1
|
Alaoui Selsouli Y, Rho HS, Eischen-Loges M, Galván-Chacón VP, Stähli C, Viecelli Y, Döbelin N, Bohner M, Tahmasebi Birgani Z, Habibović P. Optimization of a tunable process for rapid production of calcium phosphate microparticles using a droplet-based microfluidic platform. Front Bioeng Biotechnol 2024; 12:1352184. [PMID: 38600949 PMCID: PMC11004461 DOI: 10.3389/fbioe.2024.1352184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Calcium phosphate (CaP) biomaterials are amongst the most widely used synthetic bone graft substitutes, owing to their chemical similarities to the mineral part of bone matrix and off-the-shelf availability. However, their ability to regenerate bone in critical-sized bone defects has remained inferior to the gold standard autologous bone. Hence, there is a need for methods that can be employed to efficiently produce CaPs with different properties, enabling the screening and consequent fine-tuning of the properties of CaPs towards effective bone regeneration. To this end, we propose the use of droplet microfluidics for rapid production of a variety of CaP microparticles. Particularly, this study aims to optimize the steps of a droplet microfluidic-based production process, including droplet generation, in-droplet CaP synthesis, purification and sintering, in order to obtain a library of CaP microparticles with fine-tuned properties. The results showed that size-controlled, monodisperse water-in-oil microdroplets containing calcium- and phosphate-rich solutions can be produced using a flow-focusing droplet-generator microfluidic chip. We optimized synthesis protocols based on in-droplet mineralization to obtain a range of CaP microparticles without and with inorganic additives. This was achieved by adjusting synthesis parameters, such as precursor concentration, pH value, and aging time, and applying heat treatment. In addition, our results indicated that the synthesis and fabrication parameters of CaPs in this method can alter the microstructure and the degradation behavior of CaPs. Overall, the results highlight the potential of the droplet microfluidic platform for engineering CaP microparticle biomaterials with fine-tuned properties.
Collapse
Affiliation(s)
- Y. Alaoui Selsouli
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - H. S. Rho
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - M. Eischen-Loges
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - V. P. Galván-Chacón
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - C. Stähli
- RMS Foundation, Bettlach, Switzerland
| | | | | | - M. Bohner
- RMS Foundation, Bettlach, Switzerland
| | - Z. Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - P. Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
2
|
Miron RJ, Bohner M, Zhang Y, Bosshardt DD. Osteoinduction and osteoimmunology: Emerging concepts. Periodontol 2000 2024; 94:9-26. [PMID: 37658591 DOI: 10.1111/prd.12519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/23/2023] [Accepted: 07/20/2023] [Indexed: 09/03/2023]
Abstract
The recognition and importance of immune cells during bone regeneration, including around bone biomaterials, has led to the development of an entire field termed "osteoimmunology," which focuses on the connection and interplay between the skeletal system and immune cells. Most studies have focused on the "osteogenic" capacity of various types of bone biomaterials, and much less focus has been placed on immune cells despite being the first cell type in contact with implantable devices. Thus, the amount of literature generated to date on this topic makes it challenging to extract needed information. This review article serves as a guide highlighting advancements made in the field of osteoimmunology emphasizing the role of the osteoimmunomodulatory properties of biomaterials and their impact on osteoinduction. First, the various immune cell types involved in bone biomaterial integration are discussed, including the prominent role of osteal macrophages (OsteoMacs) during bone regeneration. Thereafter, key biomaterial properties, including topography, wettability, surface charge, and adsorption of cytokines, growth factors, ions, and other bioactive molecules, are discussed in terms of their impact on immune responses. These findings highlight and recognize the importance of the immune system and osteoimmunology, leading to a shift in the traditional models used to understand and evaluate biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | | | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| | | |
Collapse
|
3
|
Yu Y, Wu T, Dong L. Surface Oxygen Vacancies of Rutile Nanorods Accelerate Biomineralization. ACS OMEGA 2023; 8:20066-20072. [PMID: 37305277 PMCID: PMC10249081 DOI: 10.1021/acsomega.3c02348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023]
Abstract
Titanium dioxide (TiO2) materials have been widely used in biomedical applications of bone tissue engineering. However, the mechanism underlying the induced biomineralization onto the TiO2 surface still remains elusive. In this study, we demonstrated that the surface oxygen vacancy defects of rutile nanorods could be gradually eliminated by the regularly used annealing treatment, which restrained the heterogeneous nucleation of hydroxyapatite (HA) onto rutile nanorods in simulated body fluids (SBFs). Moreover, we also observed that the surface oxygen vacancies upregulated the mineralization of human mesenchymal stromal cells (hMSCs) on rutile TiO2 nanorod substrates. This work therefore emphasized the importance of subtle changes of surface oxygen vacancy defective features of oxidic biomaterials during the regularly used annealing treatment on their bioactive performances and provided new insights into the fundamental understanding of interactions of materials with the biological environment.
Collapse
Affiliation(s)
- Yanwen Yu
- First
People’s Hospital of Linping District, Hangzhou 311100, Zhejiang, China
| | - Tong Wu
- Guangdian
Metrology & Testing (Hangzhou) Co., Ltd., Hangzhou 310018, Zhejiang, China
| | - Lingqing Dong
- Stomatology
Hospital, School of Stomatology, Zhejiang University School of Medicine,
Zhejiang Province Clinical Research Center for Oral Diseases, Key
Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
4
|
Jin P, Liu L, Cheng L, Chen X, Xi S, Jiang T. Calcium-to-phosphorus releasing ratio affects osteoinductivity and osteoconductivity of calcium phosphate bioceramics in bone tissue engineering. Biomed Eng Online 2023; 22:12. [PMID: 36759894 PMCID: PMC9912630 DOI: 10.1186/s12938-023-01067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
Calcium phosphate (Ca-P) bioceramics, including hydroxyapatite (HA), biphasic calcium phosphate (BCP), and beta-tricalcium phosphate (β-TCP), have been widely used in bone reconstruction. Many studies have focused on the osteoconductivity or osteoinductivity of Ca-P bioceramics, but the association between osteoconductivity and osteoinductivity is not well understood. In our study, the osteoconductivity of HA, BCP, and β-TCP was investigated based on the osteoblastic differentiation in vitro and in situ as well as calvarial defect repair in vivo, and osteoinductivity was evaluated by using pluripotent mesenchymal stem cells (MSCs) in vitro and heterotopic ossification in muscles in vivo. Our results showed that the cell viability, alkaline phosphatase activity, and expression of osteogenesis-related genes, including osteocalcin (Ocn), bone sialoprotein (Bsp), alpha-1 type I collagen (Col1a1), and runt-related transcription factor 2 (Runx2), of osteoblasts each ranked as BCP > β-TCP > HA, but the alkaline phosphatase activity and expression of osteogenic differentiation genes of MSCs each ranked as β-TCP > BCP > HA. Calvarial defect implantation of Ca-P bioceramics ranked as BCP > β-TCP ≥ HA, but intramuscular implantation ranked as β-TCP ≥ BCP > HA in vivo. Further investigation indicated that osteoconductivity and osteoinductivity are affected by the Ca/P ratio surrounding the Ca-P bioceramics. Thus, manipulating the appropriate calcium-to-phosphorus releasing ratio is a critical factor for determining the osteoinductivity of Ca-P bioceramics in bone tissue engineering.
Collapse
Affiliation(s)
- Pan Jin
- grid.410654.20000 0000 8880 6009Health Science Center, Yangtze University, Jingzhou, 434023 Hubei China ,Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and MinistryGuangxi Medical University, Nanning, 530021 Guangxi China
| | - Lei Liu
- grid.452877.b0000 0004 6005 8466Articular Surgery, The Second Nanning People’s Hospital, Third Affiliated Hospital of Guangxi Medical University), Nanning, 530031 Guangxi China
| | - Lin Cheng
- grid.410654.20000 0000 8880 6009Health Science Center, Yangtze University, Jingzhou, 434023 Hubei China
| | - Xichi Chen
- grid.410654.20000 0000 8880 6009Health Science Center, Yangtze University, Jingzhou, 434023 Hubei China
| | - Shanshan Xi
- Health Science Center, Yangtze University, Jingzhou, 434023, Hubei, China.
| | - Tongmeng Jiang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199, China. .,Key Laboratory of Emergency and Trauma, Ministry of Education, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
5
|
Phothichailert S, Nowwarote N, Fournier BP, Trachoo V, Roytrakul S, Namangkalakul W, Osathanon T. Effects of decellularized extracellular matrix derived from Jagged1-treated human dental pulp stem cells on biological responses of stem cells isolated from apical papilla. Front Cell Dev Biol 2022; 10:948812. [PMID: 36081912 PMCID: PMC9445441 DOI: 10.3389/fcell.2022.948812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022] Open
Abstract
Objective: Indirect Jagged1 immobilization efficiently activates canonical Notch signaling in human dental pulp stem cells (hDPSCs). This study aimed to investigate the characteristics of the Jagged1-treated hDPSC-derived decellularized extracellular matrix (dECM) and its biological activity on odonto/osteogenic differentiation of stem cells isolated from apical papilla (SCAPs). Methods: Bioinformatic database of Jagged1-treated hDPSCs was analyzed using NetworkAnalyst. hDPSCs seeded on the Jagged1 immobilized surface were maintained with normal or osteogenic induction medium (OM) followed by decellularization procedure, dECM-N, or dECM-OM, respectively. SCAPs were reseeded on each dECM with either the normal medium or the OM. Cell viability was determined by MTT assay. Characteristics of dECMs and SCAPs were evaluated by SEM, EDX, immunofluorescent staining, and alcian blue staining. Mineralization was assessed by alizarin red S, Von Kossa, and alkaline phosphatase staining. Statistical significance was considered at p < 0.05. Results: RNA-seq database revealed upregulation of several genes involved in ECM organization, ECM–receptor interaction, and focal adhesion in Jagged1-treated hDPSCs. Immobilized Jagged1 increased the osteogenesis of the hDPSC culture with OM. dECMs showed fibrillar-like network structure and maintained major ECM proteins, fibronectin, type I-collagen, and glycosaminoglycans. A decrease in calcium and phosphate components was observed in dECMs after the decellularized process. Cell viability on dECMs did not alter by 7 days. Cell attachment and f-actin cytoskeletal organization of SCAPs proliferated on Jagged1-treated dECMs were comparable to those of the control dECMs. SCAPs exhibited significantly higher mineralization on dECM-N in OM and markedly enhanced on dECM-OM with normal medium or OM conditions. Conclusion: Jagged1-treated hDPSC-derived dECMs are biocompatible and increase odonto/osteogenic differentiation of SCAPs. The results suggested the potential of Jagged1 dECMs, which could be further developed into ECM scaffolds for application in regenerative medicine.
Collapse
Affiliation(s)
- Suphalak Phothichailert
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Nunthawan Nowwarote
- Universite Paris Cite, Faculty of Dentistry, Department of Oral Biology, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Universite, INSERM UMRS, Molecular Oral Pathophysiology, Paris, France
| | - Benjamin P.J. Fournier
- Universite Paris Cite, Faculty of Dentistry, Department of Oral Biology, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Universite, INSERM UMRS, Molecular Oral Pathophysiology, Paris, France
| | - Vorapat Trachoo
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sittiruk Roytrakul
- Proteomics Research Laboratory, Genome Institute, National Center of Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Worachat Namangkalakul
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- *Correspondence: Worachat Namangkalakul,
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Zhu Q, Chen T, Xia J, Jiang D, Wang S, Zhang Y. Preparation and characterization of two novel osteoinductive fishbone-derived biphasic calcium phosphate bone graft substitutes. J Biomater Appl 2022; 37:600-613. [PMID: 35775433 DOI: 10.1177/08853282221111969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Many studies have reported on the conversion of natural resources into xenografts with hydroxyapatite (HA) as major component, but the extraction of biphasic calcium phosphate (HA/β-TCP) from animal bones and transformation into bone graft substitutes are rarely reported. In this research, two kinds of fish bones were made into granular porous biphasic calcium phosphate bone graft substitutes with particle sizes between 500 to 1000 μm through a series of preparation procedures (Salmo salar calcined at 900°C named Sa900 and Anoplopoma fimbria calcined at 800°C named An800). The chemical composition was characterized by X-ray diffraction (XRD) and Fourier transform infrared spectroscopy (FTIR). The morphology and porous structure of the scaffolds were comparatively analyzed by scanning electron microscopy (SEM) and mercury porosimeter. The specific surface area of materials was measured by the nitrogen adsorption technique based on BET theory. Cytotoxicity and ectopic osteogenesis were also carried out to investigate the biocompatibility and osteoinductive potential of these materials. The results showed that both fishbone-derived scaffolds were composed of HA and β-TCP with different proportions, and numerous interconnected pores with different sizes were observed at the surface of materials. An800 had higher total porosity reaching 74.8% with higher interconnectivity and micropores mostly distributed at 0.27 μm and 0.12 μm, while Sa900 had a higher specific surface area and higher intraparticle porosity with nanopores mostly distributed at 0.07 μm. CCK-8 assays and Live/dead staining demonstrated excellent biocompatibility. Material-induced osteoid formation were observed on the interface of both internal pores and periphery of materials after implantation in muscle pouch of Wistar rats for 8 weeks which indicated some extent of osteoinductive potential of materials. The possible mechanism of material-induced osteogenesis and the effects of chemical composition, surface topography, and spatial structure on osteogenesis were also discussed in this paper.
