the clinic has been hampered by the lack of appropriate sites for transplantation. The lymph node is a promising alternative for cell transplantation, and offers hope for clinical application.
Collapse
|
39
|
Roberts SJ, Owen HC, Tam WL, Solie L, Van Cromphaut SJ, Van den Berghe G, Luyten FP. Humanized culture of periosteal progenitors in allogeneic serum enhances osteogenic differentiation and in vivo bone formation. Stem Cells Transl Med 2013; 3:218-28. [PMID: 24375540 DOI: 10.5966/sctm.2012-0137] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The translation of stem cell-based regenerative solutions from the laboratory to the clinic is often hindered by the culture conditions used to expand cell populations. Although fetal bovine serum (FBS) is widely used, regulatory bodies and safety concerns encourage alternative, xeno-free culturing practices. In an attempt to apply this approach to a bone-forming combination product of human periosteal progenitors (human periosteum derived cells) on a clinically used calcium phosphate carrier, FBS was substituted for human allogeneic serum (hAS) during cell expansion. It was found that cell proliferation was increased in hAS along with an apparent commitment to the osteogenic lineage, indicated by enhanced Runx2 expression, as well as alkaline phosphatase activity and matrix mineralization. Following analysis of signaling pathways, it was found that interferon-mediated signaling was downregulated, whereas JAK-STAT signaling was upregulated. STAT3 phosphorylation was enhanced in hAS-cultured human periosteum derived cells, inhibition of which ablated the proliferative effect of hAS. Furthermore, following in vivo implantation of hAS-cultured cells on NuOss scaffolds, enhanced bone formation was observed compared with FBS (71% increase, p < .001). Interestingly, the de novo-formed bone appeared to have a higher ratio of immature regions to mature regions, indicating that after 8 weeks implantation, tissue-formation processes were continuing. Integration of the implant with the environment appeared to be altered, with a decrease in calcium phosphate grain size and surface area, indicative of accelerated resorption. This study highlights the advantages of using humanized culture conditions for the expansion of human periosteal progenitors intended for bone regeneration.
Collapse
Affiliation(s)
- Scott J Roberts
- Laboratory for Developmental and Stem Cell Biology, Skeletal Biology and Engineering Research Center, Prometheus, Division of Skeletal Tissue Engineering, and Department and Laboratory of Intensive Care Medicine, KU Leuven, Leuven, Belgium; Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, The Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
40
|
In Vitro Screening of the Apatite-Forming Ability, Biointeractivity and Physical Properties of a Tricalcium Silicate Material for Endodontics and Restorative Dentistry. Dent J (Basel) 2013. [DOI: 10.3390/dj1040041] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
41
|
Overman JR, Helder MN, ten Bruggenkate CM, Schulten EAJM, Klein-Nulend J, Bakker AD. Growth factor gene expression profiles of bone morphogenetic protein-2-treated human adipose stem cells seeded on calcium phosphate scaffolds in vitro. Biochimie 2013; 95:2304-13. [PMID: 24028822 DOI: 10.1016/j.biochi.2013.08.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/27/2013] [Indexed: 12/17/2022]
Abstract
The secretome of stem cells strongly determines the outcome of tissue engineering strategies. We investigated how the secretome of human adipose stem cells (hASCs) can be affected by substrate, BMP-2 treatment, and degree of differentiation. We hypothesized that as differentiation progresses, hASCs produce increasingly more gene products associated with processes such as angiogenesis and bone remodeling. Human ASCs were treated for 15 min with BMP-2 (10 ng/ml) to enhance osteogenic differentiation, or with vehicle. Subsequently, hASCs were seeded on plastic or on biphasic calcium phosphate (BCP) consisting of 60% hydroxyapatite and 40% β-tricalcium phosphate. A PCR array for ~150 trophic factors and differentiation-related genes was performed at day 21 of culture. A limited set of factors was quantified by qPCR at days 0, 4, 14 and 21, and/or ELISA at day 21. Compared to plastic, BCP-cultured hASCs showed ≥2-fold higher expression of ~20 factors, e.g. cytokines such as IL-6, growth factors such as FGF7 and adhesion molecules such as VCAM1. Expression of another ~50 genes was decreased ≥2-fold on BCP vs. plastic, even though hASCs differentiate better on BCP than on plastic. BMP-2-treatment increased the expression of ~30 factors by hASCs seeded on BCP, while it decreased the expression of only PGF, PPARG and PTN. Substrate affected hASC secretion of Activin A and seemed to affect P1NP release. No clear association between hASC osteogenic differentiation and growth factor expression pattern was observed. Considering our observed lack of association between the degree of differentiation and the expression of factors associated with angiogenesis and bone remodeling by hASCs, future bone regeneration studies should focus more on systematically orchestrating the secretome of stem cells, rather than on inducing osteogenic differentiation of stem cells only. Short incubation with BMP-2 may be a promising treatment to enhance both osteogenic differentiation and environmental modulation.
Collapse
Affiliation(s)
- J R Overman
- Dept. Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands; Dept. Oral and Maxillofacial Surgery, Academic Centre for Dentistry Amsterdam/VU University Medical Center, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|