1
|
Klar RM, Cox JC, Houchen CJ, Raja N, Bouloussa H, Lohfeld S. The induction of bone formation by 3D-printed PLGA microsphere scaffolds in a calvarial orthotopic mouse model: a pilot study. Front Bioeng Biotechnol 2024; 12:1425469. [PMID: 39524123 PMCID: PMC11544432 DOI: 10.3389/fbioe.2024.1425469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Polymeric biodegradable microspheres are readily utilized to support targeted drug delivery for various diseases clinically. 3D printed tissue engineering scaffolds from polymer filaments with embedded microspheres or nanoparticles, as well as bulk microsphere scaffolds, have been investigated for regenerative medicine and tissue engineering. However, 3D printed scaffolds consisting only of a homogenous microsphere size with an optimized architecture that includes a unique micro- and macroporosity, have been challenging to produce and hence, have not been assessed in the literature yet. Utilizing our recently established 3D-MultiCompositional Microsphere-Adaptive Printing (3D-McMap) method, the present study evaluated the effectiveness of 3D-printed poly (lactic-co-glycolic acid) (PLGA) microsphere scaffolds, consisting of microsphere sizes 50, 100, or 200 μm, on the induction of bone formation when implanted in the calvarial murine regeneration model. Our results showed that PLGA microsphere scaffolds possess unique properties that support bone regeneration by supporting osteoconduction and stimulating, in our opinion, true spontaneous osteoinduction. The study demonstrated that PLGA microsphere-based scaffolds support bone growth in the absence of additional growth factors and promote osteogenesis primarily via their unique geometric configuration. The larger the microspheres were, the greater de novo bone formation was. This proves that bone tissue engineering scaffolds 3D printed from microspheres, enabled by the 3D-McMap method, are superior over bulk material printed scaffolds, as they possess the unique capability of spontaneous induction of new bone formation. With the addition of encapsulated modulatory bone-forming biomolecules they can substantially improve the spatiotemporal control of tissue morphogenesis, potentially leading to new innovative clinical tissue repair therapies that regenerate bone in large defects correctly and fully.
Collapse
Affiliation(s)
- Roland M. Klar
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, School of Dentistry, Kansas City, MO, United States
| | - James C. Cox
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, School of Dentistry, Kansas City, MO, United States
| | - Claire J. Houchen
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, School of Dentistry, Kansas City, MO, United States
| | - Naren Raja
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, School of Dentistry, Kansas City, MO, United States
| | - Houssam Bouloussa
- Department of Orthopedic Surgery, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States
| | - Stefan Lohfeld
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, School of Dentistry, Kansas City, MO, United States
| |
Collapse
|
2
|
Ripamonti U, Duarte R. Mechanistic insights into the spontaneous induction of bone formation. BIOMATERIALS ADVANCES 2024; 158:213795. [PMID: 38335762 DOI: 10.1016/j.bioadv.2024.213795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
The grand discovery of morphogens, or "form-generating substances", revealed that tissue morphogenesis is initiated by soluble molecular signals or morphogens primarily belonging to the transforming growth factor-β (TGF-β) supergene family. The regenerative potential of bone rests on its extracellular matrix, which is the repository of several morphogens that tightly control cellular differentiating pathways, cellular matrix deposition and remodeling. Alluringly, the matrix also contains specific factors transferred from the heterotopic implanted bone matrices initiating "Tissue Induction", as provocatively described in Nature in 1945. Later, it was found that selected genes and gene products of the TGF-β supergene family singly, synchronously, and synergistically mastermind the induction of bone formation. This review describes the phenomenon of the spontaneous and/or intrinsic osteoinductivity of calcium phosphate-based biomaterials and titanium' constructs without the applications of soluble osteogenetic molecular signals. The review shows the spontaneous induction of bone formation initiated by Ca++ activating stem cell differentiation and up-regulation of bone morphogenetic proteins genes. Expressed gene products are embedded into the concavities of the calcium phosphate-based substrata, initiating bone formation as a secondary response. Pure titanium's substrata do not initiate the spontaneous induction of bone formation. The induction of bone is solely dependent on acid, alkali and heat treatments to form apatite layers on the treated titanium surfaces. The induction of bone formation is achieved exclusively by apatite-based biomaterial surfaces. The hydroxyapatite, in its various forms and geometric configurations, finely tunes the induction of bone formation in heterotopic sites. Cellular differentiation by fine-tuning of the cellular molecular machinery is initiated by specific geometric modularity of the hydroxyapatite substrata that push cellular buttons that start the ripple-like cascade of "Tissue Induction", generating newly formed ossicles with bone marrow in heterotopic extraskeletal sites. The highlighted mechanistic insights into the spontaneous induction of bone formation are a research platform invocating selected molecular elements to construct the induction of bone formation.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Raquel Duarte
- Bone Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Internal Medicine Research Laboratory, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
3
|
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
|
4
|
Peng H, Ling T, Zhang Y, Xie T, Pei X, Zhou K, Chen A, Chen J, Zhu X, Zhang X, Zhou Z. Nanowhiskers Orchestrate Bone Formation and Bone Defect Repair by Modulating Immune Cell Behavior. ACS APPLIED MATERIALS & INTERFACES 2023; 15:9120-9134. [PMID: 36755394 DOI: 10.1021/acsami.2c21865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Immunomodulatory biomaterials have emerged as promising treatment agents for bone defects. However, it is unclear how such biomaterials control immune cell behaviors to facilitate large-segment bone defect repair. Herein, we fabricated biphasic calcium phosphate ceramics with nanowhisker structures to explore the immunoregulation features and influence on large-segment bone defect repair. We found that the nanowhisker structures markedly facilitated large-segment bone defect repair by promoting bone regeneration and scaffold resorption. Our in vitro experiment and transcriptomic analysis showed that mechanical stress derived from nanowhisker structures may activate the transcription of Egr-1 to induce early switch of macrophage phenotype to M2, which could not only facilitate osteogenic differentiation of BMSCs but also enhance the expression of osteoclast differentiation-regulating genes of M2 macrophage. In vivo study showed that the nanowhisker structures relieved local inflammatory responses by inducing early switch of macrophage phenotype from M1 to M2, which resulted in accelerated osteoclastogenesis for biomaterial resorption and osteogenesis for ectopic bone formation. Hence, we presume that nanowhisker structures may orchestrate bone formation and material resorption coupling to facilitate large-segment bone defect repair by controlling the switch of macrophage phenotype. This study provides new insight into the designing of immunomodulatory tissue engineering biomaterials for treating large-segment bone defects.
Collapse
Affiliation(s)
- Haitao Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Tingxian Ling
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Zhang
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Tianhang Xie
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xuan Pei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Kai Zhou
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Anjing Chen
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiali Chen
- West China School of Nursing, Sichuan University/Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Zongke Zhou
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
5
|
Nie Z, Hu Z, Guo X, Xiao Y, Liu X, de Bruijn JD, Bao C, Yuan H. Genesis of osteoclasts on calcium phosphate ceramics and their role in material-induced bone formation. Acta Biomater 2023; 157:625-638. [PMID: 36371003 DOI: 10.1016/j.actbio.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022]
Abstract
Innate immune responses play important roles in material-induced bone formation and such roles were further explored in the current study with an emphasis on M2 macrophages and osteoclastogenesis. With the presence of M-CSF and RANKL, M0 macrophages from FVB mouse bone marrow-derived monocytes (BMMs) fused to osteoclasts with both M2 marker and osteoclast marker at day 5, and such osteoclast formation at day 5 was enhanced when the cells were treated with IL-4 at day 3. With IL-4 treatment alone for 24 h, M0 polarized into M2 macrophages. Conditioned medium of M2 macrophages enhanced osteogenic differentiation of MC3T3-E1 (pre-osteoblasts) while osteoclast conditioned medium enhanced osteogenic differentiation of CRL-12424 (osteogenic precursors). TCPs (a typical osteoinductive material) supported M2 macrophage polarization at day 4 and osteoclast formation at day 5, while TCPb (a typical non-osteoinductive material) was less effective. Moreover, osteoclasts formed on TCPs produced osteogenic factors including S1P, Wnt10B and BMP-6, resulting osteogenic differentiation of CRL-12424 cells. Similar to in vitro testing, TCPs favored M2 macrophage polarization followed by the formation of osteoclasts in vivo, as compared to TCPb. The overall data provided evidence of a coupling between M2 macrophages, osteoclasts and material-induced bone formation: osteoclasts formed from M2 macrophages secrete osteogenic cytokines to induce osteogenic differentiation of osteogenic precursor cells to finally form bone. The current findings outlined a biological mechanism of material-induced bone formation and further rationalized the use of osteoinductive materials for bone regeneration. STATEMENT OF SIGNIFICANCE: This paper provides evidence for finding out the relationship between M2 macrophages, osteoclasts and osteogenesis in material-induced bone formation. It suggested that osteoinductive materials enhanced macrophage polarization to M2 macrophages which fuses to osteoclasts, osteoclasts subsequently secret osteogenic cytokines to differentiate finally osteogenic precursors to form bone in osteoinductive materials. The data supports scientifically the superiority of osteoinductive materials for bone regeneration in clinics.
Collapse
Affiliation(s)
- Zhangling Nie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Zhiqiao Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Xiaodong Guo
- National Center of Stomatology & National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Xian Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Joost D de Bruijn
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, the Netherlands; School of Engineering and Materials Science, Queen Mary University of London, UK
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China.
| | - Huipin Yuan
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, the Netherlands; Huipin Yuan's Lab, Sichuan, China.
| |
Collapse
|
6
|
Sasaki K, Takeshita N, Fukunaga T, Seiryu M, Sakamoto M, Oyanagi T, Maeda T, Takano-Yamamoto T. Vibration accelerates orthodontic tooth movement by inducing osteoclastogenesis via transforming growth factor-β signalling in osteocytes. Eur J Orthod 2022; 44:698-704. [DOI: 10.1093/ejo/cjac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Abstract
Background
We previously found the conditions of supplementary vibration that accelerated tooth movement and induced bone resorption in an experimental rat tooth movement model. However, the molecular biological mechanisms underlying supplementary vibration-induced orthodontic tooth movement are not fully understood. Transforming growth factor (TGF)-β upregulates osteoclastogenesis via induction of the receptor activator of nuclear factor kappa B ligand expression, thus TGF-β is considered an essential cytokine to induce bone resorption.
Objectives
The aim of this study is to examine the role of TGF-β during the acceleration of orthodontic tooth movement by supplementary vibration.
Materials and methods
In experimental tooth movement, 15 g of orthodontic force was loaded onto the maxillary right first molar for 28 days. Supplementary vibration (3 g, 70 Hz) was applied to the maxillary first molar for 3 min on days 0, 7, 14, and 21. TGF-β receptor inhibitor SB431542 was injected into the submucosal palatal and buccal areas of the maxillary first molars once every other day. The co-culture of RAW264.7 cells and MLO-Y4 cells was used as an in vitro osteoclastogenesis model.
Results
SB431542 suppressed the acceleration of tooth movement and the increase in the number of osteoclasts by supplementary vibration in our experimental rat tooth movement model. Immunohistochemical analysis showed supplementary vibration increased the number of TGF-β1-positive osteocytes in the alveolar bone on the compression side during the experimental tooth movement. Moreover, vibration-upregulated TGF-β1 in MLO-Y4 cells induced osteoclastogenesis.
Conclusions
Orthodontic tooth movement was accelerated by supplementary vibration through the promotion of the production of TGF-β1 in osteocytes and subsequent osteoclastogenesis.
