1
|
Wang Y, Zhao L, Zhou L, Chen C, Chen G. Sequential release of vascular endothelial growth factor-A and bone morphogenetic protein-2 from osteogenic scaffolds assembled by PLGA microcapsules: A preliminary study in vitro. Int J Biol Macromol 2023; 232:123330. [PMID: 36681218 DOI: 10.1016/j.ijbiomac.2023.123330] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/27/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
Bone regeneration is a complex process sequentially regulated by multiple cytokines at different stages. Vascular endothelial growth factor-A (VEGF-A) and bone morphogenetic protein-2 (BMP-2) are the two most important factors involved in this process, and the combination of the two can achieve better bone regeneration by coupling angiogenesis and osteogenesis. In this study, poly(lactic-co-glycolic acid) (PLGA) microspheres with core-shell structure (microcapsules) encapsulating VEGF-A or BMP-2 were prepared by coaxial channel injection and continuous fluid technology. The sequential release of two cytokines by microcapsules with different PLGA molecular weight and shell thickness and its performance in vitro were explored. It was demonstrated that the molecular weight of PLGA significantly affected the degradation and release kinetics of microcapsules, while the thickness of the shell can regulate the release in a finer level. VEGF-A encapsulated microcapsules with low molecular weight can induce vascular endothelial cells to form lumens structures in vitro at an early stage. And BMP-2 encapsulated microcapsules could promote osteogenic differentiation, but the effect could be delayed when the microcapsules were prepared with PLGA of 150 kDa. In conclusion, the core-shell PLGA microcapsules in this study can sequentially release VEGF-A and BMP-2 at different stages to simulate natural bone repair.
Collapse
Affiliation(s)
- Ying Wang
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Lingyan Zhao
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Lvhui Zhou
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Chen Chen
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China.
| | - Gang Chen
- Department of Prosthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu Province Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China.
| |
Collapse
|
2
|
Darjanki CM, Hananta JS, Prahasanti C, Ulfah N, Kusumawardani B, Wijaksana IKE, Aljunaid M, Nkuba A. Expression of VEGF and BMP-2 in Osteoblast cells exposed to a combination of polymethylmethacrylate (PMMA) and hydroxyapatite (HAp). J Oral Biol Craniofac Res 2023; 13:243-248. [PMID: 36818023 PMCID: PMC9930150 DOI: 10.1016/j.jobcr.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/12/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Objectives Polymethylmethacrylate (PMMA) has been widely used, but it has several fallback properties in its interaction with bone tissue, so the addition of hydroxyapatite (HAp) material aims to improve the biocompatibility, regeneration process, and osteointegration of bone implants. The HAp material can be sourced from bovine bone and processed through Good Manufacturing Practice from Tissue Bank (HApGMP), and from limestone (CaCO3) processed by Balai Besar Keramik (HApBBK).This study was to observe the expression of vascular endothelial growth factor (VEGF) and Bone morphogenetic protein-2 (BMP2) in cultured osteoblasts exposed to PMMA-HApGMP and PMMA-HApBBK as implant candidate materials. Methods Sample of PMMA and HAp materials with a mixture of PMMA and HApBBK in the first group and a mixture of PMMA and HApGMP in the second group. Twenty-four fetal rat calvarie osteoblast cell cultures were randomly divided into 6 groups: 7- and 14-day control group, 7 and 14 days PMMA-HApGMP group, 7 and 14 days PMMA-HApBBK group. The expression of VEGF and BMP-2 was seen by immunocytochemical examination. Results The one-way ANOVA with a significance value of 0.000 (p < 0.05). BMP-2 and VEGF expression was increased in the 7- and 14-days groups after exposure to PMMA-HApGMP and PMMA-HApBBK. Conclusion The application of PMMA-HApGMP and PMMA-HApBBK showed an increase in the expression of VEGF and BMP-2 in osteoblast cell cultures which indicates a potential increase in the accelerated angiogenesis and osteogenesis in the bone regeneration process of bone implants.