Collapse
Affiliation(s)
- Qingfeng Zhu
- Department of Stomatology, 12520Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Tong Chen
- Department of Stomatology, 12520Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jinfeng Xia
- 58306Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai, China
| | - Danyu Jiang
- 58306Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai, China
| | - Shaohai Wang
- Department of Stomatology, 12476Dongfang Hospital, Tongji University, Shanghai 200092, China
| | - Yuntong Zhang
- Department of Orthopeadics, 12476Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
7
|
Cámara-Torres M, Sinha R, Sanchez A, Habibovic P, Patelli A, Mota C, Moroni L. Effect of high content nanohydroxyapatite composite scaffolds prepared via melt extrusion additive manufacturing on the osteogenic differentiation of human mesenchymal stromal cells. BIOMATERIALS ADVANCES 2022; 137:212833. [PMID: 35929265 DOI: 10.1016/j.bioadv.2022.212833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
The field of bone tissue engineering seeks to mimic the bone extracellular matrix composition, balancing the organic and inorganic components. In this regard, additive manufacturing (AM) of high content calcium phosphate (CaP)-polymer composites holds great promise towards the design of bioactive scaffolds. Yet, the biological performance of such scaffolds is still poorly characterized. In this study, melt extrusion AM (ME-AM) was used to fabricate poly(ethylene oxide terephthalate)/poly(butylene terephthalate) (PEOT/PBT)-nanohydroxyapatite (nHA) scaffolds with up to 45 wt% nHA, which presented significantly enhanced compressive mechanical properties, to evaluate their in vitro osteogenic potential as a function of nHA content. While osteogenic gene upregulation and matrix mineralization were observed on all scaffold types when cultured in osteogenic media, human mesenchymal stromal cells did not present an explicitly clear osteogenic phenotype, within the evaluated timeframe, in basic media cultures (i.e. without osteogenic factors). Yet, due to the adsorption of calcium and inorganic phosphate ions from cell culture media and simulated body fluid, the formation of a CaP layer was observed on PEOT/PBT-nHA 45 wt% scaffolds, which is hypothesized to account for their bone forming ability in the long term in vitro, and osteoconductivity in vivo.
Collapse
Affiliation(s)
- Maria Cámara-Torres
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Ravi Sinha
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Alberto Sanchez
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 Donostia-San Sebastian, Spain
| | - Pamela Habibovic
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Instructive Biomaterial Engineering Department, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Alessandro Patelli
- Department of Physics and Astronomy, Padova University, Via Marzolo, 8, 35131 Padova, Italy
| | - Carlos Mota
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Lorenzo Moroni
- Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands.
| |
Collapse
|
8
|
MTA-Based Cements: Biocompatibility and Effects on the Gene Expression of Collagen Type 1 and TGF- β1. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2204698. [PMID: 35402617 PMCID: PMC8989590 DOI: 10.1155/2022/2204698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/31/2022]
Abstract
Objective This study sought to evaluate the biocompatibility of Neomineral Trioxide Aggregate (Neo-MTA), MTA Repair High Plasticity (MTA-HP), and Mineral Trioxide Aggregate-Angelus white (MTA-Ang) in fibroblasts of human dental pulp. Materials and Methods Morphology was evaluated after 24 h of incubation. LIVE/DEAD assay and cell adhesion tests were performed at 24 h of treatment. Cell proliferation assays (MTSs) and Annexin V were performed at 48 h incubation with different treatments. The expression of Col-1 and TGF-β1 was tested by endpoint PCR at 5 days of treatment. Results Morphological changes were observed in all groups. Neo-MTA and MTA-Ang were associated with increased cell viability, and all materials induced apoptosis, with a higher percentage in the MTA-HP group than in the other groups. In the LIVE/DEAD assay, there was more damage to the cell membrane in the group of cells treated with MTA-HP than in the other groups. Conclusion Neo-MTA and MTA-Ang presented similar biocompatibility, and both showed greater biocompatibility than MTA-HP. MTA-HP and MTA-Ang increased Col-1A gene expression, and Neo-MTA and MTA-Ang increased TGF-β1 gene expression in a similar way.
Collapse
|
9
|
Cetin Genc C, Yilmaz HD, Karaca B, Kiran F, Arslan YE. Nano-hydroxyapatite incorporated quince seed mucilage bioscaffolds for osteogenic differentiation of human adipose-derived mesenchymal stem cells. Int J Biol Macromol 2022; 195:492-505. [PMID: 34921891 DOI: 10.1016/j.ijbiomac.2021.12.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022]
Abstract
In this study, the therapeutic hydrocolloid quince seed mucilage (QSM) from Cydonia oblonga Miller fruit is enriched with needle-like nano-hydroxyapatite (nHAp) crystals to fabricate a novel biomimetic osteogenic bioscaffold. The molecular weight (Mw) of water-based extracted QSM was measured with GPC (8.67 × 105 g/mol), and the composite blend was prepared at a ratio of 1:1 (w/w) QSMaq and nHAp. The porous bioscaffolds were manufactured by the freeze-drying method, and evaluated in-depth with advanced analyses. The XRD, ATR-FTIR, SEM-EDX, and elemental mapping analyses revealed a uniform coated semi-crystalline structure with no covalent bindings between QSM and nHAp. Moreover, due to the hydrocolloid backbone, a supreme swelling ratio (w/w, 6523 ± 190%) with suitable pore size (208.12 ± 99.22 μm) for osteogenic development was obtained. Further, the cytocompatible bioscaffolds were evaluated for osteogenic differentiation in vitro using human adipose-derived mesenchymal stem cells (hAMSCs). The immuno/histochemical (I/HC) staining revealed that the cells with the spherical morphology invaded the pores of the prepared bioscaffolds. Also, relatively early up-regulated osteogenic markers were observed by the qRT-PCR analyses. Overall, it is believed that the QSM-nHAp bioscaffolds might be favorable in non-load bearing applications, especially in the cranio-maxillofacial region, due to their regenerative, bendable, and durable features.
Collapse
Affiliation(s)
- Cigdem Cetin Genc
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey
| | - Hilal Deniz Yilmaz
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey
| | - Burak Karaca
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, 06100 Ankara, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, 17100 Canakkale, Turkey.
| |
Collapse
|
10
|
Svensson S, Palmer M, Svensson J, Johansson A, Engqvist H, Omar O, Thomsen P. Monocytes and pyrophosphate promote mesenchymal stem cell viability and early osteogenic differentiation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:11. [PMID: 35032239 PMCID: PMC8761140 DOI: 10.1007/s10856-021-06639-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
Pyrophosphate-containing calcium phosphate implants promote osteoinduction and bone regeneration. The role of pyrophosphate for inflammatory cell-mesenchymal stem cell (MSC) cross-talk during osteogenesis is not known. In the present work, the effects of lipopolysaccharide (LPS) and pyrophosphate (PPi) on primary human monocytes and on osteogenic gene expression in human adipose-derived MSCs were evaluated in vitro, using conditioned media transfer as well as direct effect systems. Direct exposure to pyrophosphate increased nonadherent monocyte survival (by 120% without LPS and 235% with LPS) and MSC viability (LDH) (by 16-19% with and without LPS). Conditioned media from LPS-primed monocytes significantly upregulated osteogenic genes (ALP and RUNX2) and downregulated adipogenic (PPAR-γ) and chondrogenic (SOX9) genes in recipient MSCs. Moreover, the inclusion of PPi (250 μM) resulted in a 1.2- to 2-fold significant downregulation of SOX9 in the recipient MSCs, irrespective of LPS stimulation or culture media type. These results indicate that conditioned media from LPS-stimulated inflammatory monocytes potentiates the early MSCs commitment towards the osteogenic lineage and that direct pyrophosphate exposure to MSCs can promote their viability and reduce their chondrogenic gene expression. These results are the first to show that pyrophosphate can act as a survival factor for both human MSCs and primary monocytes and can influence the early MSC gene expression. Graphical abstract.
Collapse
Affiliation(s)
- Sara Svensson
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Michael Palmer
- Department of Engineering Sciences, Applied Materials Science Section, Uppsala University, Uppsala, Sweden
| | - Johan Svensson
- Department of Statistics, Umeå School of Business, Economics and Statistics, Umeå University, Umeå, Sweden
| | - Anna Johansson
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Håkan Engqvist
- Department of Engineering Sciences, Applied Materials Science Section, Uppsala University, Uppsala, Sweden
| | - Omar Omar
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Peter Thomsen
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
11
|
de Melo Pereira D, Davison N, Habibović P. Human osteoclast formation and resorptive function on biomineralized collagen. Bioact Mater 2021; 8:241-252. [PMID: 34541399 PMCID: PMC8424427 DOI: 10.1016/j.bioactmat.2021.06.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
Biomineralized collagen composite materials pose an intriguing alternative to current synthetic bone graft substitutes by offering a biomimetic composition that closely resembles native bone. We hypothesize that this composite can undergo cellular resorption and remodeling similar to natural bone. We investigate the formation and activity of human osteoclasts cultured on biomineralized collagen and pure collagen membranes in comparison to cortical bone slices. Human monocytes/macrophages from peripheral blood differentiate into multinucleated, tartrate-resistant alkaline phosphatase (TRAP)-positive osteoclast-like cells on all substrates. These cells form clear actin rings on cortical bone, but not on biomineralized collagen or pure collagen membranes. Osteoclasts form resorption pits in cortical bone, resulting in higher calcium ion concentration in cell culture medium; however, osteoclast resorption of biomineralized collagen and collagen membranes does not measurably occur. Activity of osteoclast enzymes – TRAP, carbonic anhydrase II (CA-II), and cathepsin-K (CTS-K) – is similar on all substrates, despite phenotypic differences in actin ring formation and resorption. The mesh-like structure, relatively low stiffness, and lack of RGD-containing binding domains are likely the factors responsible for preventing formation of stable actin rings on and resorption of (biomineralized) collagen membranes. This insight helps to guide further research toward the optimized design of biomineralized collagen composites as a more biomimetic bone-graft substitute. Human osteoclasts differentiated on biomineralized collagen, but no resorption occured. Porous surface of fibrillary collagen and lack of RGD ligands to the vitronectin receptor likely prevented osteoclast resorption. Quantification of resorption on uneven surfaces in comparison with bone was possible with 3D laser microscopy.