Collapse
Affiliation(s)
- Kiyo Sasaki
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
| | - Nobuo Takeshita
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
- Section of Orthodontics and Dentofacial Orthopedics, Faculty of Dental Science, Kyushu University , Fukuoka, Fukuoka , Japan
| | - Tomohiro Fukunaga
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
| | - Masahiro Seiryu
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
| | - Mayuri Sakamoto
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
| | - Toshihito Oyanagi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
| | - Toshihiro Maeda
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University , Sendai, Miyagi , Japan
- Department of Biomaterials and Bioengineering, Faculty of Dental Medicine, Hokkaido University , Sapporo, Hokkaido , Japan
| |
Collapse
|
7
|
Chen F, Tian L, Pu X, Zeng Q, Xiao Y, Chen X, Zhang X. Enhanced ectopic bone formation by strontium-substituted calcium phosphate ceramics through regulation of osteoclastogenesis and osteoblastogenesis. Biomater Sci 2022; 10:5925-5937. [PMID: 36043373 DOI: 10.1039/d2bm00348a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To explore how strontium influences osteoclastogenesis and osteoblastogenesis during material-induced ectopic bone formation, porous strontium-substituted biphasic calcium phosphate (Sr-BCP) and BCP ceramics with equivalent pore structures and comparable grain size and porosity were prepared. In vitro results showed that compared with BCP, Sr-BCP inhibited the osteoclastic differentiation of osteoclast precursors by delaying cell fusion, down-regulating the expression of osteoclast marker genes, and reducing the activity of osteoclast specific proteins, possibly due to the activated ERK signaling pathway but the suppressed p38, JNK and AKT signaling pathways. Meanwhile, Sr-BCP promoted the osteogenic differentiation of mesenchymal stem cells (MSCs) by up-regulating the osteogenic gene expression. Sr-BCP also mediated the expression of important osteoblast-osteoclast coupling factors, as evidenced by the increased Opg/Rankl ratio in mMSCs, and the reduced Rank expression and enhanced EphrinB2 expression in osteoclast precursors. Similar results were observed in an in vivo study based on a murine intramuscular implantation model. The sign of ectopic bone formation was only seen in Sr-BCP at 8 weeks. Compared to BCP, Sr-BCP obviously hindered the formation of TRAP- and CTSK-positive multinucleated osteoclast-like cells during the early implantation time up to 6 weeks, which is consistent with the in vivo PCR results. This suggested that Sr-BCP could clearly accelerate the ectopic bone formation by promoting osteogenesis but suppressing osteoclastogenesis, which might be closely related to the expression of osteoblast-osteoclast coupling factors regulated by Sr2+. These findings may help in the design and fabrication of smart bone substitutes with the desired potential for bone regeneration through modulating both osteoclastic resorption and osteoblastic synthesis.
Collapse
Affiliation(s)
- Fuying Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Qin Zeng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Xuening Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
8
|
Wang L, Wei X, Duan C, Yang J, Xiao S, Liu H, Sun J. Bone marrow mesenchymal stem cell sheets with high expression of hBD3 and CTGF promote periodontal regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112657. [PMID: 35034825 DOI: 10.1016/j.msec.2022.112657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/12/2021] [Accepted: 01/06/2022] [Indexed: 12/31/2022]
Abstract
The multi-bacterial environment of the oral cavity makes it hard for periodontal regeneration. As a class of antimicrobial peptide, beta defensin has been found to show broad-spectrum antibacterial ability. In addition, connective tissue growth factor (CTGF) is demonstrated to play a great role in multi-physiological events such as angiogenesis, wound healing and, more importantly, fibrogenesis. In this study, human β defensin 3 (hBD3) and CTGF were co-transfected into bone marrow derived mesenchymal stem cells (BMSCs) for preparing cell sheets. The transfection efficiency was detected through fluorescence of eGFP and western blot assay. Our results showed that the hBD3 and CTGF proteins were highly and stably expressed in the BMSCs after transfection. The results of RT-PCR and induced differentiation indicated that hBD3 promoted osteogenic differentiation of BMSCs, while CTGF significantly increased fibrogenic differentiation even in the presence of hBD3. The BMSCs acquired stronger capacity in terms of promoting M2 polarization of RAW 264.7 macrophages fulfilled by the transfection and secretion of hBD3 and CTGF. To further evaluate the periodontal remodeling performance of cell sheets, a coralline hydroxyapatite (CHA)-chitosan based hydrogel-human tooth system was designed to simulate the natural periodontal environment. The results showed that dense extracellular matrix, oriented fiber arrangement, and abundant collagen deposition appeared in the area of BMSCs sheets after subcutaneous transplantation. Altogether, our data showed that the lentivirus transfected BMSCs sheets had a promising application prospect for periodontal repair.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, PR China
| | - Xinbo Wei
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, PR China
| | - Cuimi Duan
- Tissue Engineering Research Center, Beijing Institute of Basic Medical Sciences, PR China
| | - Jinjin Yang
- Department of Stomatology, The Fifth Medical Center, Chinese PLA General Hospital, Xisihuan Middle Road 100, Fengtai District, Beijing 100036, PR China
| | - Shengzhao Xiao
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, PR China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, PR China.
| | - Jie Sun
- Department of Stomatology, The Fifth Medical Center, Chinese PLA General Hospital, Xisihuan Middle Road 100, Fengtai District, Beijing 100036, PR China.
| |
Collapse
|
9
|
Wu Z, Zhong Z, He W, Wu Y, Cai Y, Yang H, Hong Y. Construction of a drug-containing microenvironment for in situ bone regeneration. MATERIALS ADVANCES 2022. [DOI: 10.1039/d2ma00057a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Bioactive glass-coated hierarchical porous tricalcium phosphate ceramics were constructed as both bone scaffolds and drug delivery devices to treat S. aureus-infected bone defects.
Collapse
Affiliation(s)
- Zhen Wu
- National Engineering Research Centre for Biomaterials; Department of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Zhou Zhong
- Department of Orthopaedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Wenchao He
- National Engineering Research Centre for Biomaterials; Department of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Yanmei Wu
- National Engineering Research Centre for Biomaterials; Department of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Yuyan Cai
- National Engineering Research Centre for Biomaterials; Department of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Huilin Yang
- Department of Orthopaedics, The first Hospital Affiliated to Suzhou University, Suzhou, 215006, P. R. China
| | - Youliang Hong
- National Engineering Research Centre for Biomaterials; Department of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
10
|
Brennan MÁ, Monahan DS, Brulin B, Gallinetti S, Humbert P, Tringides C, Canal C, Ginebra MP, Layrolle P. Biomimetic versus sintered macroporous calcium phosphate scaffolds enhanced bone regeneration and human mesenchymal stromal cell engraftment in calvarial defects. Acta Biomater 2021; 135:689-704. [PMID: 34520883 DOI: 10.1016/j.actbio.2021.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023]
Abstract
In contrast to sintered calcium phosphates (CaPs) commonly employed as scaffolds to deliver mesenchymal stromal cells (MSCs) targeting bone repair, low temperature setting conditions of calcium deficient hydroxyapatite (CDHA) yield biomimetic topology with high specific surface area. In this study, the healing capacity of CDHA administering MSCs to bone defects is evaluated for the first time and compared with sintered beta-tricalcium phosphate (β-TCP) constructs sharing the same interconnected macroporosity. Xeno-free expanded human bone marrow MSCs attached to the surface of the hydrophobic β-TCP constructs, while infiltrating the pores of the hydrophilic CDHA. Implantation of MSCs on CaPs for 8 weeks in calvaria defects of nude mice exhibited complete healing, with bone formation aligned along the periphery of β-TCP, and conversely distributed within the pores of CDHA. Human monocyte-osteoclast differentiation was inhibited in vitro by direct culture on CDHA compared to β-TCP biomaterials and indirectly by administration of MSC-conditioned media generated on CDHA, while MSCs increased osteoclastogenesis in both CaPs in vivo. MSC engraftment was significantly higher in CDHA constructs, and also correlated positively with bone in-growth in scaffolds. These findings demonstrate that biomimetic CDHA are favorable carriers for MSC therapies and should be explored further towards clinical bone regeneration strategies. STATEMENT OF SIGNIFICANCE: Delivery of mesenchymal stromal cells (MSCs) on calcium phosphate (CaP) biomaterials enhances reconstruction of bone defects. Traditional CaPs are produced at high temperature, but calcium deficient hydroxyapatite (CDHA) prepared at room temperature yields a surface structure more similar to native bone mineral. The objective of this study was to compare the capacity of biomimetic CDHA scaffolds with sintered β-TCP scaffolds for bone repair mediated by MSCs for the first time. In vitro, greater cell infiltration occurred in CDHA scaffolds and following 8 weeks in vivo, MSC engraftment was higher in CDHA compared to β-TCP, as was bone in-growth. These findings demonstrate the impact of material features such as surface structure, and highlight that CDHA should be explored towards clinical bone regeneration strategies.
Collapse
Affiliation(s)
- Meadhbh Á Brennan
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Biomedical Engineering, School of Engineering; and Regenerative Medicine Institute (REMEDI), School of Medicine, National University of Ireland (NUIG), Galway, Ireland
| | - David S Monahan
- Biomedical Engineering, School of Engineering; and Regenerative Medicine Institute (REMEDI), School of Medicine, National University of Ireland (NUIG), Galway, Ireland
| | - Bénédicte Brulin
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France; INSERM, UMR 1214, ToNIC, CHU Purpan, Université Paul Sabatier, Toulouse 31024, France
| | - Sara Gallinetti
- Biomaterials, Biomechanics and Tissue Engineering Group, Dpt. Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 10-14, Barcelona 08019, Spain; Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Paul Humbert
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France
| | - Christina Tringides
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Harvard Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Cristina Canal
- Biomaterials, Biomechanics and Tissue Engineering Group, Dpt. Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 10-14, Barcelona 08019, Spain; Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Maria Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Dpt. Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 10-14, Barcelona 08019, Spain; Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain; Institute of Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Baldiri i Reixach 10-12, Barcelona 08028, Spain
| | - Pierre Layrolle
- INSERM, UMR 1238, PHY-OS, Faculty of Medicine, University of Nantes, 1 Rue Gaston Veil, Nantes 44035, France; INSERM, UMR 1214, ToNIC, CHU Purpan, Université Paul Sabatier, Toulouse 31024, France.
| |
Collapse
|
11
|
Osteogenic Competence and Potency of the Bone Induction Principle: Inductive Substrates That Initiate “Bone: Formation by Autoinduction”. J Craniofac Surg 2021; 33:971-984. [DOI: 10.1097/scs.0000000000008299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
12
|
Stokovic N, Ivanjko N, Erjavec I, Breski A, Peric M, Vukicevic S. Zoledronate Bound to Ceramics Increases Ectopic Bone Volume Induced by rhBMP6 Delivered in Autologous Blood Coagulum in Rats. Biomedicines 2021; 9:biomedicines9101487. [PMID: 34680604 PMCID: PMC8533060 DOI: 10.3390/biomedicines9101487] [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: 09/22/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Autologous bone graft substitute (ABGS) containing rhBMP6 in autologous blood coagulum (ABC) with synthetic ceramics is a novel therapeutic solution for bone repair. The aim of this study was to investigate whether the application of Zoledronate (ZOL) with ABGS might enhance the properties of newly formed bone. The effect of ZOL on bone induction was tested in a rat subcutaneous implant model. ZOL bound to synthetic ceramics was added into ABGS implants, and the quantity, quality, and longevity of the induced bone were assessed by micro-CT, histomorphometry, and histology over a period of 365 days. Local use of ZOL in the ABGS implants with ceramics had no influence on the bone volume (BV) on day 14 but subsequently significantly increased BV on days 35, 50, 105, 140, and 365 compared to the control implants. Locally applied ZOL had a similar effect in all of the applied doses (2–20 µg), while its systemic use on stimulating the BV of newly induced bone by ABGS depended on the time of application. BV was increased when ZOL was applied systemically on day 14 but had no effect when applied on day 35. The administration of ZOL bound to ceramics in ABGS increased and maintained the BV over a period of one year, offering a novel bone tissue engineering strategy for treating bone defects and spinal fusions.