Collapse
Affiliation(s)
| | | | - Chiquita Prahasanti
- Department of Periodontology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Noer Ulfah
- Department of Periodontology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Banun Kusumawardani
- Department of Biomedical Sciences, Faculty of Dentistry, University of Jember, Surabaya, Indonesia
| | - I Komang Evan Wijaksana
- Department of Periodontology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Mohammed Aljunaid
- Doctoral Program of Dental Medicine, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Dental and Oral Medicine, Faculty of Medicine, Taiz University, Taiz, Yemen
| | - Anord Nkuba
- Aqua Farms Organization. Sinza, Dar Es Salaam, Tanzania
- European Marine Biological Resource, Ghent University, Belgium
| |
Collapse
|
3
|
Quantitative Real-Time Gene Profiling of Human Alveolar Osteoblasts Using a One-Step System. Methods Mol Biol 2023; 2588:417-427. [PMID: 36418701 DOI: 10.1007/978-1-0716-2780-8_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The use of quantitative real-time reverse transcriptase PCR (qRT2-PCR) for the identification of differentially regulated genes is a powerful technology. The protocol presented here uses qRT2-PCR gene arrays to investigate the regulation of 84 angiogenic related genes in human primary alveolar osteoblasts following treatment with the bisphosphonate, zoledronic acid, and geranylgeraniol (GGOH). GGOH has potential as a therapeutic agent for medication-related osteonecrosis of the jaw, a serious side effect resulting from treatment for metastatic cancer (Zafar S, Coates DE, Cullinan MP, Drummond BK, Milne T, Seymour GJ. J Oral Pathol Med 43:711-721, 2014). The isolation of the primary osteoblast cells follows the methods described by Dillon et al. (Method Mol Biol 816:3-18, 2012) with a new RNA extraction technique described fully. The method highlights the importance of obtaining high-quality RNA which is DNA-free. Relative levels of gene expression are normalized against selected reference genes (HKG) and a number of examples of how fold regulation (2-ΔΔCq) and gene expression level (2-ΔCq) data can be presented are given.
Collapse
|
4
|
High-efficient engineering of osteo-callus organoids for rapid bone regeneration within one month. Biomaterials 2022; 288:121741. [PMID: 36031458 DOI: 10.1016/j.biomaterials.2022.121741] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/19/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023]
Abstract
Large bone defects that cannot form a callus tissue are often faced with long-time recovery. Developmental engineering-based strategies with mesenchymal stem cell (MSC) aggregates have shown enhanced potential for bone regeneration. However, MSC aggregates are different from the physiological callus tissues, which limited the further endogenous osteogenesis. This study aims to achieve engineering of osteo-callus organoids for rapid bone regeneration in cooperation with bone marrow-derived stem cell (BMSC)-loaded hydrogel microspheres (MSs) by digital light-processing (DLP) printing technology and stepwise-induction. The printed MSC-loaded MSs aggregated into osteo-callus organoids after chondrogenic induction and showed much higher chondrogenic efficiency than that of traditional MSC pellets. Moreover, the osteo-callus organoids exhibited stage-specific gene expression pattern that recapitulated endochondral ossification process, as well as a synchronized state of cell proliferation and differentiation, which highly resembled the diverse cell compositions and behaviors of developmentally endochondral ossification. Lastly, the osteo-callus organoids efficiently led to rapid bone regeneration within only 4 weeks in a large bone defect in rabbits which need 2-3 months in previous tissue engineering studies. The findings suggested that in vitro engineering of osteo-callus organoids with developmentally osteogenic properties is a promising strategy for rapid bone defect regeneration and recovery.