Collapse
Affiliation(s)
- Daniel de Melo Pereira
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Noel Davison
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
12
|
Boller LA, Shiels SM, Florian DC, Peck SH, Schoenecker JG, Duvall C, Wenke JC, Guelcher SA. Effects of nanocrystalline hydroxyapatite concentration and skeletal site on bone and cartilage formation in rats. Acta Biomater 2021; 130:485-496. [PMID: 34129957 DOI: 10.1016/j.actbio.2021.05.056] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/29/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Most fractures heal by a combination of endochondral and intramembranous ossification dependent upon strain and vascularity at the fracture site. Many biomaterials-based bone regeneration strategies rely on the use of calcium phosphates such as nano-crystalline hydroxyapatite (nHA) to create bone-like scaffolds. In this study, nHA was dispersed in reactive polymers to form composite scaffolds that were evaluated both in vitro and in vivo. Matrix assays, immunofluorescent staining, and Western blots demonstrated that nHA influenced mineralization and subsequent osteogenesis in a dose-dependent manner in vitro. Furthermore, nHA dispersed in polymeric composites promoted osteogenesis by a similar mechanism as particulated nHA. Scaffolds were implanted into a 2-mm defect in the femoral diaphysis or metaphysis of Sprague-Dawley rats to evaluate new bone formation at 4 and 8 weeks. Two formulations were tested: a poly(thioketal urethane) scaffold without nHA (PTKUR) and a PTKUR scaffold augmented with 22 wt% nHA (22nHA). The scaffolds supported new bone formation in both anatomic sites. In the metaphysis, augmentation of scaffolds with nHA promoted an intramembranous healing response. Within the diaphysis, nHA inhibited endochondral ossification. Immunohistochemistry was performed on cryo-sections of the bone/scaffold interface in which CD146, CD31, Endomucin, CD68, and Myeloperoxidase were evaluated. No significant differences in the infiltrating cell populations were observed. These findings suggest that nHA dispersed in polymeric composites induces osteogenic differentiation of adherent endogenous cells, which has skeletal site-specific effects on fracture healing. STATEMENT OF SIGNIFICANCE: Understanding the mechanism by which synthetic scaffolds promote new bone formation in preclinical models is crucial for bone regeneration applications in the clinic where complex fracture cases are seen. In this study, we found that dispersion of nHA in polymeric scaffolds promoted in vitro osteogenesis in a dose-dependent manner through activation of the PiT1 receptor and subsequent downstream Erk1/2 signaling. While augmentation of polymeric scaffolds with nHA enhanced intramembranous ossification in metaphyseal defects, it inhibited endochondral ossification in diaphyseal defects. Thus, our findings provide new insights into designing synthetic bone grafts that complement the skeletal site-specific fracture healing response.
Collapse
|
13
|
Tang Z, Chen S, Ni Y, Zhao R, Zhu X, Yang X, Zhang X. Role of Na +, K +-ATPase ion pump in osteoinduction. Acta Biomater 2021; 129:293-308. [PMID: 34087440 DOI: 10.1016/j.actbio.2021.05.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/14/2022]
Abstract
Porous biphasic calcium phosphate bioceramic (BCP) possesses osteoinductivity to induce the osteoblastic commitment of mesenchymal stem cells (MSCs) and ectopic bone formation. However, the underlying mechanism remains enigmatic. We performed a gene array analysis of MSCs cocultured with BCP to screen for candidate osteoinductive modulators. Na+, K+-ATPase (NKA), an ion transporter, therefore was identified as a crucial ion transporter in regulating the osteogenesis of the cells. NKA activator, a polyclonal antibody, enriched the cytomembrane abundance of NKA and lead to an enhanced osteogenic effect of BCP. As indicated in gene array analysis and suggested by co-immunoprecipitation assay, protein phosphatase 2A (PP2A) was elevated by BCP to dephosphorylate NKA and prevent its endocytosis. The inhibition of NKA by ouabain resulted in an adverse effect on osteoinductivity of BCP. We further altered NKA activity in mice implanted with BCP and found that the intensity and incidence of osteoinduction was increased by the NKA activator. We went one step further by investigating the potential of targeting NKA in osteoporotic bone regeneration. Activating NKA upregulated osteogenic gene expression and calcium deposition ability of osteoporotic osteoblasts. Furthermore, activation of NKA in mice ameliorated estrogen-deficiency induced bone loss, in terms of increased bone mass and improved bending strength. With this osteoinductive bioceramic derived ion transporter target, we demonstrate that the activation of NKA has significant potential to revolutionize the regeneration of bone. STATEMENT OF SIGNIFICANCE: In this study, we identified an important role of Na+, K+-ATPase (NKA) have played in osteoinductivity of biphasic calcium phosphate bioceramic (BCP). Furthermore, we demonstrated the therapeutic potential of targeting NKA in osteoporotic bone regeneration. Numerous gene and protein targets to treat osteoporosis were discovered every year, mainly obtained by genomic and proteomic screenings of a large population. In contrast, our study identified an unrevealed bone regenerating target from the upregulated genes induced by an osteoinductive biomaterial. The approach was cost-saving since it did not require a large sample pool. Furthermore, the target derived from this approach was proven to be anabolic. Identification of an anabolic agent holds significant value since most of the current anti-osteoporotic therapies are antiresorptive.
Collapse
|
14
|
Chen W, Zhu WQ, Qiu J. Impact of exogenous metal ions on peri-implant bone metabolism: a review. RSC Adv 2021; 11:13152-13163. [PMID: 35423842 PMCID: PMC8697588 DOI: 10.1039/d0ra09395e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/25/2021] [Indexed: 11/21/2022] Open
Abstract
The development of effective methods to promote the osseointegration of dental implants by surface modification is an area of intense research in dental materials science. Exogenous metal ions present in the implant and surface modifications are closely related to the bone metabolism around the implant. In the complex oral microenvironment, the release of metal ions caused by continuous corrosion of dental implants has an unfavorable impact on the surrounding tissue, and then affects osseointegration, leading to bad results such as loosening and falling off in the late stage of the implant. Besides, these ions can even be distributed in distant tissues and organs. Currently, surface modification techniques are being developed that involve different processing technologies including the introduction of exogenous metal ions with different properties onto the surface of implants to improve performance. However, most metal elements have some level of biological toxicity and can only be used within a safe concentration range to exert the optimum biological effects on recipients. In this paper, we review the adverse effects of metal ions on osseointegration and highlight the emerging applications for metal elements in improving the performance of dental implants.
Collapse
Affiliation(s)
- Wei Chen
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing 210029 PR China +86 25 69593085
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University Nanjing 210029 PR China
| | - Wen-Qing Zhu
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing 210029 PR China +86 25 69593085
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University Nanjing 210029 PR China
| | - Jing Qiu
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing 210029 PR China +86 25 69593085
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University Nanjing 210029 PR China
| |
Collapse
|
15
|
Le J, Zhongqun L, Zhaoyan W, Yijun S, Yingjin W, Yaojie W, Yanan J, Zhanrong J, Chunyang M, Fangli G, Nan X, Lingyun Z, Xiumei W, Qiong W, Xiong L, Xiaodan S. Development of methods for detecting the fate of mesenchymal stem cells regulated by bone bioactive materials. Bioact Mater 2021; 6:613-626. [PMID: 33005826 PMCID: PMC7508719 DOI: 10.1016/j.bioactmat.2020.08.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 01/07/2023] Open
Abstract
The fate of mesenchymal stem cells (MSCs) is regulated by biological, physical and chemical signals. Developments in biotechnology and materials science promoted the occurrence of bioactive materials which can provide physical and chemical signals for MSCs to regulate their fate. In order to design and synthesize materials that can precisely regulate the fate of MSCs, the relationship between the properties of materials and the fate of mesenchymal stem cells need to be clarified, in which the detection of the fate of mesenchymal stem cells plays an important role. In the past 30 years, a series of detection technologies have been developed to detect the fate of MSCs regulated by bioactive materials, among which high-throughput technology has shown great advantages due to its ability to detect large amounts of data at one time. In this review, the latest research progresses of detecting the fate of MSCs regulated by bone bioactive materials (BBMs) are systematically reviewed from traditional technology to high-throughput technology which is emphasized especially. Moreover, current problems and the future development direction of detection technologies of the MSCs fate regulated by BBMs are prospected. The aim of this review is to provide a detection technical framework for researchers to establish the relationship between the properties of BMMs and the fate of MSCs, so as to help researchers to design and synthesize BBMs better which can precisely regulate the fate of MSCs.
Collapse
Affiliation(s)
- Jiang Le
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Liu Zhongqun
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wang Zhaoyan
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, 100084, People's Republic of China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, People's Republic of China
- School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Su Yijun
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wang Yingjin
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wei Yaojie
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Jiang Yanan
- Key Lab of Advanced Technologies of Materials of Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, People's Republic of China
| | - Jia Zhanrong
- Key Lab of Advanced Technologies of Materials of Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, People's Republic of China
| | - Ma Chunyang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Gang Fangli
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Xu Nan
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Zhao Lingyun
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wang Xiumei
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wu Qiong
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, 100084, People's Republic of China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, People's Republic of China
- School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Lu Xiong
- Key Lab of Advanced Technologies of Materials of Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, People's Republic of China
| | - Sun Xiaodan
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| |
Collapse
|
16
|
Rozila I, Azari P, Munirah S, Safwani WKZW, Pingguan-Murphy B, Chua KH. Polycaprolactone-Based Scaffolds Facilitates Osteogenic Differentiation of Human Adipose-Derived Stem Cells in a Co-Culture System. Polymers (Basel) 2021; 13:polym13040597. [PMID: 33671175 PMCID: PMC7922582 DOI: 10.3390/polym13040597] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Stem cells in combination with scaffolds and bioactive molecules have made significant contributions to the regeneration of damaged bone tissues. A co-culture system can be effective in enhancing the proliferation rate and osteogenic differentiation of the stem cells. Hence, the aim of this study was to investigate the osteogenic differentiation of human adipose derived stem cells when co-cultured with human osteoblasts and seeded on polycaprolactone (PCL):hydroxyapatite (HA) scaffold; (2) Methods: Human adipose-derived stem cells (ASC) and human osteoblasts (HOB) were seeded in three different ratios of 1:2, 1:2 and 2:1 in the PCL-HA scaffolds. The osteogenic differentiation ability was evaluated based on cell morphology, proliferation rate, alkaline phosphatase (ALP) activity, calcium deposition and osteogenic genes expression levels using quantitative RT-PCR; (3) Results: The co-cultured of ASC/HOB in ratio 2:1 seeded on the PCL-HA scaffolds showed the most positive osteogenic differentiation as compared to other groups, which resulted in higher ALP activity, calcium deposition and osteogenic genes expression, particularly Runx, ALP and BSP. These genes indicate that the co-cultured ASC/HOB seeded on PCL-HA was at the early stage of osteogenic development; (4) Conclusions: The combination of co-culture system (ASC/HOB) and PCL-HA scaffolds promote osteogenic differentiation and early bone formation.
Collapse
Affiliation(s)
- Ismail Rozila
- Department of Physiology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Cyberjaya, Selangor 63000, Malaysia
| | - Pedram Azari
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Sha’ban Munirah
- Department of Rehabilitation and Physiotherapy, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan 25200, Malaysia;
| | - Wan Kamarul Zaman Wan Safwani
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (W.K.Z.W.S.); (B.P.-M.)
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (W.K.Z.W.S.); (B.P.-M.)
| | - Kien Hui Chua
- Department of Physiology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Correspondence: ; Tel.: +60-391458613
| |
Collapse
|
17
|
Ferreira SA, Young G, Jones JR, Rankin S. Bioglass/carbonate apatite/collagen composite scaffold dissolution products promote human osteoblast differentiation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111393. [PMID: 33254998 DOI: 10.1016/j.msec.2020.111393] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/30/2022]
Abstract
OssiMend® Bioactive (Collagen Matrix Inc., NJ) is a three-component porous composite bone graft device of 45S5 Bioglass/carbonate apatite/collagen. Our in vitro studies showed that conditioned media of the dissolution products of OssiMend Bioactive stimulated primary human osteoblasts to form mineralized bone-like nodules in vitro in one week, in basal culture media (no osteogenic supplements). Osteoblast differentiation was followed by gene expression analysis and a mineralization assay. In contrast, the dissolution products from commercial OssiMend (Bioglass-free carbonate apatite/collagen scaffolds), or from 45S5 Bioglass particulate alone, did not induce the mineralization of the extracellular matrix, but did induce osteoblast differentiation to mature osteoblasts, evidenced by the strong upregulation of BGLAP and IBSP mRNA levels. The calcium ions and soluble silicon species released from 45S5 Bioglass particles and additional phosphorus release from OssiMend mediated the osteostimulatory effects. Medium conditioned with OssiMend Bioactive dissolution had a much higher concentration of phosphorus and silicon than media conditioned with OssiMend and 45S5 Bioglass alone. While OssiMend and OssiMend Bioactive led to calcium precipitation in cell culture media, OssiMend Bioactive produced a higher concentration of soluble silicon than 45S5 Bioglass and higher dissolution of phosphorus than OssiMend. These in vitro results suggest that adding 45S5 Bioglass to OssiMend produces a synergistic osteostimulation effect on primary human osteoblasts. In summary, dissolution products of a Bioglass/carbonate apatite/collagen composite scaffold (OssiMend® Bioactive) stimulate human osteoblast differentiation and mineralization of extracellular matrix in vitro without any osteogenic supplements. The mineralization was faster than for dissolution products of ordinary Bioglass.