Collapse
Affiliation(s)
- Nikola Stokovic
- Laboratory for Mineralized Tissues, Centre for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia; (N.S.); (N.I.); (I.E.)
| | - Natalia Ivanjko
- Laboratory for Mineralized Tissues, Centre for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia; (N.S.); (N.I.); (I.E.)
| | - Igor Erjavec
- Laboratory for Mineralized Tissues, Centre for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia; (N.S.); (N.I.); (I.E.)
| | - Anita Breski
- Department of Pathology and Cytology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
| | - Mihaela Peric
- Department for Intracellular Communication, Centre for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia;
| | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues, Centre for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia; (N.S.); (N.I.); (I.E.)
- Correspondence:
| |
Collapse
|
13
|
He W, Wu Z, Wu Y, Zhong Z, Hong Y. Construction of the Gypsum-Coated Scaffolds for In Situ Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2021; 13:31527-31541. [PMID: 34181398 DOI: 10.1021/acsami.1c08372] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
It is significant to use functional biomaterials to rationally engineer microenvironments for in situ bone regeneration in the field of bone tissue engineering. To this end, we constructed the gypsum-coated β-tricalcium phosphate (G-TCP) scaffolds by combing a three-dimensional printing technique and an epitaxial gypsum growth method. In vitro simulation experiments showed that the as-prepared scaffolds could establish a dynamic and weakly acidic microenvironment in a simulated body liquid, in which the pH and the calcium ion concentration always changed due to the gypsum degradation and growth of bone-like apatite nanoplates on the scaffold surfaces. The cell experiments confirmed that the microenvironment established by the G-TCP surfaces promoted rapid osteogenic differentiation and proliferation of bone marrow mesenchymal stem cells (BM-MSCs). In vivo experiments confirmed that the G-TCP scaffolds had high bioactivity in modulating in situ regeneration of bone, and the bioactivity of the G-TCP scaffolds was endowed by correct pore structures, degradation of gypsum, and growth of a bone-like apatite layer. The microenvironment established by the gypsum degradation could stimulate tissue inflammation and recruit white blood cells and BM-MSCs and thus accelerating native healing cascades of the bone defects via a bone growth/remodeling-absorption cycle process. Furthermore, in vivo experiments demonstrated that after the bone defects had healed completely, the as-prepared scaffolds also degraded completely within 24 weeks.
Collapse
Affiliation(s)
- Wenchao He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Zhen Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Yanmei Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Zhou Zhong
- Department of Orthopaedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youliang Hong
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| |
Collapse
|
14
|
Hayashi K, Ishikawa K. Honeycomb scaffolds capable of ectopic osteogenesis: Histological evaluation of osteoinduction mechanism. NANO SELECT 2021. [DOI: 10.1002/nano.202000283] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Koichiro Hayashi
- Department of Biomaterials Faculty of Dental Science Kyushu University Higashi‐ku Fukuoka Japan
| | - Kunio Ishikawa
- Department of Biomaterials Faculty of Dental Science Kyushu University Higashi‐ku Fukuoka Japan
| |
Collapse
|
15
|
Wang H, Zhou Y, Li CQ, Chu TW, Wang J, Huang B. Tissue-engineered bone used in a rabbit model of lumbar intertransverse process fusion: A comparison of osteogenic capacity between two different stem cells. Exp Ther Med 2020; 19:2570-2578. [PMID: 32256736 PMCID: PMC7086144 DOI: 10.3892/etm.2020.8523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 03/15/2019] [Indexed: 12/20/2022] Open
Abstract
Spinal fusion serves an important role in the reconstruction of spinal stability via restoration of the normal spinal sequence and relief of pain. Studies have demonstrated that the fusion rate is mainly associated with the osteogenic capacity of the implanted graft. Mesenchymal stem cells (MSCs) have been successfully isolated from human degenerated cartilage endplate (CEP) and designated as CEP-derived stem cells (CESCs). Previous studies have suggested that CESCs possesses in vitro and in vivo chondrogenic potential superior to that of bone marrow (BM)-MSCs. In addition, CESCs have shown a stronger in vitro osteogenic ability. The present study aimed to further determine the in vivo three-dimensional osteogenesis efficacy of CESCs for spinal fusion. Tissue-engineered bone grafts were transplanted into a rabbit model of posterolateral lumbar intertransverse process fusion using CESCs and BM-MSCs as seed cells composited with porous hydroxyapatite (PHA). The results of manual palpation and computed tomography (CT) scan reconstruction indicated that the CESCs/PHA group had a higher fusion rate than the BM-MSCs/PHA group, although the difference was not observed to be statistically significant. In addition, RT-qPCR results revealed that the in vitro CESCs/PHA composite expressed significantly higher levels of osteogenic-specific mRNA compared with the BM-MSCs/PHA composite. Finally, micro-CT and semi-quantitative histological analysis further demonstrated that the newly formed bone quality of the CESCs/PHA group was significantly higher than that of the BM-MSCs/PHA group in the intertransverse process fusion model. Therefore, the study indicated that CESCs possess superior in vivo osteogenesis capacity compared with BM-MSCs, and might serve as an important alternative seed cell source for bone tissue engineering. These results may provide the foundation for a biological solution to spinal fusion or other bone defect issues.
Collapse
Affiliation(s)
- Hai Wang
- Department of Orthopaedics, Xingsha Branch, Hunan Provincial People's Hospital, Changsha, Hunan 410000, P.R. China.,Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China.,Department of Orthopaedic and Trauma Surgery, Hunan Provincial People's Hospital, Changsha, Hunan 410000, P.R. China.,Faculty of Life Science, Kunming University of Science and Technology, Kunming, Yunnan 650093, P.R. China
| | - Yue Zhou
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Chang-Qing Li
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Tong-Wei Chu
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Jian Wang
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Bo Huang
- Department of Orthopaedics, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
16
|
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
|
17
|
Bighetti ACC, Cestari TM, Santos PS, Arantes RVN, Paini S, Assis GF, Costa BC, de Oliveira FA, Tokuhara CK, de Oliveira RC, Taga R. In vitro and in vivo assessment of CaP materials for bone regenerative therapy. The role of multinucleated giant cells/osteoclasts in bone regeneration. J Biomed Mater Res B Appl Biomater 2019; 108:282-297. [PMID: 31009176 DOI: 10.1002/jbm.b.34388] [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: 10/29/2018] [Revised: 03/22/2019] [Accepted: 03/30/2019] [Indexed: 12/15/2022]
Abstract
In this work, bone formation/remodeling/maturation was correlated with the presence of multinucleated giant cells (MGCs)/osteoclasts (tartrate-resistant acid phosphatase [TRAP]-positive cells) on the surface of beta-tricalcium phosphate (β-TCP), sintered deproteinized bovine bone (sDBB), and carbonated deproteinized bovine bone (cDBB) using a maxillary sinus augmentation (MSA) in a New Zealand rabbit model. Microtomographic, histomorphometric, and immunolabeling for TRAP-cells analyses were made at 15, 30, and 60 days after surgery. In all treatments, a faster bone formation/remodeling/maturation and TRAP-positive cells activity occurred in the osteotomy region of the MSA than in the middle and submucosa regions. In the β-TCP, the granules were rapidly reabsorbed by TRAP-positive cells and replaced by bone tissue. β-TCP enabled quick bone regeneration/remodeling and full bone and marrow restoration until 60 days, but with a significant reduction in MSA volume. In cDBB and sDBB, the quantity of TRAP-positive cells was smaller than in β-TCP, and these cells were associated with granule surface preparation for osteoblast-mediated bone formation. After 30 days, more than 80% of granule surfaces were surrounded and integrated by bone tissue without signs of degradation, preserving the MSA volume. Overall, the materials tested in a standardized preclinical model led to different bone formation/remodeling/maturation within the same repair process influenced by different microenvironments and MGCs/osteoclasts. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 108B:282-297, 2020.
Collapse
Affiliation(s)
- Ana Carolina Cestari Bighetti
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Tania Mary Cestari
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Paula Sanches Santos
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Ricardo Vinicius Nunes Arantes
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Suelen Paini
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Gerson Francisco Assis
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Bruna Carolina Costa
- Physics Department, Advanced Materials Laboratory, São Paulo State University, UNESP, Avenue Luiz Edmundo Carrijo Coube 14-01, Bauru, São Paulo, 17033-360, Brazil
| | - Flávia Amadeu de Oliveira
- Laboratory of Biochemistry of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Cintia Kazuko Tokuhara
- Laboratory of Biochemistry of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Rodrigo Cardoso de Oliveira
- Laboratory of Biochemistry of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Rumio Taga
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| |
Collapse
|
18
|
Humbert P, Brennan MÁ, Davison N, Rosset P, Trichet V, Blanchard F, Layrolle P. Immune Modulation by Transplanted Calcium Phosphate Biomaterials and Human Mesenchymal Stromal Cells in Bone Regeneration. Front Immunol 2019; 10:663. [PMID: 31001270 PMCID: PMC6455214 DOI: 10.3389/fimmu.2019.00663] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/11/2019] [Indexed: 12/22/2022] Open
Abstract
A wide variety of biomaterials have been developed as both stabilizing structures for the injured bone and inducers of bone neoformation. They differ in chemical composition, shape, porosity, and mechanical properties. The most extensively employed and studied subset of bioceramics are calcium phosphate materials (CaPs). These materials, when transplanted alongside mesenchymal stem cells (MSCs), lead to ectopic (intramuscular and subcutaneous) and orthotopic bone formation in preclinical studies, and effective fracture healing in clinical trials. Human MSC transplantation in pre-clinical and clinical trials reveals very low engraftment in spite of successful clinical outcomes and their therapeutic actions are thought to be primarily through paracrine mechanisms. The beneficial role of transplanted MSC could rely on their strong immunomodulatory effect since, even without long-term engraftment, they have the ability to alter both the innate and adaptive immune response which is critical to facilitate new bone formation. This study presents the current knowledge of the immune response to the implantation of CaP biomaterials alone or in combination with MSC. In particular the central role of monocyte-derived cells, both macrophages and osteoclasts, in MSC-CaP mediated bone formation is emphasized. Biomaterial properties, such as macroporosity and surface microstructure, dictate the host response, and the ultimate bone healing cascade. Understanding intercellular communications throughout the inflammation, its resolution and the bone regeneration phase, is crucial to improve the current therapeutic strategies or develop new approaches.
Collapse
Affiliation(s)
- Paul Humbert
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | - Meadhbh Á. Brennan
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Noel Davison
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- Instructure Labs, B.V., The Hague, Netherlands
| | - Philippe Rosset
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Centre Hospitalier Universitaire de Tours, Tours, France
| | - Valérie Trichet
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | | | - Pierre Layrolle
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| |
Collapse
|
19
|
Osteogenesis by foamed and 3D-printed nanostructured calcium phosphate scaffolds: Effect of pore architecture. Acta Biomater 2018; 79:135-147. [PMID: 30195084 DOI: 10.1016/j.actbio.2018.09.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/31/2018] [Accepted: 09/05/2018] [Indexed: 01/12/2023]
Abstract
There is an urgent need of synthetic bone grafts with enhanced osteogenic capacity. This can be achieved by combining biomaterials with exogenous growth factors, which however can have numerous undesired side effects, but also by tuning the intrinsic biomaterial properties. In a previous study, we showed the synergistic effect of nanostructure and pore architecture of biomimetic calcium deficient hydroxyapatite (CDHA) scaffolds in enhancing osteoinduction, i.e. fostering the differentiation of mesenchymal stem cells to bone forming cells. This was demonstrated by assessing bone formation after implanting the scaffolds intramuscularly. The present study goes one step forward, since it analyzes the effect of the geometrical features of the same CDHA scaffolds, obtained either by 3D-printing or by foaming, on the osteogenic potential and resorption behaviour in a bony environment. After 6 and 12 weeks of intraosseous implantation, both bone formation and material degradation had been drastically affected by the macropore architecture of the scaffolds. Whereas nanostructured CDHA was shown to be highly osteoconductive both in the robocast and foamed scaffolds, a superior osteogenic capacity was observed in the foamed scaffolds, which was associated with their higher intrinsic osteoinductive potential. Moreover, they showed a significantly higher cell-mediated degradation than the robocast constructs, with a simultaneous and progressive replacement of the scaffold by new bone. In conclusion, these results demonstrate that the control of macropore architecture is a crucial parameter in the design of synthetic bone grafts, which allows fostering both material degradation and new bone formation. Statement of Significance 3D-printing technologies open new perspectives for the design of patient-specific bone grafts, since they allow customizing the external shape together with the internal architecture of implants. In this respect, it is important to design the appropriate pore geometry to maximize the bone healing capacity of these implants. The present study analyses the effect of pore architecture of nanostructured hydroxyapatite scaffolds, obtained either by 3D-printing or foaming, on the osteogenic potential and scaffold resorption in an in vivo model. While nanostructured hydroxyapatite showed excellent osteoconductive properties irrespective of pore geometry, we demonstrated that the spherical, concave macropores of foamed scaffolds significantly promoted both material resorption and bone regeneration compared to the 3D-printed scaffolds with orthogonal-patterned struts and therefore prismatic, convex macropores.