Collapse
|
5
|
Kang F, Yi Q, Gu P, Dong Y, Zhang Z, Zhang L, Bai Y. Controlled growth factor delivery system with osteogenic-angiogenic coupling effect for bone regeneration. J Orthop Translat 2022; 31:110-125. [PMID: 34976731 PMCID: PMC8671819 DOI: 10.1016/j.jot.2021.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/01/2021] [Accepted: 11/15/2021] [Indexed: 11/19/2022] Open
Abstract
Objective Bone regeneration involves a coordinated cascade of events that are regulated by several cytokines and growth factors, among which bone morphogenic protein-2 (BMP-2), vascular endothelial growth factor (VEGF) and fibroblast growth factor-2 (FGF-2) play important roles. In this study, we investigated the effects of dual release of the three growth factors on bone regeneration in femur defects. Methods A composite consisting of Gelatin microparticles loaded with VEGF/FGF-2 and poly(lactic-co-glycolic acid)-poly(ethylene glycol)-carboxyl (PLGA-PEG-COOH) microparticles loaded with BMP-2 encapsulated in a nano hydroxyapatite-poly actic-co-glycolic acid (nHA-PLGA) scaffold was prepared for the dual release of the growth factors. Results On the 14th day, decreased release rate of BMP-2 compared with FGF-2 and VEGF was observed. However, after 14 days, compared to FGF-2 and VEGF, BMP-2 showed an increased release rate. Controlled dual release of BMP-2 and VEGF, FGF-2 resulted in a significant osteogenic differentiation of bone mesenchymal stem cells (BMSCs). Moreover, effects of the composite scaffold on functional connection of osteoblast-vascular cells during bone development were evaluated. The synergistic effects of dual delivery of growth factors were shown to promote the expression of VEGF in BMSCs. Increased secretion of VEGF from BMSCs promoted the proliferation and angiogenic differentiation of human umbilical vein endothelial cells (HUVECs) in the co-culture system. At 12 weeks after implantation, blood vessel and bone formation were analyzed by micro-CT and histology. The composite scaffold significantly promoted the formation of blood vessels and new bone in femur defects. Conclusions These findings demonstrate that dual delivery of angiogenic factors and osteogenic factors from Gelatin and PLGA-PEG-COOH microparticles-based composite scaffolds exerted an osteogenic-angiogenic coupling effect on bone regeneration. This approach will inform on the development of appropriate designs of high-performance bioscaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Fei Kang
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, 400038, China
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Qiying Yi
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Gu
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yuhan Dong
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Ziyang Zhang
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Lijuan Zhang
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Bai
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Corresponding author. School of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, PR China.
| |
Collapse
|
6
|
Xie C, Ye J, Liang R, Yao X, Wu X, Koh Y, Wei W, Zhang X, Ouyang H. Advanced Strategies of Biomimetic Tissue-Engineered Grafts for Bone Regeneration. Adv Healthc Mater 2021; 10:e2100408. [PMID: 33949147 DOI: 10.1002/adhm.202100408] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/16/2021] [Indexed: 12/21/2022]
Abstract
The failure to repair critical-sized bone defects often leads to incomplete regeneration or fracture non-union. Tissue-engineered grafts have been recognized as an alternative strategy for bone regeneration due to their potential to repair defects. To design a successful tissue-engineered graft requires the understanding of physicochemical optimization to mimic the composition and structure of native bone, as well as the biological strategies of mimicking the key biological elements during bone regeneration process. This review provides an overview of engineered graft-based strategies focusing on physicochemical properties of materials and graft structure optimization from macroscale to nanoscale to further boost bone regeneration, and it summarizes biological strategies which mainly focus on growth factors following bone regeneration pattern and stem cell-based strategies for more efficient repair. Finally, it discusses the current limitations of existing strategies upon bone repair and highlights a promising strategy for rapid bone regeneration.
Collapse
Affiliation(s)
- Chang Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
| | - Jinchun Ye
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Xudong Yao
- The Fourth Affiliated Hospital Zhejiang University School of Medicine Yiwu 322000 China
| | - Xinyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Yiwen Koh
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Wei Wei
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| | - Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| |
Collapse
|
7
|
Freeman FE, Burdis R, Kelly DJ. Printing New Bones: From Print-and-Implant Devices to Bioprinted Bone Organ Precursors. Trends Mol Med 2021; 27:700-711. [PMID: 34090809 DOI: 10.1016/j.molmed.2021.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022]
Abstract
Regenerating large bone defects remains a significant clinical challenge, motivating increased interest in additive manufacturing and 3D bioprinting to engineer superior bone graft substitutes. 3D bioprinting enables different biomaterials, cell types, and growth factors to be combined to develop patient-specific implants capable of directing functional bone regeneration. Current approaches to bioprinting such implants fall into one of two categories, each with their own advantages and limitations. First are those that can be 3D bioprinted and then directly implanted into the body and second those that require further in vitro culture after bioprinting to engineer more mature tissues prior to implantation. This review covers the key concepts, challenges, and applications of both strategies to regenerate damaged and diseased bone.