Collapse
Affiliation(s)
- Silvia A Ferreira
- National Heart & Lung Institute, Imperial College London, London, UK.
| | - Gloria Young
- Department of Materials, Imperial College London, London, UK.
| | - Julian R Jones
- Department of Materials, Imperial College London, London, UK.
| | - Sara Rankin
- National Heart & Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
18
|
Nanosheets-incorporated bio-composites containing natural and synthetic polymers/ceramics for bone tissue engineering. Int J Biol Macromol 2020; 164:1960-1972. [DOI: 10.1016/j.ijbiomac.2020.08.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/20/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
|
19
|
Zheng G, Xie ZY, Wang P, Wu YF, Shen HY. Recent advances of single-cell RNA sequencing technology in mesenchymal stem cell research. World J Stem Cells 2020; 12:438-447. [PMID: 32742561 PMCID: PMC7360991 DOI: 10.4252/wjsc.v12.i6.438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/13/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells with great potential for clinical applications. However, little is known about their cell heterogeneity at a single-cell resolution, which severely impedes the development of MSC therapy. In this review, we focus on advances in the identification of novel surface markers and functional subpopulations of MSCs made by single-cell RNA sequencing and discuss their participation in the pathophysiology of stem cells and related diseases. The challenges and future directions of single-cell RNA sequencing in MSCs are also addressed in this review.
Collapse
Affiliation(s)
- Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Zhong-Yu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yan-Feng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Hui-Yong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
20
|
Othman Z, Mohren R, Cillero-Pastor B, Shen Z, Lacroix Y, Guttenplan A, Tahmasebi Birgani Z, Eijssen L, Luider T, van Rijt S, Habibovic P. Comparative proteomic analysis of human mesenchymal stromal cell behavior on calcium phosphate ceramics with different osteoinductive potential. Mater Today Bio 2020; 7:100066. [PMID: 32642640 PMCID: PMC7334494 DOI: 10.1016/j.mtbio.2020.100066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 01/13/2023] Open
Abstract
In recent years, synthetic calcium phosphate (CaP) ceramics have emerged as an alternative to bone grafts in the treatment of large critical-sized bone defects. To successfully substitute for bone grafts, materials must be osteoinductive, that is, they must induce osteogenic differentiation and subsequent bone formation in vivo. Although a set of osteoinductive CaP ceramics has been developed, the precise biological mechanism by which a material directs cells toward osteogenesis and the role of individual chemical and physical properties in this mechanism remain incompletely understood. Here, we used proteomics to compare serum protein adsorption to two CaP ceramics with different osteoinductive potential, namely an osteoinductive β-tricalcium phosphate (TCP) and a non-osteoinductive hydroxyapatite (HA). Moreover, we analyzed the protein profiles of human mesenchymal stromal cells (hMSCs) cultured on these two ceramics. The serum protein adsorption experiments in the absence of cells highlighted the proteins that are highly abundant in the serum and/or have a high affinity to CaP. The extent of adsorption was suggested to be affected by the available surface area for binding and by the ion exchange dynamics on the surface. Several proteins were uniquely expressed by hMSCs on TCP and HA surfaces. Proteins identified as enriched on TCP were involved in processes related to wound healing, cell proliferation, and the production of extracellular matrix. On the other hand, proteins that were enriched on HA were involved in processes related to protein production, translation, localization, and secretion. In addition, we performed a separate proteomics analysis on TCP, HA, and two biphasic calcium phosphates with known osteoinductive potential and performed a clustering analysis on a combination of a set of proteins found to be enriched on osteoinductive materials with a set of proteins already known to be involved in osteogenesis. This yielded two protein networks potentially involved in the process of osteoinduction – one consisting of collagen fragments and collagen-related enzymes and a second consisting of endopeptidase inhibitors and regulatory proteins. The results of this study show that protein profiling can be a useful tool to help understand the effect of biomaterial properties on the interactions between a biomaterial and a biological system. Such understanding will contribute to the design and development of improved biomaterials for (bone) regenerative therapies.
Collapse
Affiliation(s)
- Z. Othman
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - R.J.C. Mohren
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - B. Cillero-Pastor
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Z. Shen
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Y.S.N.W. Lacroix
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - A.P.M. Guttenplan
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Z. Tahmasebi Birgani
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - L. Eijssen
- Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs School for Mental Health and Neuroscience Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - T.M. Luider
- Laboratory of Neuro-Oncology and Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Center, Wytemaweg 80, P.O. Box 2040, 3000 CA, Rotterdam, the Netherlands
| | - S. van Rijt
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - P. Habibovic
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
- Corresponding author.
| |
Collapse
|
21
|
Hybrid Bone Scaffold Induces Bone Bridging in Goat Calvarial Critical Size Defects Without Growth Factor Augmentation. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-019-00144-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract
In the present study, a series of four different scaffolds were comparatively evaluated in a goat calvarial critical size defect model. Such studies are only rarely reported in the literature. In our work, E1001(1k), a member of a large combinational library of tyrosine-derived polycarbonates (TyrPC), was used to prepare two calcium phosphate hybrid, biodegradable bone scaffolds. In one formulation, the widely used β-tricalcium phosphate (β-TCP) was incorporated into the polymer scaffold. In the second formulation, a coating of dicalcium phosphate dihydrate (DCPD, also known as brushite) was used as the mineral phase. These scaffolds were evaluated for bone regeneration in goat calvarial 20-mm critical size defects (CSD) after 16 weeks. Results were compared with chronOS (a clinically used product) and E1001(1k)/β-TCP scaffolds, augmented with 400 μg of recombinant human bone morphogenetic protein-2 (rhBMP-2). Microcomputed tomography (micro-CT) and histomorphometry were used to assess bone regeneration within the defects. Histomorphometry showed that rhBMP-2-augmented E1001(1k)/β-TCP scaffolds completely healed the defect in all animals within 16 weeks. Among the hybrid scaffolds that were not augmented with rhBMP-2, the degree of bone regeneration within the defect area was low for the clinically used chronOS, which is a poly(lactide co-ε-caprolactone)/β-TCP hybrid scaffold. Similar results were obtained for E1001(1k)/β-TCP scaffolds, indicating that replacing poly(lactide co-ε-caprolactone) with E1001(1k) does not improve bone regeneration is this model. However, a statistically significant improvement of bone regeneration was observed for E1001(1k)/DCPD scaffolds. These scaffolds resulted in significant levels of bone regeneration in all animals and in complete bridging of the defect in three of six tests. This is the first report of a synthetic bone scaffold being able to heal a critical size calvarial defect in a large animal model without the addition of exogenous growth factors.
Lay Summary
Reconstruction of large bone defects is a significant clinical problem. The overwhelming majority of all research results are obtained in vitro or in small animal models (mouse, rat, rabbit) that cannot predict the clinical outcomes in humans. We address this problem by conducting our studies in a goat calvarial critical size defect model, which is widely regarded as predictive of human outcomes. Among the three rhBMP-2-free scaffolds tested, only one specific formulation, E1001(1k)/DCPD, resulted in massive bone ingrowth into the center of the defect in all animals and in complete bridging of the defect 50% of the animals. This is the first time, a synthetic bone scaffold was able to heal a critical size calvarial defect in a large animal model without the addition of biological growth factors. Given the high cost of biologically enhanced bone grafts and the regulatory complexities of their FDA market clearance, the development of E1001(1k)/DCPD hybrid scaffolds addresses a significant clinical need.
Collapse
|
22
|
Zhu J, Yang S, Cai K, Wang S, Qiu Z, Huang J, Jiang G, Wang X, Fang X. Bioactive poly (methyl methacrylate) bone cement for the treatment of osteoporotic vertebral compression fractures. Theranostics 2020; 10:6544-6560. [PMID: 32483469 PMCID: PMC7255031 DOI: 10.7150/thno.44428] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: Poly (methyl methacrylate) (PMMA) bone cement is one of the most commonly used biomaterials for augmenting/stabilizing osteoporosis-induced vertebral compression fractures (OVCFs), such as percutaneous vertebroplasty (PVP) and balloon kyphoplasty (BKP). However, its clinical applications are limited by its poor performance in high compressive modulus and weak bonding to bone. To address these issues, a bioactive composite bone cement was developed for the treatment of osteoporotic vertebral compression fractures, in which mineralized collagen (MC) was incorporated into the PMMA bone cement (MC-PMMA). Methods: The in vitro properties of PMMA and MC-PMMA composite bone cement were determined, including setting time, compressive modulus, adherence, proliferation, and osteogenic differentiation of rat bone mesenchymal stem cells. The in vivo properties of both cements were evaluated in an animal study (36 osteoporotic New Zealand female rabbits divided equally between the two bone cement groups; PVP at L5) and a small-scale and short-term clinical study (12 patients in each of the two bone cement groups; follow-up: 2 years). Results: In terms of value for PMMA bone cement, the handling properties of MC-PMMA bone cement were not significantly different. However, both compressive strength and compressive modulus were found to be significantly lower. In the rabbit model study, at 8 and 12 weeks post-surgery, bone regeneration was more significant in MC-PMMA bone cement (cortical bone thickness, osteoblast area, new bone area, and bone ingrowth %; each significantly higher). In the clinical study, at a follow-up of 2 years, both the Visual Analogue Score and Oswestry Disability Index were significantly reduced when MC-PMMA cement was used. Conclusions: MC-PMMA bone cement demonstrated good adaptive mechanical properties and biocompatibility and may be a promising alternative to commercial PMMA bone cements for the treatment of osteoporotic vertebral fractures in clinical settings. While the present results for MC-PMMA bone cement are encouraging, further study of this cement is needed to explore its viability as an ideal alternative for use in PVP and BKP.
Collapse
Affiliation(s)
- Jinjin Zhu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
- Department of Spinal Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo 315020, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Kaiwen Cai
- Department of Spinal Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo 315020, China
| | - Shuo Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zhiye Qiu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Junfei Huang
- Shimadzu (China) Co., Ltd. Shenzhen Branch, Shenzhen 518042, China
| | - Guoqiang Jiang
- Department of Spinal Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo 315020, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiangqian Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| |
Collapse
|
23
|
Maazouz Y, Rentsch I, Lu B, Santoni BLG, Doebelin N, Bohner M. In vitro measurement of the chemical changes occurring within β-tricalcium phosphate bone graft substitutes. Acta Biomater 2020; 102:440-457. [PMID: 31756552 DOI: 10.1016/j.actbio.2019.11.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/18/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Several mechanisms proposed to explain the osteoinductive potential of calcium phosphates involve surface mineralization ("bioactivity") and mention the occurrence of concentration gradients between the inner and the outer part of the implanted material. Determining the evolution of the local chemical environment occurring inside the pores of an implanted bone graft substitute (BGS) is therefore highly relevant. A quantitative and fast method was developed to measure the chemical changes occurring within the pores of β-Tricalcium Phosphate (β-TCP) granules incubated in a simulated body fluid. A factorial design of experiment was used to test the effect of particle size, specific surface area, microporosity, and purity of the β-TCP granules. Large pH, calcium and phosphate concentration changes were observed inside the BGS and lasted for several days. The kinetics and magnitude of these changes (up to 2 pH units) largely depended on the processing and properties of the granules. Interestingly, processing parameters that increased the kinetics and magnitude of the local chemical changes are parameters considered to favor calcium phosphate osteoinduction, suggesting that the model might be useful to predict the osteoinductive potential of BGSs. STATEMENT OF SIGNIFICANCE: Recent results suggest that in situ mineralization of biomaterials (polymers, ceramics, metals) might be key in their ability to trigger ectopic bone formation. This is the reason why the effect on in situ mineralization of various synthesis parameters of β-tricalcium phosphate granules was studied (size, microporosity, specific surface area, and Ca/P molar ratio). To the best of our knowledge, this is the first article devoted to the chemical changes occurring within the pores of a bone graft substitute. We believe that the manuscript will prove to be highly important in the design and mechanistic understanding of drug-free osteoinductive biomaterials.