Collapse
|
20
|
Klar RM. The Induction of Bone Formation: The Translation Enigma. Front Bioeng Biotechnol 2018; 6:74. [PMID: 29938204 PMCID: PMC6002665 DOI: 10.3389/fbioe.2018.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/22/2018] [Indexed: 11/25/2022] Open
Abstract
A paradigmatic shift in the way of thinking is what bone tissue engineering science requires to decrypt the translation conundrum from animal models into human. The deductive work of Urist (1965), who discerned the principle of bone induction from the pioneering works of Senn, Huggins, Lacroix, Levander, and other bone regenerative scientists, provided the basis that has assisted future bone tissue regenerative scientists to extend the bone tissue engineering field and its potential uses for bone regenerative medicine in humans. However, major challenges remain that are preventing the formation of bone by induction clinically. Growing experimental evidence is indicating that bone inductive studies are non-translatable from animal models into a clinical environment. This is preventing bone tissue engineering from reaching the next phase in development. Countless studies are trying to discern how the formation of bone by induction functions mechanistically, so as to try and solve this enigmatic problem. However, are the correct questions being asked? Why do bone inductive animal studies not translate into humans? Why do bone induction principles not yield the same extent of bone formation as an autogenous bone graft? What are bone tissue engineering scientists missing? By critically re-assessing the past and present discoveries of the bone induction field, this review article attempts to re-discover the field of bone formation by induction, identifying some key features that may have been missed. These include a detailed library of all proteins in bones and their arrangement in the 3D superstructure of the bone together with some other important criteria not considered by tissue engineering scientists. The review therefore not only re-iterates possible avenues of research that need to be re-explored but also seeks to guide present and future scientists in how they assess their own research in light of experimental design and results. By addressing these issues bone formation by induction without autografts might finally become clinically viable.
Collapse
Affiliation(s)
- Roland M. Klar
- Laboratory of Biomechanics and Experimental Orthopaedics, Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
21
|
Zhang W, Zhu J, Ma T, Liu C, Hai B, Du G, Wang H, Li N, Leng H, Xu Y, Song C. Comparison of the effects of once-weekly and once-daily rhPTH (1-34) injections on promoting fracture healing in rodents. J Orthop Res 2018; 36:1145-1152. [PMID: 28960481 DOI: 10.1002/jor.23750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/22/2017] [Indexed: 02/04/2023]
Abstract
To compare the efficacy of once-weekly and once-daily subcutaneous injections of teriparatide (recombinant human parathyroid hormone 1-34) on fracture healing, 50 adult male Sprague-Dawley rats were subjected to a unilateral tibia fracture and received internal fixation with a Kirschner needle. Based on the injection dose and frequency, the rats were randomly divided into five groups (n = 10 each): subcutaneous injections of saline or 10 µg/kg/w, 20 µg/kg/w, 10 µg/kg/d, and 20 µg/kg/d teriparatide. Four weeks later, the rats were euthanatized, and the fractured tibiae were assessed using X-rays, dual-energy X-ray absorptiometry, micro-computed tomography, the three-point bending biomechanics test, and histology. Compared to the saline control group, either daily or weekly subcutaneous injections of teriparatide significantly increased bone mass, improved the bone microarchitecture, and promoted fracture healing (p < 0.05). There were no significant differences in bone mineral density (BMD), bone microstructure or bone strength between the 20 µg/kg/w and 10 µg/kg/d groups (p > 0.05). Teriparatide 20 µg weekly injections promoted bone fracture healing to the same extent as teriparatide 10 µg daily injections, which can dramatically decrease the cumulative dosage of teriparatide injections. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1145-1152, 2018.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| | - Junxiong Zhu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| | - Teng Ma
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| | - Can Liu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| | - Bao Hai
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| | - Guohong Du
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Hong Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| | - Nan Li
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, 49 North Garden Rd Haidian District, Beijing, China
| |
Collapse
|
22
|
Ripamonti U. Functionalized Surface Geometries Induce: " Bone: Formation by Autoinduction". Front Physiol 2018; 8:1084. [PMID: 29467661 PMCID: PMC5808255 DOI: 10.3389/fphys.2017.01084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/08/2017] [Indexed: 12/15/2022] Open
Abstract
The induction of tissue formation, and the allied disciplines of tissue engineering and regenerative medicine, have flooded the twenty-first century tissue biology scenario and morphed into high expectations of a fulfilling regenerative dream of molecularly generated tissues and organs in assembling human tissue factories. The grand conceptualization of deploying soluble molecular signals, first defined by Turing as forms generating substances, or morphogens, stemmed from classic last century studies that hypothesized the presence of morphogens in several mineralized and non-mineralized mammalian matrices. The realization of morphogens within mammalian matrices devised dissociative extractions and chromatographic procedures to isolate, purify, and finally reconstitute the cloned morphogens, found to be members of the transforming growth factor-β (TGF-β) supergene family, with insoluble signals or substrata to induce de novo tissue induction and morphogenesis. Can we however construct macroporous bioreactors per se capable of inducing bone formation even without the exogenous applications of the osteogenic soluble molecular signals of the TGF-β supergene family? This review describes original research on coral-derived calcium phosphate-based macroporous constructs showing that the formation of bone is independent of the exogenous application of the osteogenic soluble signals of the TGF-β supergene family. Such signals are the molecular bases of the induction of bone formation. The aim of this review is to primarily describe today's hottest topic of biomaterials' science, i.e., to construct and define osteogenetic biomaterials' surfaces that per se, in its own right, do initiate the induction of bone formation. Biomaterials are often used to reconstruct osseous defects particularly in the craniofacial skeleton. Edentulism did spring titanium implants as tooth replacement strategies. No were else that titanium surfaces require functionalized geometric nanotopographic cues to set into motion osteogenesis independently of the exogenous application of the osteogenic soluble molecular signals. Inductive morphogenetic surfaces are the way ahead of biomaterials' science: the connubium of stem cells on primed functionalized surfaces precisely regulates gene expression and the induction of the osteogenic phenotype.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory, Faculty of Health Sciences, School of Oral Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
23
|
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
|
24
|
Barba A, Diez-Escudero A, Maazouz Y, Rappe K, Espanol M, Montufar EB, Bonany M, Sadowska JM, Guillem-Marti J, Öhman-Mägi C, Persson C, Manzanares MC, Franch J, Ginebra MP. Osteoinduction by Foamed and 3D-Printed Calcium Phosphate Scaffolds: Effect of Nanostructure and Pore Architecture. ACS APPLIED MATERIALS & INTERFACES 2017; 9:41722-41736. [PMID: 29116737 DOI: 10.1021/acsami.7b14175] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Some biomaterials are osteoinductive, that is, they are able to trigger the osteogenic process by inducing the differentiation of mesenchymal stem cells to the osteogenic lineage. Although the underlying mechanism is still unclear, microporosity and specific surface area (SSA) have been identified as critical factors in material-associated osteoinduction. However, only sintered ceramics, which have a limited range of porosities and SSA, have been analyzed so far. In this work, we were able to extend these ranges to the nanoscale, through the foaming and 3D-printing of biomimetic calcium phosphates, thereby obtaining scaffolds with controlled micro- and nanoporosity and with tailored macropore architectures. Calcium-deficient hydroxyapatite (CDHA) scaffolds were evaluated after 6 and 12 weeks in an ectopic-implantation canine model and compared with two sintered ceramics, biphasic calcium phosphate and β-tricalcium phosphate. Only foams with spherical, concave macropores and not 3D-printed scaffolds with convex, prismatic macropores induced significant ectopic bone formation. Among them, biomimetic nanostructured CDHA produced the highest incidence of ectopic bone and accelerated bone formation when compared with conventional microstructured sintered calcium phosphates with the same macropore architecture. Moreover, they exhibited different bone formation patterns; in CDHA foams, the new ectopic bone progressively replaced the scaffold, whereas in sintered biphasic calcium phosphate scaffolds, bone was deposited on the surface of the material, progressively filling the pore space. In conclusion, this study demonstrates that the high reactivity of nanostructured biomimetic CDHA combined with a spherical, concave macroporosity allows the pushing of the osteoinduction potential beyond the limits of microstructured calcium phosphate ceramics.
Collapse
Affiliation(s)
- Albert Barba
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Bone Healing Group, Small Animal Surgery Department, Veterinary School, Universitat Autònoma de Barcelona , 08193 Bellaterra, Barcelona, Spain
| | - Anna Diez-Escudero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Yassine Maazouz
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Katrin Rappe
- Bone Healing Group, Small Animal Surgery Department, Veterinary School, Universitat Autònoma de Barcelona , 08193 Bellaterra, Barcelona, Spain
| | - Montserrat Espanol
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Edgar B Montufar
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Mar Bonany
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Joanna M Sadowska
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Jordi Guillem-Marti
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Caroline Öhman-Mägi
- Materials in Medicine Group, Division of Applied Materials Science, Department of Engineering Sciences, Uppsala University , 751 21 Uppsala, Sweden
| | - Cecilia Persson
- Materials in Medicine Group, Division of Applied Materials Science, Department of Engineering Sciences, Uppsala University , 751 21 Uppsala, Sweden
| | - Maria-Cristina Manzanares
- Human Anatomy and Embryology Unit, Department of Pathology and Experimental Therapeutics, Universitat de Barcelona , 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Franch
- Bone Healing Group, Small Animal Surgery Department, Veterinary School, Universitat Autònoma de Barcelona , 08193 Bellaterra, Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC) , 08028 Barcelona, Spain
| |
Collapse
|
25
|
Bouler J, Pilet P, Gauthier O, Verron E. Biphasic calcium phosphate ceramics for bone reconstruction: A review of biological response. Acta Biomater 2017; 53:1-12. [PMID: 28159720 DOI: 10.1016/j.actbio.2017.01.076] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/04/2017] [Accepted: 01/27/2017] [Indexed: 12/23/2022]
Abstract
Autologous bone graft is considered as the gold standard in bone reconstructive surgery. However, the quantity of bone available is limited and the harvesting procedure requires a second surgical site resulting in severe complications. Due to these limits, scientists and clinicians have considered alternatives to autologous bone graft. Calcium phosphates (CaPs) biomaterials including biphasic calcium phosphate (BCP) ceramics have proven efficacy in numerous clinical indications. Their specific physico-chemical properties (HA/TCP ratio, dual porosity and subsequent interconnected architecture) control (regulate/condition) the progressive resorption and the bone substitution process. By describing the most significant biological responses reported in the last 30years, we review the main events that made their clinical success. We also discuss about their exciting future applications as osteoconductive scaffold for delivering various bioactive molecules or bone cells in bone tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE Nowadays, BCPs are definitely considered as the gold standard of bone substitutes in bone reconstructive surgery. Among the numerous clinical studies in literature demonstrating the performance of BCP, Passuti et al. and Randsford et al. studies largely contributed to the emergence of the BCPs. It could be interesting to come back to the main events that made their success and could explain their large adhesion from scientists to clinicians. This paper aims to review the most significant biological responses reported in the last 30years, of these BCP-based materials. We also discuss about their exciting future applications as osteoconductive scaffold for delivering various bioactive molecules or bone cells in bone tissue engineering and regenerative medicine.