Collapse
Affiliation(s)
- Fiona E Freeman
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ross Burdis
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
8
|
Lou Y, Wang H, Ye G, Li Y, Liu C, Yu M, Ying B. Periosteal Tissue Engineering: Current Developments and Perspectives. Adv Healthc Mater 2021; 10:e2100215. [PMID: 33938636 DOI: 10.1002/adhm.202100215] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/18/2021] [Indexed: 12/22/2022]
Abstract
Periosteum, a highly vascularized bilayer connective tissue membrane plays an indispensable role in the repair and regeneration of bone defects. It is involved in blood supply and delivery of progenitor cells and bioactive molecules in the defect area. However, sources of natural periosteum are limited, therefore, there is a need to develop tissue-engineered periosteum (TEP) mimicking the composition, structure, and function of natural periosteum. This review explores TEP construction strategies from the following perspectives: i) different materials for constructing TEP scaffolds; ii) mechanical properties and surface topography in TEP; iii) cell-based strategies for TEP construction; and iv) TEP combined with growth factors. In addition, current challenges and future perspectives for development of TEP are discussed.
Collapse
Affiliation(s)
- Yiting Lou
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Guanchen Ye
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Yongzheng Li
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Chao Liu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Binbin Ying
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| |
Collapse
|
9
|
Lee SS, Kim JH, Jeong J, Kim SHL, Koh RH, Kim I, Bae S, Lee H, Hwang NS. Sequential growth factor releasing double cryogel system for enhanced bone regeneration. Biomaterials 2020; 257:120223. [DOI: 10.1016/j.biomaterials.2020.120223] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/26/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022]
|
10
|
Subbiah R, Cheng A, Ruehle MA, Hettiaratchi MH, Bertassoni LE, Guldberg RE. Effects of controlled dual growth factor delivery on bone regeneration following composite bone-muscle injury. Acta Biomater 2020; 114:63-75. [PMID: 32688092 DOI: 10.1016/j.actbio.2020.07.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
The objective of this study was to investigate the controlled release of two growth factors (BMP-2 and VEGF) as a treatment strategy for bone healing in clinically challenging composite injuries, consisting of a femoral segmental bone defect and volumetric muscle loss. This is the first investigation of dual growth factor delivery in a composite injury model using an injectable delivery system consisting of heparin microparticles and alginate gel. The loading efficiency of growth factors into these biomaterials was found to be >90%, revealing a strong affinity of VEGF and BMP-2 to heparin and alginate. The system could achieve simultaneous or tunable release of VEGF and BMP-2 by varying the loading strategy. Single growth factor delivery (VEGF or BMP-2 alone) significantly enhanced vascular growth in vitro. However, no synergistic effect was observed for dual growth factor (BMP-2 + VEGF) delivery in vitro. Effective bone healing was achieved in all treatment groups (BMP-2, simultaneous or tunable delivery of BMP-2 and VEGF) in the composite injury model. The mechanics of the regenerated bone reached a maximum strength of ~52% of intact bone with tunable delivery of VEGF and BMP-2. Overall, simultaneous or tunable co-delivery of low-dose BMP-2 and VEGF failed to fully restore the mechanics of bone in this injury model. Given the severity of the composite injury, VEGF alone may not be sufficient to establish mature and stable blood vessels when compared with previous studies co-delivering BMP-2+VEGF enhanced bone tissue regeneration. Hence, future studies are warranted to develop an alternative treatment strategy focusing on better control over growth factor dose, spatiotemporal delivery, and additional growth factors to regenerate fully functional bone tissue. STATEMENT OF SIGNIFICANCE: We have developed an injectable delivery system consisting of heparin microparticles and an alginate hydrogel that is capable of delivering multiple growth factors in a tunable manner. We used this delivery system to deliver BMP-2 and VEGF in a rodent model of composite bone-muscle injury that mimics clinical type III open fractures. An advanced treatment strategy is necessary for these injuries in order to avoid the negative side effects of high doses of growth factors and because it has been shown that the addition of a muscle injury in this model attenuates the bone regenerative effect of BMP-2. This is the first study to test the effects of dual growth factor delivery (BMP-2/VEGF) on bone healing in a composite bone-muscle injury model and is expected to open up new directions in protein delivery for regenerative medicine.