Collapse
|
24
|
Hughes EAB, Chipara M, Hall TJ, Williams RL, Grover LM. Chemobrionic structures in tissue engineering: self-assembling calcium phosphate tubes as cellular scaffolds. Biomater Sci 2020; 8:812-822. [DOI: 10.1039/c9bm01010f] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A diverse range of complex patterns and mineralised hierarchical microstructures can be derived from chemobrionic systems. In this work, we explore chemobrionic calcium phosphate tubes as cellular scaffolds.
Collapse
Affiliation(s)
- Erik A. B. Hughes
- School of Chemical Engineering
- University of Birmingham
- UK
- NIHR Surgical Reconstruction and Microbiology Research Centre
- Queen Elizabeth Hospital
| | - Miruna Chipara
- School of Chemical Engineering
- University of Birmingham
- UK
| | - Thomas J. Hall
- School of Chemical Engineering
- University of Birmingham
- UK
| | | | - Liam M. Grover
- School of Chemical Engineering
- University of Birmingham
- UK
| |
Collapse
|
25
|
Lee CC, Hirasawa N, Garcia KG, Ramanathan D, Kim KD. Stem and progenitor cell microenvironment for bone regeneration and repair. Regen Med 2019; 14:693-702. [PMID: 31393221 DOI: 10.2217/rme-2018-0044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells reside in their native microenvironment, which provides dynamic physical and chemical cues essential to their survival, proliferation and function. A typical cell-based therapeutic approach requires the mesenchymal stem cells (MSC) to depart their native microenvironment, transplant to in-vivo environment, differentiate toward multiple lineages and participate in bone formation. The long-term survival, function and fate of MSC are dependent on the microenvironment in which they are transplanted. Transplantation of morselized autologous bone, which contains both stem cells and their native microenvironment, results in a good clinical outcome. However, implantation of bone graft substitutes does not provide the complete and dynamic microenvironment for MSC. Current bone graft therapeutics may need to be improved further to provide an optimal engineered MSC microenvironment.
Collapse
Affiliation(s)
- Charles C Lee
- Department of Cell Biology & Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | | | | | - Dinesh Ramanathan
- Department of Neurological Surgery, School of Medicine, University of California, Davis, CA, USA
| | - Kee D Kim
- Department of Neurological Surgery, School of Medicine, University of California, Davis, CA, USA
| |
Collapse
|
26
|
Bageshlooyafshar B, Vakilian S, Kehtari M, Eslami-Arshaghi T, Rafeie F, Ramezanifard R, Rahchamani R, Mohammadi-Sangcheshmeh A, Mostafaloo Y, Seyedjafari E. Zinc silicate mineral-coated scaffold improved in vitro osteogenic differentiation of equine adipose-derived mesenchymal stem cells. Res Vet Sci 2019; 124:444-451. [DOI: 10.1016/j.rvsc.2017.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 08/09/2017] [Accepted: 09/14/2017] [Indexed: 01/30/2023]
|
27
|
Lei Q, Lin D, Huang WX, Wu D, Chen J. [Effects of calcium ion on the migration and osteogenic differentiation of human osteoblasts]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 36:602-608. [PMID: 30593103 DOI: 10.7518/hxkq.2018.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aimed to investigate the effect of calcium ion (Ca²⁺) on the migration and osteogenic differentiation of human osteoblasts and explore the proper concentration and correlation mechanism. METHODS A series of Ca²⁺ solutions with different concentrations was prepared. Osteoblast migration was assessed by Transwell assay, and proliferation was studied via the CCK-8 colorimetric assay. The mRNA expression of osteogenic genes was examined via reverse transcription-polymerase chain reaction (RT-PCR), and the mineralized nodule was examined by alizarin red-S method. After calcium sensitive receptor (CaSR) antagonism, Ca²⁺-induced migration and osteogenic differentiation were analyzed. RESULTS In the migration experiment, 2, 4, and 6 mmol·L⁻¹ Ca²⁺ could promoted osteoblast migration at three timepoints (8, 16, and 24 h), whereas 10 mmol·L⁻¹ Ca²⁺ considerably inhibited migration at 8 h. The Ca²⁺ concentration range of 2-10 mmol·L⁻¹ could promote proliferation, osteogenic differentiation, and mineralization of human osteoblasts. Moreover, mineralization was predominantly induced by 8 and 10 mmol·L⁻¹ Ca²⁺. CaSR antagonism could reduce Ca²⁺-induced migration and osteogenic differentiation of human osteoblasts. CONCLUSIONS Low Ca²⁺ concentration favored osteoblast migration, whereas high Ca²⁺ concentration favored osteogenic differentiation. The Ca²⁺ concentrations of 4 and 6 mmol·L⁻¹ could substantially induce osteoblast migration and osteogenic differentiation, and the Ca²⁺-CaSR pathway participated in signal transduction.
Collapse
Affiliation(s)
- Qun Lei
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Dong Lin
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Wen-Xiu Huang
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Dong Wu
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Jiang Chen
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| |
Collapse
|
28
|
Othman Z, Fernandes H, Groot AJ, Luider TM, Alcinesio A, Pereira DDM, Guttenplan APM, Yuan H, Habibovic P. The role of ENPP1/PC-1 in osteoinduction by calcium phosphate ceramics. Biomaterials 2019; 210:12-24. [PMID: 31048198 DOI: 10.1016/j.biomaterials.2019.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/15/2019] [Accepted: 04/15/2019] [Indexed: 12/20/2022]
Abstract
In the past decade, calcium phosphate (CaP) ceramics have emerged as alternatives to autologous bone grafts for the treatment of large, critical-sized bone defects. In order to be effective in the regeneration of such defects, ceramics must show osteoinductive behaviour, defined as the ability to induce de novo heterotopic bone formation. While a set of osteoinductive CaP ceramics has been developed, the exact processes underlying osteoinduction, and the role of the physical and chemical properties of the ceramics, remain largely unknown. Previous studies have focused on the role of the transcriptome to shed light on the mechanism of osteoinduction at the mRNA level. To complement these studies, a proteomic analysis was performed to study the behaviour of hMSCs on osteoinductive and non-osteoinductive CaPs. The results of this analysis suggest that plasma cell glycoprotein 1 (PC-1), encoded by the ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) gene, plays a key role in the process of osteoinduction by CaP ceramics. Validation experiments have confirmed that indeed, the mRNA expression of ENPP1 and the production of PC-1 are higher on osteoinductive than on non-osteoinductive CaP ceramics, a trend that was also observed for other osteogenic markers such as bone morphogenetic protein 2 (BMP2) and osteopontin (OPN), but not for alkaline phosphatase (ALP). Our results also showed that the expression of PC-1 is restricted to those cells which are in direct contact with the CaP ceramic surface, plausibly due to the localised depletion of calcium and inorganic phosphate ions from the supersaturated cell culture medium as CaP crystallises on the ceramic surface. Replicating the surface of the osteoinductive ceramic in polystyrene resulted in a significant decrease in ENPP1 expression, suggesting that surface structural properties alone are not sufficient to induce ENPP1 expression. Finally, knocking down ENPP1 expression in hMSCs resulted in increased BMP2 expression, both at the mRNA and protein level, suggesting that ENPP1 is a negative regulator of BMP-2 signalling. Taken together, this study shows, for the first time, that ENPP1/PC-1 plays an important role in CaP-induced osteogenic differentiation of hMSCs and thus possibly osteoinduction by CaP ceramics. Furthermore, we have identified a crucial role for the interfacial (chemical) events occurring on the CaP ceramic surface in the process of osteoinduction. This knowledge can contribute to the development of new bone graft substitutes, with improved osteoinductive potential.
Collapse
Affiliation(s)
- Ziryan Othman
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Hugo Fernandes
- Faculty of Medicine, University of Coimbra, Health Science Campus, Central Unit, Azinhaga de Santa Comba, 3000-354, Coimbra, Portugal
| | - Arjan J Groot
- Department of Radiation Oncology (MaastRO), GROW - School for Oncology & Developmental Biology, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Theo M Luider
- Laboratory of Neuro-Oncology and Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, the Netherlands
| | - Alessandro Alcinesio
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, 12 Mansfield Rd, OX1 3TA, Oxford, UK
| | - Daniel de Melo Pereira
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Alexander P M Guttenplan
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Huipin Yuan
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands.
| |
Collapse
|
29
|
Pereira RC, Benelli R, Canciani B, Scaranari M, Daculsi G, Cancedda R, Gentili C. Beta-tricalcium phosphate ceramic triggers fast and robust bone formation by human mesenchymal stem cells. J Tissue Eng Regen Med 2019; 13:1007-1018. [PMID: 30811859 DOI: 10.1002/term.2848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022]
Abstract
Due to their osteoconductive and inductive properties, a variety of calcium phosphate (CaP) scaffolds are commonly used in orthopaedics as graft material to heal bone defects. In this study, we have used two CaP scaffolds with different hydroxyapatite (HA) and β-tricalcium phosphate (β-TCP) ratios (MBCP®; 60/40 and MBCP+ ®; 20/80) to investigate their intrinsic capacity to favour human bone marrow stem cells (hBMSCs) osteogenic differentiation capacity. We report that MBCP+ ® showed in in vitro culture model a higher rate of calcium ion release in comparison with MBCP®. In two defined coculture systems, the hBMSC seeded onto MBCP+ ® presented an increased amount of VEGF secretion, resulting in an enhanced endothelial cell proliferation and capillary formation compared with hBMSC seeded onto MBCP®. When both ceramics combined with hBMSC were implanted in a nude mouse model, we observed a faster osteogenic differentiation and enhancement mature bone deposition sustained by the presence of a vast host vasculature within the MBCP+ ® ceramics. Bone formation was observed in samples highly positive to the activation of calcium sensing receptor protein (CaSr) on the surface of seeded hBMSC that also shown higher BMP-2 protein expression. With these data we provide valuable insights in the possible mechanisms of ossification and angiogenesis by hBMSC that we believe to be primed by calcium ions released from CaP scaffolds. Evidences could lead to an optimization of ceramic scaffolds to prime bone repair.
Collapse
Affiliation(s)
- Rui C Pereira
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Roberto Benelli
- Laboratory of Immunology, IRCCS AOU San Martino, Genoa, Italy
| | - Barbara Canciani
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Monica Scaranari
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Guy Daculsi
- INSERM LIOAD U791, Dental Faculty, Nantes University, Nantes, France
| | - Ranieri Cancedda
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Chiara Gentili
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
30
|
Sato T, Anada T, Hamai R, Shiwaku Y, Tsuchiya K, Sakai S, Baba K, Sasaki K, Suzuki O. Culture of hybrid spheroids composed of calcium phosphate materials and mesenchymal stem cells on an oxygen-permeable culture device to predict in vivo bone forming capability. Acta Biomater 2019; 88:477-490. [PMID: 30844570 DOI: 10.1016/j.actbio.2019.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 12/30/2022]
Abstract
Three-dimensional (3-D) cell culture can better mimic physiological conditions in which cells interact with adjacent cells and the extracellular matrix than monolayer culture. We have developed a 3-D cell culture device, the Oxy chip, which can be used to generate and supply oxygen to cell spheroids to prevent hypoxia. Here, we used the Oxy chip to generate hybrid spheroids comprising calcium phosphate (CaP) particles (hydroxyapatite (HA), β-tricalcium phosphate (β-TCP) or octacalcium phosphate (OCP)) and mesenchymal stem cells (MSCs, C3H10T1/2 cells or D1 cells) that can be used to analyze cell differentiation mechanisms. We showed that the 3-D cell-cell and cell-material interactions and oxygenation offered by the Oxy chip promoted osteoblastic differentiation of MSCs. We also used histomorphometric analysis of hematoxylin and eosin staining, quality analyses by μCT and collagen orientation observation with picrosirius red staining in bone regeneration following implantation of three CaPs in a critical-sized defect in mouse calvaria. The in vivo bone formation capacity of the three tested CaP materials was OCP ≥ β-TCP > HA: the newly formed bone by OCP had a structure relatively close to that of the calvaria intact bone. When MSCs were 3-D cultured with the CaP materials using the Oxy chip, the in vitro osteogenic capacity of these materials was highly similar to trends observed in vivo. The in vitro alkaline phosphatase activity of D1 cells had the highest correlation with in vivo bone volume (R = 0.900). Chemical and FTIR spectroscopic analyses confirmed that differentiation of D1 cells could be associated with amorphous calcium phosphate (ACP) precipitation concomitant with OCP hydrolysis. Taken together, hybrid spheroid cultures using the Oxy chip can be used to screen and predict bone forming potential of bone substitute materials. STATEMENT OF SIGNIFICANCE: An oxygen permeable-culture chip (Oxy chip) can be used to induce formation of cell spheroids by mesenchymal stem cells (MSCs). Use of the Oxy chip avoids hypoxia in the spheroid core and enhances MSC osteoblastic differentiation relative to conventional spheroid culture methods. The present study showed that the Oxy chip mimics the in vivo environment associated with bone formation and can be used to generate hybrid spheroids consisting of calcium phosphates and MSCs that are useful for analyzing cell differentiation mechanisms. Bone formation analysis following implantation of calcium phosphate materials in mouse calvaria defects showed positive correlation with the in vitro results. We propose that hybrid spheroids cultured on the Oxy chip can be used to screen and predict the bone forming potential of bone substitute materials.