Collapse
|
26
|
Zhang W, Liu C, Hai B, Du G, Wang H, Leng H, Xu Y, Song C. A Convenient In Vivo Model Using Small Interfering RNA Silencing to Rapidly Assess Skeletal Gene Function. PLoS One 2016; 11:e0167222. [PMID: 27893850 PMCID: PMC5125699 DOI: 10.1371/journal.pone.0167222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
It is difficult to study bone in vitro because it contains various cell types that engage in cross-talk. Bone biologically links various organs, and it has thus become increasingly evident that skeletal physiology must be studied in an integrative manner in an intact animal. We developed a model using local intraosseous small interfering RNA (siRNA) injection to rapidly assess the effects of a target gene on the local skeletal environment. In this model, 160-g male Sprague-Dawley rats were treated for 1-2 weeks. The left tibia received intraosseous injection of a parathyroid hormone 1 receptor (Pth1r) or insulin-like growth factor 1 receptor (Igf-1r) siRNA transfection complex loaded in poloxamer 407 hydrogel, and the right tibia received the same volume of control siRNA. All the tibias received an intraosseous injection of recombinant human parathyroid hormone (1-34) (rhPTH (1-34)) or insulin-like growth factor-1 (IGF-1). Calcein green and alizarin red were injected 6 and 2 days before euthanasia, respectively. IGF-1R and PTH1R expression levels were detected via RT-PCR assays and immunohistochemistry. Bone mineral density (BMD), microstructure, mineral apposition rates (MARs), and strength were determined by dual-energy X-ray absorptiometry, micro-CT, histology and biomechanical tests. The RT-PCR and immunohistochemistry results revealed that IGF-1R and PTH1R expression levels were dramatically diminished in the siRNA-treated left tibias compared to the right tibias (both p<0.05). Using poloxamer 407 hydrogel as a controlled-release system prolonged the silencing effect of a single dose of siRNA; the mRNA expression levels of IGF-1R were lower at two weeks than at one week (p<0.01). The BMD, bone microstructure parameters, MAR and bone strength were significantly decreased in the left tibias compared to the right tibias (all p<0.05). This simple and convenient local intraosseous siRNA injection model achieved gene silencing with very small quantities of siRNA over a short treatment period (≤7 days).
Collapse
Affiliation(s)
- Wen Zhang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Can Liu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Bao Hai
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Guohong Du
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Hong Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
- * E-mail:
| |
Collapse
|
27
|
Ripamonti U, Parak R, Klar RM, Dickens C, Dix-Peek T, Duarte R. The synergistic induction of bone formation by the osteogenic proteins of the TGF-β supergene family. Biomaterials 2016; 104:279-96. [DOI: 10.1016/j.biomaterials.2016.07.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/12/2016] [Accepted: 07/16/2016] [Indexed: 12/28/2022]
|
28
|
Liu X, Li M, Zhu Y, Yeung KWK, Chu PK, Wu S. The modulation of stem cell behaviors by functionalized nanoceramic coatings on Ti-based implants. Bioact Mater 2016; 1:65-76. [PMID: 29744396 PMCID: PMC5883996 DOI: 10.1016/j.bioactmat.2016.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 12/25/2022] Open
Abstract
Nanoceramic coating on the surface of Ti-based metallic implants is a clinical potential option in orthopedic surgery. Stem cells have been found to have osteogenic capabilities. It is necessary to study the influences of functionalized nanoceramic coatings on the differentiation and proliferation of stem cells in vitro or in vivo. In this paper, we summarized the recent advance on the modulation of stem cells behaviors through controlling the properties of nanoceramic coatings, including surface chemistry, surface roughness and microporosity. In addition, mechanotransduction pathways have also been discussed to reveal the interaction mechanisms between the stem cells and ceramic coatings on Ti-based metals. In the final part, the osteoinduction and osteoconduction of ceramic coating have been also presented when it was used as carrier of BMPs in new bone formation. The effects of basic physical properties like roughness, topography and porous stucture of ceramic coatings on the stem cells behaviors on Ti-based alloys have been reviewed together. The chemical way to modulate the cell behaviors is also discussed in this review paper; and the related mechanotransduction pathways have been described in this paper.
Collapse
Affiliation(s)
- Xiangmei Liu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Faculty of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Man Li
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Faculty of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Yizhou Zhu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Faculty of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - K W K Yeung
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong Shenzhen Hospital, 1 Haiyuan 1st Road, Futian District, Shenzhen, China.,Division of Spine Surgery, Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Paul K Chu
- Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Shuilin Wu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Faculty of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
29
|
Ripamonti U, Duarte R, Parak R, Dickens C, Dix-Peek T, Klar RM. Redundancy and Molecular Evolution: The Rapid Induction of Bone Formation by the Mammalian Transforming Growth Factor-β3 Isoform. Front Physiol 2016; 7:396. [PMID: 27660615 PMCID: PMC5014861 DOI: 10.3389/fphys.2016.00396] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/25/2016] [Indexed: 11/26/2022] Open
Abstract
The soluble osteogenic molecular signals of the transforming growth factor-β (TGF-β) supergene family are the molecular bases of the induction of bone formation and postnatal bone tissue morphogenesis with translation into clinical contexts. The mammalian TGF-β3 isoform, a pleiotropic member of the family, controls a vast array of biological processes including the induction of bone formation. Recombinant hTGF-β3 induces substantial bone formation when implanted with either collagenous bone matrices or coral-derived macroporous bioreactors in the rectus abdominis muscle of the non-human primate Papio ursinus. In marked contrast, the three mammalian TGF-βs do not initiate the induction of bone formation in rodents and lagomorphs. The induction of bone by hTGF-β3/preloaded bioreactors is orchestrated by inducing fibrin-fibronectin rings that structurally organize tissue patterning and morphogenesis within the macroporous spaces. Induced advancing extracellular matrix rings provide the structural anchorage for hyper chromatic cells, interpreted as differentiating osteoblasts re-programmed by hTGF-β3 from invading myoblastic and/or pericytic differentiated cells. Runx2 and Osteocalcin expression are significantly up-regulated correlating to multiple invading cells differentiating into the osteoblastic phenotype. Bioreactors pre-loaded with recombinant human Noggin (hNoggin), a BMPs antagonist, show down-regulation of BMP-2 and other profiled osteogenic proteins' genes resulting in minimal bone formation. Coral-derived macroporous constructs preloaded with binary applications of hTGF-β3 and hNoggin also show down-regulation of BMP-2 with the induction of limited bone formation. The induction of bone formation by hTGF-β3 is via the BMPs pathway and it is thus blocked by hNoggin. Our systematic studies in P. ursinus with translational hTGF-β3 in large cranio-mandibulo-facial defects in humans are now requesting the re-evaluation of "Bone: formation by autoinduction" in primate models including humans.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory, Faculty of Health Sciences, School of Oral Health Sciences, University of the WitwatersrandJohannesburg, South Africa
| | - Raquel Duarte
- Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the WitwatersrandJohannesburg, South Africa
| | - Ruqayya Parak
- Bone Research Laboratory, Faculty of Health Sciences, School of Oral Health Sciences, University of the WitwatersrandJohannesburg, South Africa
- Department of Oral Biological Sciences, School of Oral Health Sciences, University of the WitwatersrandJohannesburg, South Africa
| | - Caroline Dickens
- Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the WitwatersrandJohannesburg, South Africa
| | - Therese Dix-Peek
- Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the WitwatersrandJohannesburg, South Africa
| | - Roland M. Klar
- Bone Research Laboratory, Faculty of Health Sciences, School of Oral Health Sciences, University of the WitwatersrandJohannesburg, South Africa
- Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the WitwatersrandJohannesburg, South Africa
| |
Collapse
|
30
|
Ripamonti U, Klar RM, Parak R, Dickens C, Dix-Peek T, Duarte R. Tissue segregation restores the induction of bone formation by the mammalian transforming growth factor-β(3) in calvarial defects of the non-human primate Papio ursinus. Biomaterials 2016; 86:21-32. [PMID: 26874889 DOI: 10.1016/j.biomaterials.2016.01.071] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 01/31/2016] [Indexed: 12/12/2022]
Abstract
A diffusion molecular hypothesis from the dura and/or the leptomeninges below that would control the induction of calvarial membranous bone formation by the recombinant human transforming growth factor-β3 (hTGF-β3) was investigated. Coral-derived calcium carbonate-based macroporous constructs (25 mm diameter; 3.5/4 mm thickness) with limited hydrothermal conversion to hydroxyapatite (7% HA/CC) were inserted into forty calvarial defects created in 10 adult Chacma baboons Papio ursinus. In 20 defects, an impermeable nylon foil membrane (SupraFOIL(®)) was inserted between the cut endocranial bone and the underlying dura mater. Twenty of the macroporous constructs were preloaded with hTGF-β3 (125 μg in 1000 μl 20 mM sodium succinate, 4% mannitol pH4.0), 10 of which were implanted into defects segregated by the SupraFOIL(®) membrane, and 10 into non-segregated defects. Tissues were harvested on day 90, processed for decalcified and undecalcified histology and quantitative real-time polymerase chain reaction (qRT-PCR). Segregated untreated macroporous specimens showed a reduction of bone formation across the macroporous spaces compared to non-segregated constructs. qRT-PCR of segregated untreated specimens showed down regulation of osteogenic protein-1 (OP-1), osteocalcin (OC), bone morphogenetic protein-2 (BMP-2), RUNX-2 and inhibitor of DNA binding-2 and -3 (ID2,ID3) and up regulation of TGF-β3, a molecular signalling pathway inhibiting the induction of membranous bone formation. Non-segregated hTGF-β3/treated constructs also showed non-osteogenic expression profiles when compared to non-segregated untreated specimens. Segregated hTGF-β3/treated 7% HA/CC constructs showed significantly greater induction of bone formation across the macroporous spaces and, compared to non-segregated hTGF-β3/treated constructs, showed up regulation of OP-1, OC, BMP-2, RUNX-2, ID2 and ID3. Similar up-regulated expression profiles were seen for untreated non-segregated constructs. TGF-β signalling via ID genes creates permissive or refractory micro-environments that regulate the induction of calvarial bone formation which is controlled by the exogenous hTGF-β3 upon segregation of the calvarial defects. The dura is the common regulator of the induction of calvarial bone formation modulated by the presence or absence of the SupraFOIL(®) membrane with or without hTGF-β3.