Collapse
|
31
|
Shih YV, Varghese S. Tissue engineered bone mimetics to study bone disorders ex vivo: Role of bioinspired materials. Biomaterials 2019; 198:107-121. [PMID: 29903640 PMCID: PMC6281816 DOI: 10.1016/j.biomaterials.2018.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/25/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
Recent advances in materials development and tissue engineering has resulted in a substantial number of bioinspired materials that recapitulate cardinal features of bone extracellular matrix (ECM) such as dynamic inorganic and organic environment(s), hierarchical organization, and topographical features. Bone mimicking materials, as defined by its self-explanatory term, are developed based on the current understandings of the natural bone ECM during development, remodeling, and fracture repair. Compared to conventional plastic cultures, biomaterials that resemble some aspects of the native environment could elicit a more natural molecular and cellular response relevant to the bone tissue. Although current bioinspired materials are mainly developed to assist tissue repair or engineer bone tissues, such materials could nevertheless be applied to model various skeletal diseases in vitro. This review summarizes the use of bioinspired materials for bone tissue engineering, and their potential to model diseases of bone development and remodeling ex vivo. We largely focus on biomaterials, designed to re-create different aspects of the chemical and physical cues of native bone ECM. Employing these bone-inspired materials and tissue engineered bone surrogates to study bone diseases has tremendous potential and will provide a closer portrayal of disease progression and maintenance, both at the cellular and tissue level. We also briefly touch upon the application of patient-derived stem cells and introduce emerging technologies such as organ-on-chip in disease modeling. Faithful recapitulation of disease pathologies will not only offer novel insights into diseases, but also lead to enabling technologies for drug discovery and new approaches for cell-based therapies.
Collapse
Affiliation(s)
- Yuru Vernon Shih
- Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA.
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA; Department of Materials Science and Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
32
|
Xie ZY, Wang P, Wu YF, Shen HY. Long non-coding RNA: The functional regulator of mesenchymal stem cells. World J Stem Cells 2019; 11:167-179. [PMID: 30949295 PMCID: PMC6441937 DOI: 10.4252/wjsc.v11.i3.167] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/07/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a subset of multipotent stroma cells residing in various tissues of the body. Apart from supporting the hematopoietic stem cell niche, MSCs possess strong immunoregulatory ability and multiple differentiation potentials. These powerful capacities allow the extensive application of MSCs in clinical practice as an effective treatment for diseases. Therefore, illuminating the functional mechanism of MSCs will help to improve their curative effect and promote their clinical use. Long noncoding RNA (LncRNA) is a novel class of noncoding RNA longer than 200 nt. Recently, multiple studies have demonstrated that LncRNA is widely involved in growth and development through controlling the fate of cells, including MSCs. In this review, we highlight the role of LncRNA in regulating the functions of MSCs and discuss their participation in the pathogenesis of diseases and clinical use in diagnosis and treatment.
Collapse
Affiliation(s)
- Zhong-Yu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yan-Feng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong Province, China
| | - Hui-Yong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
33
|
Griffanti G, Jiang W, Nazhat SN. Bioinspired mineralization of a functionalized injectable dense collagen hydrogel through silk sericin incorporation. Biomater Sci 2019; 7:1064-1077. [DOI: 10.1039/c8bm01060a] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The incorporation of silk sericin into injectable dense collagen hydrogels represents a powerful approach to mimic the biomineralization process, together with the osteogenic stimulation of seeded mesenchymal stem cells, in vitro.
Collapse
Affiliation(s)
- Gabriele Griffanti
- Department of Mining and Materials Engineering
- McGill University
- Montréal
- Canada
| | - Wenge Jiang
- Department of Mining and Materials Engineering
- McGill University
- Montréal
- Canada
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering
- McGill University
- Montréal
- Canada
| |
Collapse
|
34
|
Wang J, Chen X, Guo B, Yang X, Zhou Y, Zhu X, Zhang K, Fan Y, Tu C, Zhang X. A serum protein adsorption profile on BCP ceramics and influence of the elevated adsorption of adhesive proteins on the behaviour of MSCs. J Mater Chem B 2018; 6:7383-7395. [PMID: 32254739 DOI: 10.1039/c8tb02283f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Protein adsorption plays a key role in bone repair and regeneration by affecting cell behavior. In this study, a biphasic calcium phosphate (BCP) ceramic, with excellent osteoinductivity, was chosen to investigate its serum protein adsorption profile using isobaric tags for relative and absolute quantification (iTRAQ) proteomics technology. 281 differentially adsorbed serum proteins and the involved biological processes were confirmed by the combination of Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The differentially adsorbed adhesive proteins in the extracellular matrix (ECM)-receptor interaction pathway were further selected to investigate their roles in the behavior of mesenchymal stem cells (MSCs). Pre-coating and blockage experiments revealed that both adsorbed vitronectin (VN) and laminin (LN) could promote the attachment, proliferation and osteogenic differentiation of MSCs on the BCP ceramic by interacting with different integrin subunits. It is revealed that the up-regulated expressions of integrin α2, αv and β3, β5 could contribute to VN-mediated MSC functions, and the elevated gene expressions of α6 and β1, β4 could be related to the LN-participated process. The above results proved that the preferential protein adsorption on a biomaterial should be vital for modulating MSC functions in the course of material-mediated osteoinductivity.
Collapse
Affiliation(s)
- Jing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Song M, Yu B, Kim S, Hayashi M, Smith C, Sohn S, Kim E, Lim J, Stevenson RG, Kim RH. Clinical and Molecular Perspectives of Reparative Dentin Formation: Lessons Learned from Pulp-Capping Materials and the Emerging Roles of Calcium. Dent Clin North Am 2018; 61:93-110. [PMID: 27912821 DOI: 10.1016/j.cden.2016.08.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The long-term use of calcium hydroxide and the recent increase in the use of hydraulic calcium-silicate cements as direct pulp-capping materials provide important clues in terms of how reparative dentin may be induced to form a "biological seal" to protect the underlying pulp tissues. In this review article, we discuss clinical and molecular perspectives of reparative dentin formation based on evidence learned from the use of these pulp-capping materials. We also discuss the emerging role of calcium as an odontoinductive component in these pulp-capping materials.
Collapse
Affiliation(s)
- Minju Song
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA; Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Bo Yu
- Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Sol Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA; Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Marc Hayashi
- Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Colby Smith
- Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Suhjin Sohn
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Euiseong Kim
- Microscope Center, Department of Conservative Dentistry, Oral Science Research Center, Yonsei University College of Dentistry, 50 Yonsei-Ro, 03772, Seoul, Korea
| | - James Lim
- Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Richard G Stevenson
- Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA; Section of Restorative Dentistry, UCLA School of Dentistry, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Bowers DT, Brown JL. Nanofibers as Bioinstructive Scaffolds Capable of Modulating Differentiation through Mechanosensitive Pathways for Regenerative Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 5:22-29. [PMID: 31179378 DOI: 10.1007/s40883-018-0076-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bioinstructive scaffolds encode information in the physical shape and size of materials to direct cell responses. Electrospinning nanofibers is a process that offers control over scaffold architecture and fiber diameter, while providing extended linear length of fibers. This review summarizes tissue engineering literature that has utilized nanofiber scaffolds to direct stem cell differentiation for various tissues including musculoskeletal, vascular, immunological and nervous system tissues. Nanofibers are also considered for their extracellular matrix mimetic characteristics that can preserve stem cell differentiation capacity. These topics are considered in the context of focal adhesion and integrin signaling. Regenerative engineering will be enhanced by construction of scaffolds encoded with shape information to cause an attached cell to create the intended tissue at that region. Nanofibers are likely to be a bioinstructive scaffold in future regenerative engineering development as we pursue the Grand Challenges of engineering tissues.
Collapse
|
37
|
McMillan A, Nguyen MK, Gonzalez-Fernandez T, Ge P, Yu X, Murphy WL, Kelly DJ, Alsberg E. Dual non-viral gene delivery from microparticles within 3D high-density stem cell constructs for enhanced bone tissue engineering. Biomaterials 2018; 161:240-255. [PMID: 29421560 PMCID: PMC5826638 DOI: 10.1016/j.biomaterials.2018.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/24/2017] [Accepted: 01/02/2018] [Indexed: 01/03/2023]
Abstract
High-density mesenchymal stem cell (MSC) aggregates can be guided to form bone-like tissue via endochondral ossification in vitro when culture media is supplemented with proteins, such as growth factors (GFs), to first guide the formation of a cartilage template, followed by culture with hypertrophic factors. Recent reports have recapitulated these results through the controlled spatiotemporal delivery of chondrogenic transforming growth factor-β1 (TGF-β1) and chondrogenic and osteogenic bone morphogenetic protein-2 (BMP-2) from microparticles embedded within human MSC aggregates to avoid diffusion limitations and the lengthy, costly in vitro culture necessary with repeat exogenous supplementation. However, since GFs have limited stability, localized gene delivery is a promising alternative to the use of proteins. Here, mineral-coated hydroxyapatite microparticles (MCM) capable of localized delivery of Lipofectamine-plasmid DNA (pDNA) nanocomplexes encoding for TGF-β1 (pTGF-β1) and BMP-2 (pBMP-2) were incorporated, alone or in combination, within MSC aggregates from three healthy porcine donors to induce sustained production of these transgenes. Three donor populations were investigated in this work due to the noted MSC donor-to-donor variability in differentiation capacity documented in the literature. Delivery of pBMP-2 within Donor 1 aggregates promoted chondrogenesis at week 2, followed by an enhanced osteogenic phenotype at week 4. Donor 2 and 3 aggregates did not promote robust glycosaminoglycan (GAG) production at week 2, but by week 4, Donor 2 aggregates with pTGF-β1/pBMP-2 and Donor 3 aggregates with both unloaded MCM and pBMP-2 enhanced osteogenesis compared to controls. These results demonstrate the ability to promote osteogenesis in stem cell aggregates through controlled, non-viral gene delivery within the cell masses. These findings also indicate the need to screen donor MSC regenerative potential in response to gene transfer prior to clinical application. Taken together, this work demonstrates a promising gene therapy approach to control stem cell fate in biomimetic 3D condensations for treatment of bone defects.
Collapse
Affiliation(s)
- Alexandra McMillan
- Department of Pathology Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Tomas Gonzalez-Fernandez
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBERG), Trinity College Dublin and Royal College of Surgeons in Dublin, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Dublin, Ireland
| | - Peilin Ge
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Xiaohua Yu
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA; Materials Science Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBERG), Trinity College Dublin and Royal College of Surgeons in Dublin, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Dublin, Ireland
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA; Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA; The National Center for Regenerative Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA; School of Dentistry, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
38
|
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: 148] [Impact Index Per Article: 24.7] [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
|
39
|
Aquino-Martínez R, Angelo AP, Pujol FV. Calcium-containing scaffolds induce bone regeneration by regulating mesenchymal stem cell differentiation and migration. Stem Cell Res Ther 2017; 8:265. [PMID: 29145866 PMCID: PMC5689169 DOI: 10.1186/s13287-017-0713-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/18/2017] [Accepted: 10/26/2017] [Indexed: 11/17/2022] Open
Abstract
Background Osteoinduction and subsequent bone formation rely on efficient mesenchymal stem cell (MSC) recruitment. It is also known that migration is induced by gradients of growth factors and cytokines. Degradation of Ca2+-containing biomaterials mimics the bone remodeling compartment producing a localized calcium-rich osteoinductive microenvironment. The aim of our study was to determine the effect of calcium sulfate (CaSO4) on MSC migration. In addition, to evaluate the influence of CaSO4 on MSC differentiation and the potential molecular mechanisms involved. Methods A circular calvarial bone defect (5 mm diameter) was created in the parietal bone of 35 Balb-C mice. We prepared and implanted a cell-free agarose/gelatin scaffold alone or in combination with different CaSO4 concentrations into the bone defects. After 7 weeks, we determined the new bone regenerated by micro-CT and histological analysis. In vitro, we evaluated the CaSO4 effects on MSC migration by both wound healing and agarose spot assays. Osteoblastic gene expression after BMP-2 and CaSO4 treatment was also evaluated by qPCR. Results CaSO4 increased MSC migration and bone formation in a concentration-dependent manner. Micro-CT analysis showed that the addition of CaSO4 significantly enhanced bone regeneration compared to the scaffold alone. The histological evaluation confirmed an increased number of endogenous cells recruited into the cell-free CaSO4-containing scaffolds. Furthermore, MSC migration in vitro and active AKT levels were attenuated when CaSO4 and BMP-2 were in combination. Addition of LY294002 and Wortmannin abrogated the CaSO4 effects on MSC migration. Conclusions Specific CaSO4 concentrations induce bone regeneration of calvarial defects in part by acting on the host’s undifferentiated MSCs and promoting their migration. Progenitor cell recruitment is followed by a gradual increment in osteoblast gene expression. Moreover, CaSO4 regulates BMP-2-induced MSC migration by differentially activating the PI3K/AKT pathway. Altogether, these results suggest that CaSO4 scaffolds could have potential applications for bone regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0713-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rubén Aquino-Martínez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Alcira P Angelo
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francesc Ventura Pujol
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
40
|
Hayrapetyan A, Bongio M, Leeuwenburgh SCG, Jansen JA, van den Beucken JJJP. Effect of Nano-HA/Collagen Composite Hydrogels on Osteogenic Behavior of Mesenchymal Stromal Cells. Stem Cell Rev Rep 2017; 12:352-64. [PMID: 26803618 PMCID: PMC4879177 DOI: 10.1007/s12015-016-9644-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study aimed to comparatively evaluate the in vitro effect of nanosized hydroxyapatite and collagen (nHA/COL) based composite hydrogels (with different ratios of nHA and COL) on the behavior of human mesenchymal stromal cells (MSCs), isolated from either adipose tissue (AT-MSCs) or bone marrow (BM-MSCs). We hypothesized that (i) nHA/COL composite hydrogels would promote the osteogenic differentiation of MSCs in an nHA concentration dependent manner, and that (ii) AT-MSCs would show higher osteogenic potential compared to BM-MSCs, due to their earlier observed higher proliferation and osteogenic differentiation potential in 2D in vitro cultures [1]. The obtained results indicated that AT-MSCs show indeed high proliferation, differentiation and mineralization capacities in nHA/COL constructs compared to BM-MSCs, but this effect was irrespective of nHA concentration. Based on the results of alkaline phosphatase (ALP) activity and osteocalcin (OCN) protein level, the osteogenic differentiation of BM-MSCs started in the beginning of the culture period and for AT-MSCs at the end of the culture period. At a molecular level, both cell types showed high expression of osteogenic markers (bone morphogenic protein 2 [BMP2], runt-related transcription factor 2 [RUNX2], OCN or COL1) in both an nHA concentration and time dependent manner. In conclusion, AT-MSCs demonstrated higher osteogenic potential in nHA/COL based 3D micro-environments compared to BM-MSCs, in which proliferation and osteogenic differentiation were highly promoted in a time dependent manner, irrespective of nHA amount in the constructs. The fact that AT-MSCs showed high proliferation and mineralization potential is appealing for their application in future pre-clinical research as an alternative cell source for BM-MSCs.
Collapse
Affiliation(s)
- Astghik Hayrapetyan
- Department of Biomaterials, Radboudumc, Ph van Leijdenlaan 25, 6525 ex, Nijmegen, The Netherlands
| | - Matilde Bongio
- Department of Biomaterials, Radboudumc, Ph van Leijdenlaan 25, 6525 ex, Nijmegen, The Netherlands
| | - Sander C G Leeuwenburgh
- Department of Biomaterials, Radboudumc, Ph van Leijdenlaan 25, 6525 ex, Nijmegen, The Netherlands
| | - John A Jansen
- Department of Biomaterials, Radboudumc, Ph van Leijdenlaan 25, 6525 ex, Nijmegen, The Netherlands
| | | |
Collapse
|
41
|
Zhang J, Dalbay MT, Luo X, Vrij E, Barbieri D, Moroni L, de Bruijn JD, van Blitterswijk CA, Chapple JP, Knight MM, Yuan H. Topography of calcium phosphate ceramics regulates primary cilia length and TGF receptor recruitment associated with osteogenesis. Acta Biomater 2017; 57:487-497. [PMID: 28456657 PMCID: PMC5489417 DOI: 10.1016/j.actbio.2017.04.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/09/2017] [Accepted: 04/05/2017] [Indexed: 12/26/2022]
Abstract
The surface topography of synthetic biomaterials is known to play a role in material-driven osteogenesis. Recent studies show that TGFβ signalling also initiates osteogenic differentiation. TGFβ signalling requires the recruitment of TGFβ receptors (TGFβR) to the primary cilia. In this study, we hypothesize that the surface topography of calcium phosphate ceramics regulates stem cell morphology, primary cilia structure and TGFβR recruitment to the cilium associated with osteogenic differentiation. We developed a 2D system using two types of tricalcium phosphate (TCP) ceramic discs with identical chemistry. One sample had a surface topography at micron-scale (TCP-B, with a bigger surface structure dimension) whilst the other had a surface topography at submicron scale (TCP-S, with a smaller surface structure dimension). In the absence of osteogenic differentiation factors, human bone marrow stromal cells (hBMSCs) were more spread on TCP-S than on TCP-B with alterations in actin organization and increased primary cilia prevalence and length. The cilia elongation on TCP-S was similar to that observed on glass in the presence of osteogenic media and was followed by recruitment of transforming growth factor-β RII (p-TGFβ RII) to the cilia axoneme. This was associated with enhanced osteogenic differentiation of hBMSCs on TCP-S, as shown by alkaline phosphatase activity and gene expression for key osteogenic markers in the absence of additional osteogenic growth factors. Similarly, in vivo after a 12-week intramuscular implantation in dogs, TCP-S induced bone formation while TCP-B did not. It is most likely that the surface topography of calcium phosphate ceramics regulates primary cilia length and ciliary recruitment of p-TGFβ RII associated with osteogenesis and bone formation. This bioengineering control of osteogenesis via primary cilia modulation may represent a new type of biomaterial-based ciliotherapy for orthopedic, dental and maxillofacial surgery applications. STATEMENT OF SIGNIFICANCE The surface topography of synthetic biomaterials plays important roles in material-driven osteogenesis. The data presented herein have shown that the surface topography of calcium phosphate ceramics regulates mesenchymal stromal cells (e.g., human bone marrow mesenchymal stromal cells, hBMSCs) with respect to morphology, primary cilia structure and TGFβR recruitment to the cilium associated with osteogenic differentiation in vitro. Together with bone formation in vivo, our results suggested a new type of biomaterial-based ciliotherapy for orthopedic, dental and maxillofacial surgery by the bioengineering control of osteogenesis via primary cilia modulation.
Collapse
|
42
|
Li X, Wang M, Deng Y, Chen X, Xiao Y, Zhang X. Fabrication and Properties of Ca-P Bioceramic Spherical Granules with Interconnected Porous Structure. ACS Biomater Sci Eng 2017; 3:1557-1566. [DOI: 10.1021/acsbiomaterials.7b00232] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Xiangfeng Li
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Menglu Wang
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yanglong Deng
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xuening Chen
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yumei Xiao
- 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
|
43
|
Sun X, Su W, Ma X, Zhang H, Sun Z, Li X. Comparison of the osteogenic capability of rat bone mesenchymal stem cells on collagen, collagen/hydroxyapatite, hydroxyapatite and biphasic calcium phosphate. Regen Biomater 2017; 5:93-103. [PMID: 29644091 PMCID: PMC5888729 DOI: 10.1093/rb/rbx018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/24/2017] [Accepted: 05/28/2017] [Indexed: 12/29/2022] Open
Abstract
Collagen (COL), collagen/hydroxyapatite (COL/HA), HA and biphasic calcium phosphate were prepared as representative bone grafting materials with composition analogous to bone, and their structural characteristics were analyzed. The rat bone mesenchymal stem cells (BMSCs) were further seeded onto four groups of materials, and BMSCs grown in basic medium and standard osteogenic medium were set as controls of a reference model to show the basic and osteogenic behavior of cells without the intervention of materials. Cellular behaviors were characterized, including proliferation, spreading morphology and expression of osteogenesis factors. The rat BMSCs proliferated properly with time on four groups of materials as well on two groups of controls, and typical cuboidal, polygonal and extremely-elongated morphologies of cells were observed. According to the real-time polymerase chain reaction data, a higher osteogenic gene expression level was dependent upon the growing morphology but not the proliferation rate of cells, and the osteogenic differentiation capacity of cells onto four groups of materials varied in specific genes. In general, BMSCs exhibited the highest osteogenic capacity onto COL/HA, but the poorest onto HA. The growing behaviors of cells on materials were further discussed in comparison with the cases of OC and BC of the reference model. The present attempt to comparatively analyze cell experimental data with a reference model is expected to be useful for revealing the difference in the osteogenic capability of MSCs onto materials or even the bioactivity of materials.
Collapse
Affiliation(s)
- Xiaoyu Sun
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Wen Su
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Xiaomin Ma
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Huaiying Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Zhe Sun
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| | - Xudong Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P.R. China
| |
Collapse
|
44
|
Groen N, Yuan H, Hebels DGAJ, Koçer G, Mbuyi F, LaPointe V, Truckenmüller R, van Blitterswijk CA, Habibović P, de Boer J. Linking the Transcriptional Landscape of Bone Induction to Biomaterial Design Parameters. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1603259. [PMID: 27991696 DOI: 10.1002/adma.201603259] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/07/2016] [Indexed: 06/06/2023]
Abstract
New engineering possibilities allow biomaterials to serve as active orchestrators of the molecular and cellular events of tissue regeneration. Here, the molecular control of tissue regeneration for calcium phosphate (CaP)-based materials is established by defining the parameters critical for tissue induction and those are linked to the molecular circuitry controlling cell physiology. The material properties (microporosity, ion composition, protein adsorption) of a set of synthesized osteoinductive and noninductive CaP ceramics are parameterized and these properties are correlated to a transcriptomics profile of osteogenic cells grown on the materials in vitro. Using these data, a genetic network controlling biomaterial-induced bone formation is built. By isolating the complex material properties into single-parameter test conditions, it is verified that a subset of these genes is indeed controlled by surface topography and ions released from the ceramics, respectively. The gene network points to a decisive role for extracellular matrix deposition in osteoinduction by genes such as tenascin C and hyaluronic acid synthase 2, which are controlled by calcium and phosphate ions as well as surface topography. This work provides insight into the biomaterial composition and material engineering aspects of bone void filling and can be used as a strategy to explore the interface between biomaterials and tissue regeneration.
Collapse
Affiliation(s)
- Nathalie Groen
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
| | - Huipin Yuan
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
- Xpand Biotechnology B.V, Professor Bronkhorstlaan 10, 3723, MB, Bilthoven, The Netherlands
| | - Dennie G A J Hebels
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Gülistan Koçer
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
| | - Faustin Mbuyi
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
| | - Vanessa LaPointe
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Clemens A van Blitterswijk
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Jan de Boer
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| |
Collapse
|
45
|
Cheng L, Wang T, Zhu J, Cai P. Osteoinduction of Calcium Phosphate Ceramics in Four Kinds of Animals for 1 Year: Dog, Rabbit, Rat, and Mouse. Transplant Proc 2017; 48:1309-14. [PMID: 27320611 DOI: 10.1016/j.transproceed.2015.09.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/03/2015] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Bone grafts are in great demand. Synthetic materials have been extensively studied as substitutes for autografts. Calcium phosphate ceramics are promising synthetic bone replacement materials. Because they share chemical similarities with human bone mineral, they show excellent biocompatibility and osteoinductivity. OBJECTIVE Calcium phosphate ceramics have been used to fill bone defects in preclinical study in a variety of animals. This study aimed to investigate the osteogenesis ability of calcium phosphate ceramics in 4 kinds of animals. METHODS Φ3 × 5 mm hydroxyapatite/β-tricalcium phosphate (HA/β-TCP) cylinders were implanted into the dorsal muscle of rats and mice, whereas Φ5 × 10 mm cylinders were implanted into the dorsal muscle of dogs and rabbits. One year after implantation, the ceramics were harvested to perform hematoxylin and eosin (HE) staining and Masson-trichrome staining. The new bone tissues were observed and the area percentage of new bone was compared in the 4 kinds of animals. RESULTS A large number of new bone and bone marrow tissues were observed in dogs, rabbits, and mice, but not in rats; and the area percentage of new bone in mice was significantly higher than that in dogs and rabbits (P < .05). Calcium phosphate ceramics have good biocompability and biological safety, and the degree of ease of osteogenesis was as follows: mouse > dog > rabbit > rat. CONCLUSION To achieve better effects for bone transplantation, mouse should be chosen as the preferred experimental model based on these advantages: economic, convenience, and osteogenesis ability.