Collapse
Affiliation(s)
- U Ripamonti
- Bone Research Laboratory, School of Oral Health Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Roland Manfred Klar
- Bone Research Laboratory, School of Oral Health Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Molecular and Cellular Biology Laboratories, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ruqayya Parak
- Bone Research Laboratory, School of Oral Health Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Department of Oral Biological Sciences, School of Oral Health Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline Dickens
- Molecular and Cellular Biology Laboratories, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Therese Dix-Peek
- Molecular and Cellular Biology Laboratories, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Raquel Duarte
- Molecular and Cellular Biology Laboratories, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
31
|
Tan J, Fu X, Sun CG, Liu C, Zhang XH, Cui YY, Guo Q, Ma T, Wang H, Du GH, Yin X, Liu ZJ, Leng HJ, Xu YS, Song CL. A single CT-guided percutaneous intraosseous injection of thermosensitive simvastatin/poloxamer 407 hydrogel enhances vertebral bone formation in ovariectomized minipigs. Osteoporos Int 2016. [PMID: 26223190 DOI: 10.1007/s00198-015-3230-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
UNLABELLED The ultimate goal of osteoporosis treatment is prevention of fragile fracture. Local treatment targeting specific bone may decrease the incidence of osteoporotic fractures. We developed an injectable, thermosensitive simvastatin/poloxamer 407 hydrogel; a single CT-guided percutaneous intraosseous injection augmented vertebrae in ovariectomized minipigs. INTRODUCTION The greatest hazard associated with osteoporosis is local fragility fractures. An adjunct, local treatment might be helpful to decrease the incidence of osteoporotic fracture. Studies have found that simvastatin stimulates bone formation, but the skeletal bioavailability of orally administered is low. Directly delivering simvastatin to the specific bone that is prone to fractures may reinforce the target bone and reduce the incidence of fragility fractures. METHODS We developed an injectable, thermosensitive simvastatin/poloxamer 407 hydrogel, conducted scanning electron microscopy, rheological, and drug release analyses to evaluate the delivery system; injected it into the lumbar vertebrae of ovariectomized minipigs via minimally invasive CT-guided percutaneous vertebral injection. Three months later, BMD, microstructures, mineral apposition rates, and strength were determined by DXA, micro-CT, histology, and biomechanical test; expression of VEGF, BMP2, and osteocalcin were analyzed by immunohistochemistry and Western blots. RESULTS Poloxamer 407 is an effective controlled delivery system for intraosseous-injected simvastatin. A single injection of the simvastatin/poloxamer 407 hydrogel significantly increased BMD, bone microstructure, and strength; the bone volume fraction and trabecular thickness increased nearly 150 %, bone strength almost doubled compared with controls (all P < 0.01); and induced higher expression of VEGF, BMP2, and osteocalcin. CONCLUSIONS CT-guided percutaneous vertebral injection of a single simvastatin/poloxamer 407 thermosensitive hydrogel promotes bone formation in ovariectomized minipigs. The underlying mechanism appears to involve the higher expression of VEGF and BMP-2.
Collapse
MESH Headings
- Absorptiometry, Photon/methods
- Animals
- Bone Density/drug effects
- Bone Morphogenetic Protein 2/metabolism
- Chemistry, Physical
- Drug Combinations
- Drug Delivery Systems
- Drug Evaluation, Preclinical/methods
- Female
- Hydrogel, Polyethylene Glycol Dimethacrylate
- Injections, Spinal
- Lumbar Vertebrae/diagnostic imaging
- Lumbar Vertebrae/metabolism
- Lumbar Vertebrae/physiopathology
- Microscopy, Electron, Scanning
- Osteogenesis/drug effects
- Osteoporosis/diagnostic imaging
- Osteoporosis/drug therapy
- Osteoporosis/physiopathology
- Ovariectomy
- Poloxamer/administration & dosage
- Poloxamer/chemistry
- Poloxamer/pharmacology
- Poloxamer/therapeutic use
- Radiography, Interventional
- Rheology
- Simvastatin/administration & dosage
- Simvastatin/pharmacology
- Simvastatin/therapeutic use
- Swine
- Swine, Miniature
- Tomography, X-Ray Computed
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- J Tan
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - X Fu
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - C G Sun
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, 100191, China
| | - C Liu
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China
| | - X H Zhang
- Department of Pharmacology, Peking University Third Hospital, Beijing, 100191, China
| | - Y Y Cui
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Q Guo
- Department of Neurology, Peking University Third Hospital, Beijing, 100191, China
| | - T Ma
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - H Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, 100191, China
| | - G H Du
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, 100191, China
| | - X Yin
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, 100191, China
| | - Z J Liu
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, 100191, China
| | - H J Leng
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, 100191, China
| | - Y S Xu
- Department of Neurology, Peking University Third Hospital, Beijing, 100191, China
| | - C L Song
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China.
- Beijing Key Laboratory of Spinal Diseases, Beijing, 100191, China.
| |
Collapse
|
32
|
Shiwaku Y, Neff L, Nagano K, Takeyama KI, de Bruijn J, Dard M, Gori F, Baron R. The Crosstalk between Osteoclasts and Osteoblasts Is Dependent upon the Composition and Structure of Biphasic Calcium Phosphates. PLoS One 2015; 10:e0132903. [PMID: 26193362 PMCID: PMC4507990 DOI: 10.1371/journal.pone.0132903] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/22/2015] [Indexed: 11/18/2022] Open
Abstract
Biphasic calcium phosphates (BCPs), consisting of hydroxyapatite (HA) and β-tricalcium phosphate (β-TCP), exhibit good biocompatibility and osteoconductivity, maintaining a balance between resorption of the biomaterial and formation of new bone. We tested whether the chemical composition and/or the microstructure of BCPs affect osteoclasts (OCs) differentiation and/or their ability to crosstalk with osteoblasts (OBs). To this aim, OCs were cultured on BCPs with HA content of 5, 20 or 60% and their differentiation and activity were assessed. We found that OC differentiation is partially impaired by increased HA content, but not by the presence of micropores within BCP scaffolds, as indicated by TRAP staining and gene profile expression. We then investigated whether the biomaterial-induced changes in OC differentiation also affect their ability to crosstalk with OBs and regulate OB function. We found that BCPs with low percentage of HA favored the expression of positive coupling factors, including sphingosine-kinase 1 (SPHK1) and collagen triple helix repeat containing 1 (Cthrc1). In turn, the increase of these secreted coupling factors promotes OB differentiation and function. All together our studies suggest that the chemical composition of biomaterials affects not only the differentiation and activity of OCs but also their potential to locally regulate bone formation.
Collapse
Affiliation(s)
- Yukari Shiwaku
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Lynn Neff
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Kenichi Nagano
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Ken-Ichi Takeyama
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States of America
| | | | - Michel Dard
- Department of Periodontology and Implant Dentistry, New York University College of Dentistry, New York, NY, United States of America
| | - Francesca Gori
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Roland Baron
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, United States of America
- Department of Medicine, Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
33
|
Tan J, Yang N, Fu X, Cui Y, Guo Q, Ma T, Yin X, Leng H, Song C. Single-dose local simvastatin injection improves implant fixation via increased angiogenesis and bone formation in an ovariectomized rat model. Med Sci Monit 2015; 21:1428-39. [PMID: 25982481 PMCID: PMC4448596 DOI: 10.12659/msm.892247] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Statins have been reported to promote bone formation. However, taken orally, their bioavailability is low to the bones. Implant therapies require a local repair response, topical application of osteoinductive agents, or biomaterials that promote implant fixation. Material/Methods The present study evaluated the effect of a single local injection of simvastatin on screw fixation in an ovariectomized rat model of osteoporosis. Results Dual-energy X-ray absorptiometry, micro-computed tomography, histology, and biomechanical tests revealed that 5 and 10 mg simvastatin significantly improved bone mineral density by 18.2% and 22.4%, respectively (P<0.05); increased bone volume fraction by 51.0% and 57.9%, trabecular thickness by 16.4% and 18.9%, trabeculae number by 112.0% and 107.1%, and percentage of osseointegration by 115.7% and 126.3%; and decreased trabeculae separation by 34.1% and 36.6%, respectively (all P<0.01). Bone mineral apposition rate was significantly increased (P<0.01). Furthermore, implant fixation was significantly increased (P<0.05), and bone morphogenetic protein 2 (BMP2) expression was markedly increased. Local injection of a single dose of simvastatin also promoted angiogenesis. Vessel number, volume, thickness, surface area, and vascular volume per tissue volume were significantly increased (all P<0.01). Vascular endothelial growth factor (VEGF), VEGF receptor-2, von Willebrand factor, and platelet endothelial cell adhesion molecule-1 expression were enhanced. Conclusions A single local injection of simvastatin significantly increased bone formation, promoted osseointegration, and enhanced implant fixation in ovariectomized rats. The underlying mechanism appears to involve enhanced BMP2 expression and angiogenesis in the target bone.
Collapse
Affiliation(s)
- Jie Tan
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| | - Ning Yang
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| | - Xin Fu
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| | - Yueyi Cui
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| | - Qi Guo
- Department of Neurology, Peking University Third Hospital, Beijing, China (mainland)
| | - Teng Ma
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| | - Xiaoxue Yin
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| | - Huijie Leng
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| | - Chunli Song
- Department of Orthopaedic, Peking University Third Hospital, Beijing, China (mainland)
| |
Collapse
|
34
|
Ripamonti U, Dix-Peek T, Parak R, Milner B, Duarte R. Profiling bone morphogenetic proteins and transforming growth factor-βs by hTGF-β3 pre-treated coral-derived macroporous bioreactors: The power of one. Biomaterials 2015; 49:90-102. [DOI: 10.1016/j.biomaterials.2015.01.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/13/2015] [Accepted: 01/20/2015] [Indexed: 12/27/2022]
|
35
|
Tsukanaka M, Fujibayashi S, Otsuki B, Takemoto M, Matsuda S. Osteoinductive potential of highly purified porous β-TCP in mice. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:132. [PMID: 25698341 DOI: 10.1007/s10856-015-5469-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 02/01/2015] [Indexed: 06/04/2023]
Abstract
Material-induced osteoinduction of calcium phosphate ceramics has been reported in specific animals. We previously reported that recruitment of tartrate-resistant acid phosphatase (TRAP)-positive cells might be one of the main factors responsible for the difference in the occurrence of material-induced osteoinduction between dogs and rats. In this study, we evaluated the osteoinductive potential of highly purified porous beta-tricalcium phosphate materials (HPP-β-TCP) with two different porosities, 75 and 60 % (Olympus Terumo Biomaterials, Tokyo, Japan), implanted into subcutaneous pockets of FVB and C57BL/6 mice. Twelve weeks after implantation, histological examination and gene expression analysis using reverse transcription-polymerase chain reaction were performed. We observed osteoinduction in half of the HPP-β-TCP materials with 60 % porosity implanted into FVB mice. This group of mice also exhibited the most TRAP-positive cells. Significantly more vessels were found in FVB mice than in C57BL/6 mice, but the greatest number of vessels was counted in implants from materials with 75 % porosity implanted into FVB mice, which did not show osteoinduction. These results indicate that recruitment of TRAP-positive cells is one factor responsible for osteoinduction caused by HPP-β-TCP materials in both FVB mice and dogs. Vessel formation seems to be necessary but appears to be less influential for osteoinduction than TRAP-positive cell recruitment.
Collapse
Affiliation(s)
- Masako Tsukanaka
- Center for Implant and Radiostereometric Research Oslo, Oslo University Hospital Ullevål, Kirkeveien 166, 0407, Oslo, Norway,
| | | | | | | | | |
Collapse
|
36
|
Lee JH, Baek HR, Lee KM, Zheng GB, Shin SJ, Jin YZ. The inhibitory effect of zoledronate on early-stage osteoinduction by recombinant human bone morphogenetic protein 2 in an osteoporosis model. Growth Factors 2015; 33:220-8. [PMID: 26099999 DOI: 10.3109/08977194.2015.1058259] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study evaluated the effect of the combined treatment of intravenous zoledronic acid (ZA, 0.08 mg/kg) and rhBMP-2 (5 µg) on osteogenesis in a calvarial defect model of ovariectomized SD rats. New bone formation was evaluated 4 or 8 weeks after calvarial defect implantation using micro-CT and histology. Micro-CT results revealed that the rhBMP-2 group showed significantly higher calvarial defect coverage ratio compared with the ZA + rhBMP-2 group at 4 weeks. In addition, bone formation indices were significantly lower in ZA + rhBMP-2 group when compared with the rhBMP-2 group after 4 weeks, which indicates a negative effect of ZA on the initial bone formation and the bone quality. At 8 weeks, the negative effect induced by ZA treatment was alleviated as time passed. Histological examination showed similar results to the micro-CT measurements. In conclusion, although ZA treatment lowered the new bone formation induced by rhBMP-2 initially, as time passed, the negative effect was decreased.
Collapse
Affiliation(s)
- Jae Hyup Lee
- a Department of Orthopedic Surgery , College of Medicine, Seoul National University, SMG-SNU Boramae Medical Center , Seoul , Republic of Korea and
- b Institute of Medical and Biological Engineering, Seoul National University Medical Research Center , Seoul , Republic of Korea
| | - Hae-Ri Baek
- a Department of Orthopedic Surgery , College of Medicine, Seoul National University, SMG-SNU Boramae Medical Center , Seoul , Republic of Korea and
| | - Kyung Mee Lee
- a Department of Orthopedic Surgery , College of Medicine, Seoul National University, SMG-SNU Boramae Medical Center , Seoul , Republic of Korea and
| | - Guang Bin Zheng
- a Department of Orthopedic Surgery , College of Medicine, Seoul National University, SMG-SNU Boramae Medical Center , Seoul , Republic of Korea and
| | - Sung Joon Shin
- a Department of Orthopedic Surgery , College of Medicine, Seoul National University, SMG-SNU Boramae Medical Center , Seoul , Republic of Korea and
| | - Yuan Zhe Jin
- a Department of Orthopedic Surgery , College of Medicine, Seoul National University, SMG-SNU Boramae Medical Center , Seoul , Republic of Korea and
| |
Collapse
|
37
|
Re-evaluating the induction of bone formation in primates. Biomaterials 2014; 35:9407-22. [DOI: 10.1016/j.biomaterials.2014.07.053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 07/21/2014] [Indexed: 12/20/2022]
|
38
|
Davison NL, ten Harkel B, Schoenmaker T, Luo X, Yuan H, Everts V, Barrère-de Groot F, de Bruijn JD. Osteoclast resorption of beta-tricalcium phosphate controlled by surface architecture. Biomaterials 2014; 35:7441-51. [PMID: 24927681 DOI: 10.1016/j.biomaterials.2014.05.048] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/16/2014] [Indexed: 01/13/2023]
Abstract
A resorbable bone graft substitute should mimic native bone in its capacity to support bone formation and be remodeled by osteoclasts (OCl) or other multinucleated cells such as foreign body giant cells (FBGC). We hypothesize that by changing the scale of surface architecture of beta-tricalcium phosphate (TCP), cellular resorption can be influenced. CD14(+) monocyte precursors were isolated from human peripheral blood (n = 4 independent donors) and differentiated into OCl or FBGC on the surface of TCP discs comprising either submicron- or micron-scale surface topographical features (TCPs and TCPb, respectively). On submicrostructured TCPs, OCl survived, fused, differentiated, and extensively resorbed the substrate; however, on microstructured TCPb, OCl survival, TRAP activation, and fusion were attenuated. Importantly, no resorption was observed on microstructured TCPb. By confocal microscopy, OCl formed on TCPs contained numerous actin rings allowing for resorption, but not on TCPb. In comparison, FBGC could not resorb either TCP material, suggesting that osteoclast-specific machinery is necessary to resorb TCP. By tuning surface architecture, it appears possible to control osteoclast resorption of calcium phosphate. This approach presents a useful strategy in the design of resorbable bone graft substitutes.
Collapse
Affiliation(s)
- Noel L Davison
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, Netherlands; Xpand Biotechnology BV, 3723 MB Bilthoven, Netherlands.
| | - Bas ten Harkel
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute, University of Amsterdam and VU University Amsterdam, 1081 BT Amsterdam, Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute, University of Amsterdam and VU University Amsterdam, 1081 BT Amsterdam, Netherlands
| | - Xiaoman Luo
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, Netherlands; Xpand Biotechnology BV, 3723 MB Bilthoven, Netherlands
| | - Huipin Yuan
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, Netherlands; Xpand Biotechnology BV, 3723 MB Bilthoven, Netherlands
| | - Vincent Everts
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute, University of Amsterdam and VU University Amsterdam, 1081 BT Amsterdam, Netherlands
| | | | - Joost D de Bruijn
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522 NB Enschede, Netherlands; Xpand Biotechnology BV, 3723 MB Bilthoven, Netherlands; School of Engineering and Materials Science (SEMS), Queen Mary University of London, E1 4NS London, United Kingdom
| |
Collapse
|
39
|
Liposomal clodronate inhibition of osteoclastogenesis and osteoinduction by submicrostructured beta-tricalcium phosphate. Biomaterials 2014; 35:5088-97. [PMID: 24698521 DOI: 10.1016/j.biomaterials.2014.03.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/07/2014] [Indexed: 12/27/2022]
Abstract
Bone graft substitutes such as calcium phosphates are subject to the innate inflammatory reaction, which may bear important consequences for bone regeneration. We speculate that the surface architecture of osteoinductive β-tricalcium phosphate (TCP) stimulates the differentiation of invading monocyte/macrophages into osteoclasts, and that these cells may be essential to ectopic bone formation. To test this, porous TCP cubes with either submicron-scale surface architecture known to induce ectopic bone formation (TCPs, positive control) or micron-scale, non-osteoinductive surface architecture (TCPb, negative control) were subcutaneously implanted on the backs of FVB strain mice for 12 weeks. Additional TCPs samples received local, weekly injections of liposome-encapsulated clodronate (TCPs + LipClod) to deplete invading monocyte/macrophages. TCPs induced osteoclast formation, evident by positive tartrate resistant acid phosphatase (TRAP) cytochemical staining and negative macrophage membrane marker F4/80 immunostaining. No TRAP positive cells were found in TCPb or TCPs + LipClod, only F4/80 positive macrophages and foreign body giant cells. TCPs stimulated subcutaneous bone formation in all implants, while no bone could be found in TCPb or TCPs + LipClod. In agreement, expression of bone and osteoclast gene markers was upregulated in TCPs versus both TCPb and TCPs + LipClod, which were equivalent. In summary, submicron-scale surface structure of TCP induced osteoclastogenesis and ectopic bone formation in a process that is blocked by monocyte/macrophage depletion.
Collapse
|
40
|
Klar RM, Duarte R, Dix-Peek T, Ripamonti U. The induction of bone formation by the recombinant human transforming growth factor-β3. Biomaterials 2014; 35:2773-88. [DOI: 10.1016/j.biomaterials.2013.12.062] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/19/2013] [Indexed: 01/22/2023]
|
41
|
Mantripragada VP, Lecka-Czernik B, Ebraheim NA, Jayasuriya AC. An overview of recent advances in designing orthopedic and craniofacial implants. J Biomed Mater Res A 2013; 101:3349-64. [PMID: 23766134 PMCID: PMC4854641 DOI: 10.1002/jbm.a.34605] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/11/2013] [Accepted: 01/11/2013] [Indexed: 01/22/2023]
Abstract
Great deal of research is still going on in the field of orthopedic and craniofacial implant development to resolve various issues being faced by the industry today. Despite several disadvantages of the metallic implants, they continue to be used, primarily because of their superior mechanical properties. In order to minimize the harmful effects of the metallic implants and its by-products, several modifications are being made to these materials, for instance nickel-free stainless steel, cobalt-chromium and titanium alloys are being introduced to eliminate the toxic effects of nickel being released from the alloys, introduce metallic implants with lower modulus, reduce the cost of these alloys by replacing rare elements with less expensive elements etc. New alloys like tantalum, niobium, zirconium, and magnesium are receiving attention given their satisfying mechanical and biological properties. Non-oxide ceramics like silicon nitride and silicon carbide are being currently developed as a promising implant material possessing a combination of properties such as good wear and corrosion resistance, increased ductility, good fracture and creep resistance, and relatively high hardness in comparison to alumina. Polymer/magnesium composites are being developed to improve mechanical properties as well as retain polymer's property of degradation. Recent advances in orthobiologics are proving interesting as well. This paper thus deals with the latest improvements being made to the existing implant materials and includes new materials being introduced in the field of biomaterials.
Collapse
|
42
|
Klar RM, Duarte R, Dix-Peek T, Dickens C, Ferretti C, Ripamonti U. Calcium ions and osteoclastogenesis initiate the induction of bone formation by coral-derived macroporous constructs. J Cell Mol Med 2013; 17:1444-57. [PMID: 24106923 PMCID: PMC4117557 DOI: 10.1111/jcmm.12125] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 08/12/2013] [Indexed: 01/06/2023] Open
Abstract
Coral-derived calcium carbonate/hydroxyapatite macroporous constructs of the genus Goniopora with limited hydrothermal conversion to hydroxyapatite (7% HA/CC) initiate the induction of bone formation. Which are the molecular signals that initiate pattern formation and the induction of bone formation? To evaluate the role of released calcium ions and osteoclastogenesis, 7% HA/CC was pre-loaded with either 500 μg of the calcium channel blocker, verapamil hydrochloride, or 240 μg of the osteoclast inhibitor, biphosphonate zoledronate, and implanted in the rectus abdominis muscle of six adult Chacma baboons Papio ursinus. Generated tissues on days 15, 60 and 90 were analysed by histomorphometry and qRT-PCR. On day 15, up-regulation of type IV collagen characterized all the implanted constructs correlating with vascular invasion. Zoledronate-treated specimens showed an important delay in tissue patterning and morphogenesis with limited bone formation. Osteoclastic inhibition yielded minimal, if any, bone formation by induction. 7% HA/CC pre-loaded with the Ca++ channel blocker verapamil hydrochloride strongly inhibited the induction of bone formation. Down-regulation of bone morphogenetic protein-2 (BMP-2) together with up-regulation of Noggin genes correlated with limited bone formation in 7% HA/CC pre-loaded with either verapamil or zoledronate, indicating that the induction of bone formation by coral-derived macroporous constructs is via the BMPs pathway. The spontaneous induction of bone formation is initiated by a local peak of Ca++ activating stem cell differentiation and the induction of bone formation.
Collapse
Affiliation(s)
- Roland M Klar
- Bone Research Laboratory, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | | | | | | |
Collapse
|
43
|
Guo YJ, Wang YY, Chen T, Wei YT, Chu LF, Guo YP. Hollow carbonated hydroxyapatite microspheres with mesoporous structure: Hydrothermal fabrication and drug delivery property. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:3166-72. [DOI: 10.1016/j.msec.2013.03.040] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 03/03/2013] [Accepted: 03/23/2013] [Indexed: 10/27/2022]
|
44
|
Amini AR, Laurencin CT, Nukavarapu SP. Bone tissue engineering: recent advances and challenges. Crit Rev Biomed Eng 2013; 40:363-408. [PMID: 23339648 DOI: 10.1615/critrevbiomedeng.v40.i5.10] [Citation(s) in RCA: 1364] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The worldwide incidence of bone disorders and conditions has trended steeply upward and is expected to double by 2020, especially in populations where aging is coupled with increased obesity and poor physical activity. Engineered bone tissue has been viewed as a potential alternative to the conventional use of bone grafts, due to their limitless supply and no disease transmission. However, bone tissue engineering practices have not proceeded to clinical practice due to several limitations or challenges. Bone tissue engineering aims to induce new functional bone regeneration via the synergistic combination of biomaterials, cells, and factor therapy. In this review, we discuss the fundamentals of bone tissue engineering, highlighting the current state of this field. Further, we review the recent advances of biomaterial and cell-based research, as well as approaches used to enhance bone regeneration. Specifically, we discuss widely investigated biomaterial scaffolds, micro- and nano-structural properties of these scaffolds, and the incorporation of biomimetic properties and/or growth factors. In addition, we examine various cellular approaches, including the use of mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), adult stem cells, induced pluripotent stem cells (iPSCs), and platelet-rich plasma (PRP), and their clinical application strengths and limitations. We conclude by overviewing the challenges that face the bone tissue engineering field, such as the lack of sufficient vascularization at the defect site, and the research aimed at functional bone tissue engineering. These challenges will drive future research in the field.
Collapse
Affiliation(s)
- Ami R Amini
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|
45
|
Santo VE, Gomes ME, Mano JF, Reis RL. Controlled release strategies for bone, cartilage, and osteochondral engineering--Part II: challenges on the evolution from single to multiple bioactive factor delivery. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:327-52. [PMID: 23249320 DOI: 10.1089/ten.teb.2012.0727] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The development of controlled release systems for the regeneration of bone, cartilage, and osteochondral interface is one of the hot topics in the field of tissue engineering and regenerative medicine. However, the majority of the developed systems consider only the release of a single growth factor, which is a limiting step for the success of the therapy. More recent studies have been focused on the design and tailoring of appropriate combinations of bioactive factors to match the desired goals regarding tissue regeneration. In fact, considering the complexity of extracellular matrix and the diversity of growth factors and cytokines involved in each biological response, it is expected that an appropriate combination of bioactive factors could lead to more successful outcomes in tissue regeneration. In this review, the evolution on the development of dual and multiple bioactive factor release systems for bone, cartilage, and osteochondral interface is overviewed, specifically the relevance of parameters such as dosage and spatiotemporal distribution of bioactive factors. A comprehensive collection of studies focused on the delivery of bioactive factors is also presented while highlighting the increasing impact of platelet-rich plasma as an autologous source of multiple growth factors.