Collapse
Affiliation(s)
- L Cheng
- Medical School (Nursing School), Chengdu University, Chengdu, China
| | - T Wang
- Medical School (Nursing School), Chengdu University, Chengdu, China
| | - J Zhu
- Medical School (Nursing School), Chengdu University, Chengdu, China
| | - P Cai
- Medical School (Nursing School), Chengdu University, Chengdu, China.
| |
Collapse
|
46
|
Clarke SA, Martin J, Nelson J, Hornez JC, Bohner M, Dunne N, Buchanan F. Surrogate Outcome Measures of In Vitro Osteoclast Resorption of β Tricalcium Phosphate. Adv Healthc Mater 2017; 6. [PMID: 27930865 DOI: 10.1002/adhm.201600947] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/01/2016] [Indexed: 01/12/2023]
Abstract
Introduction of porosity to calcium phosphate scaffolds for bone repair has created a new challenge when measuring bioresorption in vitro, rendering traditional outcome measures redundant. The aim of this study is to identify a surrogate endpoint for use with 3D scaffolds. Murine RAW 264.7 cells are cultured on dense discs of β-tricalcium phosphate in conditions to stimulate osteoclast (OC) formation. Multinucleated OCs are visible from day 6 with increases at days 8 and 10. Resorption pits are first observed at day 6 with much larger pits visible at days 8, 10, and 12. The concentration of calcium ions in the presence of cells is significantly higher than cell-free cultures at days 3 and 9. Using linear regression analysis, Ca ion release could account for 35.9% of any subsequent change in resorption area. The results suggest that Ca ion release is suitable to measure resorption of a beta-tricalcium phosphate ceramic substrate in vitro. This model could replace the more accepted resorption pit assay in circumstances where quantification of pits is not possible, e.g., when characterizing 3D tissue engineered bone scaffolds.
Collapse
Affiliation(s)
- Susan A. Clarke
- School of Nursing and Midwifery; Medical Biology Centre; 97, Lisburn Road Belfast BT9 7BL UK
| | - Joanne Martin
- School of Mechanical and Aerospace Engineering; Queen's University Belfast; Ashby Building, Stranmillis Rd Belfast BT9 5AH UK
| | - John Nelson
- School of Biological Sciences; Queens University Belfast; MBC, 97 Lisburn Rd Belfast BT9 7BL UK
| | | | - Marc Bohner
- Skeletal Substitutes Group; RMS Foundation; Bischmattstr. 12 CH-2544 Bettlach Switzerland
| | - Nicholas Dunne
- School of Mechanical and Aerospace Engineering; Queen's University Belfast; Ashby Building, Stranmillis Rd Belfast BT9 5AH UK
| | - Fraser Buchanan
- School of Mechanical and Aerospace Engineering; Queen's University Belfast; Ashby Building, Stranmillis Rd Belfast BT9 5AH UK
| |
Collapse
|
47
|
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: 6.1] [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]
|
48
|
Pujari-Palmer M, Pujari-Palmer S, Lu X, Lind T, Melhus H, Engstrand T, Karlsson-Ott M, Engqvist H. Pyrophosphate Stimulates Differentiation, Matrix Gene Expression and Alkaline Phosphatase Activity in Osteoblasts. PLoS One 2016; 11:e0163530. [PMID: 27701417 PMCID: PMC5049792 DOI: 10.1371/journal.pone.0163530] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 09/09/2016] [Indexed: 12/29/2022] Open
Abstract
Pyrophosphate is a potent mitogen, capable of stimulating proliferation in multiple cell types, and a critical participant in bone mineralization. Pyrophosphate can also affect the resorption rate and bioactivity of orthopedic ceramics. The present study investigated whether calcium pyrophosphate affected proliferation, differentiation and gene expression in early (MC3T3 pre-osteoblast) and late stage (SAOS-2 osteosarcoma) osteoblasts. Pyrophosphate stimulated peak alkaline phosphatase activity by 50% and 150% at 100μM and 0.1μM in MC3T3, and by 40% in SAOS-2. The expression of differentiation markers collagen 1 (COL1), alkaline phosphatase (ALP), osteopontin (OPN), and osteocalcin (OCN) were increased by an average of 1.5, 2, 2 and 3 fold, by high concentrations of sodium pyrophosphate (100μM) after 7 days of exposure in MC3T3. COX-2 and ANK expression did not differ significantly from controls in either treatment group. Though both high and low concentrations of pyrophosphate stimulate ALP activity, only high concentrations (100μM) stimulated osteogenic gene expression. Pyrophosphate did not affect proliferation in either cell type. The results of this study confirm that chronic exposure to pyrophosphate exerts a physiological effect upon osteoblast differentiation and ALP activity, specifically by stimulating osteoblast differentiation markers and extracellular matrix gene expression.
Collapse
Affiliation(s)
- Michael Pujari-Palmer
- Division of Applied Material Science, Department of Engineering Sciences, Uppsala University, Uppsala, Sweden
| | - Shiuli Pujari-Palmer
- Division of Applied Material Science, Department of Engineering Sciences, Uppsala University, Uppsala, Sweden
| | - Xi Lu
- Division of Applied Material Science, Department of Engineering Sciences, Uppsala University, Uppsala, Sweden
| | - Thomas Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Håkan Melhus
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Thomas Engstrand
- Stockholm Craniofacial Centre, Department of Reconstructive Plastic Surgery, Karolinska University Hospital, Stockholm, Sweden
- Department of Materials Chemistry, Polymer section, Uppsala University, Uppsala, Sweden
| | - Marjam Karlsson-Ott
- Division of Applied Material Science, Department of Engineering Sciences, Uppsala University, Uppsala, Sweden
- * E-mail:
| | - Hakan Engqvist
- Division of Applied Material Science, Department of Engineering Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
49
|
Sun L, Danoux CB, Wang Q, Pereira D, Barata D, Zhang J, LaPointe V, Truckenmüller R, Bao C, Xu X, Habibovic P. Independent effects of the chemical and microstructural surface properties of polymer/ceramic composites on proliferation and osteogenic differentiation of human MSCs. Acta Biomater 2016; 42:364-377. [PMID: 27318269 DOI: 10.1016/j.actbio.2016.06.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/06/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Within the general aim of finding affordable and sustainable regenerative solutions for damaged and diseased tissues and organs, significant efforts have been invested in developing synthetic alternatives to natural bone grafts, such as autografts. Calcium phosphate (CaP) ceramics are among widely used synthetic bone graft substitutes, but their mechanical properties and bone regenerative capacity are still outperformed by their natural counterparts. In order to improve the existing synthetic bone graft substitutes, it is imperative to understand the effects of their individual properties on a biological response, and to find a way to combine the desired properties into new, improved functional biomaterials. To this end, we studied the independent effects of the chemical composition and surface microstructure of a poly(lactic acid)/hydroxyapatite (PLA/HA) composite material on the proliferation and osteogenic differentiation of clinically relevant bone marrow-derived human mesenchymal stromal cells (hMSCs). While the molecular weight of the polymer and presence/absence of the ceramic phase were used as the chemical variables, a soft embossing technique was used to pattern the surfaces of all materials with either pits or pillars with identical microscale dimensions. The results indicated that, while cell morphology was affected by both the presence and availability of HA and by the surface microstructure, the effect of the latter parameter on cell proliferation was negligible. The osteogenic differentiation of hMSCs, and in particular the expression of bone morphogenetic protein 2 (BMP-2) and osteopontin (OP) were significantly enhanced when cells were cultured on the composite based on low-molecular-weight PLA, as compared to the high-molecular-weight PLA-based composite and the two pure polymers. The OP expression on the low-molecular-weight PLA-based composite was further enhanced when the surface was patterned with pits. Taken together, within this experimental set up, the individual effect of the chemistry, and in particular of the presence of CaP, was more pronounced than the individual effect of the surface microstructure, although their combined effects were, in some cases, synergistic. The approach presented here opens new routes to study the interactions of biomaterials with the biological environment in greater depths, which can serve as a starting point for developing biomaterials with improved bioactivity. STATEMENT OF SIGNIFICANCE The aim of the this study was to obtain insight into independent effects of the chemical composition and surface microstructure of a poly(lactic acid)/hydroxyapatite (PLA/HA) composite material on the morphology, proliferation and osteogenic differentiation of clinically relevant bone marrow-derived human mesenchymal stromal cells (hMSCs). While the need for synthetic alternatives for natural bone in bone regenerative strategies is rapidly increasing, the clinical performance of synthetic biomaterials needs to be further improved. To do this successfully, we believe that a better understanding of the relationship between a property of a material and a biological response is imperative. This study is a step forward in this direction, and we are therefore convinced that it will be of interest to the readers of Acta Biomaterialia.
Collapse
|
50
|
Danoux C, Pereira D, Döbelin N, Stähli C, Barralet J, van Blitterswijk C, Habibovic P. The Effects of Crystal Phase and Particle Morphology of Calcium Phosphates on Proliferation and Differentiation of Human Mesenchymal Stromal Cells. Adv Healthc Mater 2016; 5:1775-85. [PMID: 27232450 DOI: 10.1002/adhm.201600184] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/18/2016] [Indexed: 12/12/2022]
Abstract
Calcium phosphate (CaP) ceramics are extensively used for bone regeneration; however, their clinical performance is still considered inferior to that of patient's own bone. To improve the performance of CaP bone graft substitutes, it is important to understand the effects of their individual properties on a biological response. The aim of this study is to investigate the effects of the crystal phase and particle morphology on the behavior of human mesenchymal stromal cells (hMSCs). To study the effect of the crystal phase, brushite, monetite, and octacalcium phosphate (OCP) are produced by controlling the precipitation conditions. Brushite and monetite are produced as plate-shaped and as needle-shaped particles, to further investigate the effect of particle morphology. Proliferation of hMSCs is inhibited on OCP as compared to brushite and monetite in either morphology. Brushite needles consistently show the lowest expression of most osteogenic markers, whereas the expression on OCP is in general high. There is a trend toward a higher expression of the osteogenic markers on plate-shaped than on needle-shaped particles for both brushite and monetite. Within the limits of CaP precipitation, these data indicate the effect of both crystal phase and particle morphology of CaPs on the behavior of hMSCs.
Collapse
Affiliation(s)
- Charlène Danoux
- Department of Tissue Regeneration University of Twente P.O. Box 217 7500 AE Enschede The Netherlands
| | - Daniel Pereira
- MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University P.O. Box 616 6200 MD Maastricht The Netherlands
| | - Nicola Döbelin
- RMS Foundation P.O. Box 203 2544 Bettlach Switzerland
- Institute of Geological Sciences University of Bern Baltzerstrasse 1‐3 3012 Bern Switzerland
| | | | - Jake Barralet
- Faculty of Dentistry McGill University 3640 University Street Montreal QC H3 A 2B2 Canada
| | - Clemens van Blitterswijk
- Department of Tissue Regeneration University of Twente P.O. Box 217 7500 AE Enschede The Netherlands
- MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University P.O. Box 616 6200 MD Maastricht The Netherlands
| | - Pamela Habibovic
- Department of Tissue Regeneration University of Twente P.O. Box 217 7500 AE Enschede The Netherlands
- MERLN Institute for Technology‐Inspired Regenerative Medicine Maastricht University P.O. Box 616 6200 MD Maastricht The Netherlands
| |
Collapse
|