Collapse
Affiliation(s)
- Vítor E Santo
- 3Bs Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
| | | | | | | |
Collapse
|
46
|
Ripamonti U, Teare J, Ferretti C. A Macroporous Bioreactor Super Activated by the Recombinant Human Transforming Growth Factor-β(3). Front Physiol 2012; 3:172. [PMID: 22701102 PMCID: PMC3369251 DOI: 10.3389/fphys.2012.00172] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 05/11/2012] [Indexed: 01/12/2023] Open
Abstract
Macroporous single phase hydroxyapatite (HA) and biphasic HA/β-tricalcium phosphate with 33% post-sinter hydroxyapatite (HA/β-TCP) were combined with 25 or 125 μg recombinant human transforming growth factor-β3 (hTGF-β3) to engineer a super activated bioreactor implanted in orthotopic calvarial and heterotopic rectus abdominis muscle sites and harvested on day 30 and 90. Coral-derived calcium carbonate fully converted (100%) and partially converted to 5 and 13% hydroxyapatite/calcium carbonate (5 and 13% HA/CC) pre-loaded with 125 and 250 μg hTGF-β3, and 1:5 and 5:1 binary applications of hTGF-β3: hOP-1 by weight, were implanted in the rectus abdominis and harvested on day 20 and 30, respectively, to monitor spatial/temporal morphogenesis by high doses of hTGF-β3. Bone formation was assessed on decalcified paraffin-embedded sections by measuring the fractional volume of newly formed bone. On day 30 and 90, single phase HA implants showed greater amounts of bone when compared to biphasic specimens; 5 and 13% HA/CC pre-loaded with 125 and 250 μg hTGF-β3 showed substantial induction of bone formation; 250 μg hTGF-β3 induced as yet unreported massive induction of bone formation as early as 20 days prominently outside the profile of the macroporous constructs. The induction of bone formation is controlled by the implanted ratio of the recombinant morphogens, i.e., the 1:5 hTGF-β3:hOP-1 ratio by weight was greater than the inverse ratio. The unprecedented tissue induction by single doses of 250 μg hTGF-β3 resulting in rapid bone morphogenesis of vast mineralized ossicles with multiple trabeculations surfaced by contiguous secreting osteoblasts is the novel molecular and morphological frontier for the induction of bone formation in clinical contexts.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Unit, Faculty of Health Sciences, School of Physiology, Medical Research Council/University of the Witwatersrand Johannesburg, South Africa
| | | | | |
Collapse
|
47
|
The influence of genetic factors on the osteoinductive potential of calcium phosphate ceramics in mice. Biomaterials 2012; 33:5696-705. [PMID: 22594974 DOI: 10.1016/j.biomaterials.2012.04.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 04/07/2012] [Indexed: 12/20/2022]
Abstract
The efficacy of calcium phosphate (CaP) ceramics in healing large bone defects is, in general, not as high as that of autologous bone grafting. Recently, we reported that CaP ceramics with osteoinductive properties were as efficient in healing an ilium defect of a sheep as autologous bone graft was, which makes this subclass of CaP ceramics a powerful alternative for bone regeneration. Although osteoinduction by CaP ceramics has been shown in several large animal models it is sporadically reported in mice. Because the lack of a robust mouse model has delayed understanding of the mechanism, we screened mice from 11 different inbred mouse strains for their responsiveness to subcutaneous implantation of osteoinductive tricalcium phosphate (TCP). In only two strains (FVB and 129S2) the ceramic induced bone formation, and in particularly, in FVB mice, bone was found in all the tested mice. We also demonstrated that other CaP ceramics induced bone formation at the same magnitude as that observed in other animal models. Furthermore, VEGF did not significantly increase TCP induced bone formation. The mouse model here described can accelerate research of osteoinductive mechanisms triggered by CaP ceramics and potentially the development of therapies for bone regeneration.
Collapse
|
48
|
Deng M, Cushnie EK, Lv Q, Laurencin CT. Poly(lactide-co-glycolide)-Hydroxyapatite Composites: The Development of Osteoinductive Scaffolds for Bone Regenerative Engineering. ACTA ACUST UNITED AC 2012. [DOI: 10.1557/opl.2012.737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ABSTRACTRegenerative engineering represents a new multidisciplinary paradigm to engineer complex tissues, organs, or organ systems through the integration of tissue engineering with advanced materials science, stem cell science and developmental biology. While possessing elements of tissue engineering, regenerative medicine, and morphogenesis, regenerative engineering is distinct from these individual disciplines since it specifically focuses on the integration and subsequent response of stem cells to biomaterials. One goal of regenerative engineering is the design of materials capable of inducing associated cells toward highly specialized functions. For example, the interaction of cells with calcium phosphate surfaces has proven to be an important signaling modality in promoting osteogenic differentiation. A biodegradable polymer-ceramic composite system has been developed from poly(lactide-co-glycolide) and in situ synthesized hydroxyapatite based on the three-dimensional sintered microsphere matrix platform. We have systematically optimized scaffold physico-chemical, mechanical, and structural properties for bone tissue regeneration applications by varying several parameters such as solution pH, polymer:ceramic ratio, sintering time and sintering temperature. The bioactivity of composite scaffolds is attributed to their ability to deliver calcium ions to surrounding medium and allow for reprecipitation of calcium phosphate on the scaffold surface. Furthermore, the composite scaffolds have demonstrated increased loading capacity of osteoinductive growth factor (BMP-2) and a more sustained release profile due to a greater number of adsorption sites provided by the ionic calcium and phosphate groups as well as a larger matrix surface area. In vitro cell studies were performed to investigate the efficacy of this composite system to induce osteogenic differentiation of human adipose-derived stem cells. Cells cultured on the ceramic containing scaffolds exhibited significantly higher expression of osteoblastic markers and greater extracellular matrix mineralization than non-ceramic containing scaffolds, indicating the potential for the ceramic phase to promote osteogenic differentiation. In addition, loaded BMP-2 retained its bioactivity as a mitogen and osteoinductive agent during the differentiation of adipose-derived stem cells into mature osteoblasts. In vivo evaluation using a critical-sized ulnar defect model in New Zealand white rabbits demonstrated the ability of composite scaffolds to support cellular infiltration throughout the scaffold pore structure and vascularization of new tissue, as well as facilitate formation of newly mineralized bone tissue. The work described herein provides strong evidence for the potential of polymer-ceramic composite scaffolds to function as osteoinductive bone graft substitutes, and paves the way for future development of advanced tissue-inducing materials.
Collapse
|
49
|
Ripamonti U, Roden LC, Renton LF. Osteoinductive hydroxyapatite-coated titanium implants. Biomaterials 2012; 33:3813-23. [PMID: 22364700 DOI: 10.1016/j.biomaterials.2012.01.050] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 01/27/2012] [Indexed: 12/20/2022]
Abstract
Previous studies have shown that heterotopic induction of bone formation by calcium phosphate-based macroporous constructs is set into motion by the geometry of the implanted substrata, i.e. a sequence of repetitive concavities assembled within the macroporous spaces. The aim of this study was to construct osteoinductive titanium implants that per se, and without the exogenous application of the osteogenic soluble molecular signals of the transforming growth factor-β supergene family, would initiate the induction of bone formation. To generate intrinsically osteoinductive titanium implants for translation in clinical contexts, titanium grade Ti-6A1-4V cylinders of 15 mm in length and 3.85 mm in diameter, with or without concavities, were plasma sprayed with crystalline hydroxyapatite resulting in a uniform layer of 30 μm in thickness. Before coating, experimental titanium implants were prepared with a sequence of 36 repetitive concavities 1600 μm in diameter and 800 μm in depth, spaced a distance of 1000 μm apart. Mandibular molars and premolars were extracted to prepare edentulous mandibular ridges for later implantation. Planar and geometric hydroxyapatite-coated titanium constructs were implanted in the left and right edentulized hemi-mandibles, respectively, after a healing period of 7-8 months, 3 per hemi-mandible. Three planar and three geometric implants were implanted in the left and right tibiae, respectively; additionally, planar and geometric constructs were also inserted in the rectus abdominis muscle. Six animals were euthanized at 30 and 90 days after implantation; one animal had to be euthanized 5 days after surgery and the remaining animal was euthanized 31 months after implantation. Undecalcified longitudinal sections were precision-sawed, ground and polished to 40-60 μm; all sections were stained with a modified Goldner's trichrome. Undecalcified specimen block preparation was performed using the EXAKT precision cutting and grinding system. Histomorphometric analyses of bone in contact (BIC) showed that on day 30 there was no difference between the geometric vs. planar control implants; on day 90, the ratio of BIC to surface within the geometric implants was greater than on the standard planar implants in both mandibular and tibial sites; 31 months after implantation, selected concavities cut into the geometric implants harvested from the rectus abdominis muscle showed the spontaneous induction of bone formation with mineralized bone surfaced by osteoid seams. These data in non-human primates indicate that geometrically-constructed plasma-sprayed titanium implants are per se osteogenic, the concavities providing a unique microenvironment to initiate bone differentiation by induction.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory, School of Physiology, Medical Research Council/University of the Witwatersrand, Johannesburg, 2193 Parktown, South Africa.
| | | | | |
Collapse
|
50
|
Abstract
The new strategy of tissue engineering, and regenerative medicine at large, is to construct biomimetic matrices to mimic nature's hierarchical structural assemblages and mechanisms of simplicity and elegance that are conserved throughout genera and species. There is a direct spatial and temporal relationship of morphologic and molecular events that emphasize the biomimetism of the remodeling cycles of the osteonic corticocancellous bone versus the "geometric induction of bone formation," that is, the induction of bone by "smart" concavities assembled in biomimetic matrices of macroporous calcium phosphate-based constructs. The basic multicellular unit of the corticocancellous bone excavates a trench across the bone surface, leaving in its wake a hemiosteon rather than an osteon, that is, a trench with cross-sectional geometric cues of concavities after cyclic episodes of osteoclastogenesis, eventually leading to osteogenesis. The concavities per se are geometric regulators of growth-inducing angiogenesis and osteogenesis as in the remodeling processes of the corticocancellous bone. The concavities act as a powerful geometric attractant for myoblastic/myoendothelial and/or endothelial/pericytic stem cells, which differentiate into bone-forming cells. The lacunae, pits, and concavities cut by osteoclastogenesis within the biomimetic matrices are the driving morphogenetic cues that induce bone formation in a continuum of sequential phases of resorption/dissolution and formation. To induce the cascade of bone differentiation, the soluble osteogenic molecular signals of the transforming growth factor β supergene family must be reconstituted with an insoluble signal or substratum that triggers the bone differentiation cascade. By carving a series of repetitive concavities into solid and/or macroporous biomimetic matrices of highly crystalline hydroxyapatite or biphasic hydroxyapatite/β-tricalcium phosphate, we were able to embed smart biologic functions within intelligent scaffolds for tissue engineering of bone. The concavities assembled in the bioceramic constructs biomimetize the remodeling cycle of the corticocancellous bone and are endowed with multifunctional pleiotropic self-assembly capacities, initiating angiogenesis and bone formation by induction without the exogenous applications of the osteogenic-soluble molecular signals of the transforming growth factor β supergene family. The incorporation of specific biologic activities into biomimetic matrices by manipulating the geometry of the substratum, defined as geometric induction of bone formation, is now helping to engineer therapeutic osteogenesis in clinical contexts.
Collapse
|