Collapse
|
11
|
Dreyer CH, Kjaergaard K, Ding M, Qin L. Vascular endothelial growth factor for in vivo bone formation: A systematic review. J Orthop Translat 2020; 24:46-57. [PMID: 32642428 PMCID: PMC7334443 DOI: 10.1016/j.jot.2020.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/29/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To achieve optimal bone formation one of the most influential parameters has been mentioned to be adequate blood supply. Vascular endothelial growth factor (VEGF) is hereby of particular interest in bone regeneration, because of its primary ability to induce neovascularization and chemokine affection for endothelial cells (EC), and is considered to be the main regulator of vascular formation. However, the growth factor has yet to be implemented in a clinical setting in orthopaedic intervention surgery. We hypothesised that the development of VEGF in vivo for bone formation in the last decade had progressed towards clinical application since the latest systematic review from 2008. OBJECTIVE This systematic review recapped the last 13 years of in vivo bone regeneration using vascular endothelial growth factor (VEGF). METHOD A total of 1374 articles were identified using the PubMed search string (vegf or "vascular endothelial growth factor") and (osteogen∗ or "bone formation" or "bone regeneration"). By 3 selection phases 24 published articles were included by the criteria of being in vivo, using only VEGF for bone formation, published after 2007 and written in English. Articles in vitro, written in different languages than English and older than 2007 was excluded. The most recent systematic review on this subject was published in 2008, with the latest included study from 01 to 11-2007. All included studies were classified based on animal, type of defect, scaffold, control group, type of VEGF, release rate, dosage of VEGF, time of evaluation and results. Each study was evaluated for risk of bias by modified CAMARADES quality assessment for the use in experimental animal studies. The score was calculated by peer review journal publication, use of control group, randomisation of groups, justified VEGF dosage, blinding of results, details on animal model, sample size calculation, comply with ethics and no conflict of interest. RESULTS No clinical trials or human application studies were obtained from our search. Experimentally, 11 articles using solely VEGF for bone formation had a group or a timepoint significantly better than the corresponding control group. 18 articles revealed no significant difference of VEGF compared to the control group and 1 article reported a significant decreased bone growth using VEGF compared to control. CONCLUSION Based on these results no clinical studies have yet been performed. However, indications in the best use of VEGF from experimental studies could be made towards that the optimal release is within the first three weeks, in defect models, with the best effect before eight weeks. Future designs should incorporate this with standardised and reproducible models for verification towards clinical practice. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This systematic review aims to assess the existing literature to focus on methodologies and outcomes that can provide future knowledge regarding the solitary use of VEGF for bone regeneration in a clinical setting.
Collapse
Affiliation(s)
- Chris H. Dreyer
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
- Musculoskeletal Research Laboratory, Department of Orthopaedic Surgery & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
- Acute Medicine, Department of Emergency Medicine, Slagelse Hospital, Slagelse, Denmark
| | - Kristian Kjaergaard
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
| | - Ming Ding
- Orthopaedic Research Laboratory, Department of Orthopaedics & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, 5000, Odense C, Denmark
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedic Surgery & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
12
|
Freeman FE, Pitacco P, van Dommelen LHA, Nulty J, Browe DC, Shin JY, Alsberg E, Kelly DJ. 3D bioprinting spatiotemporally defined patterns of growth factors to tightly control tissue regeneration. SCIENCE ADVANCES 2020; 6:eabb5093. [PMID: 32851179 PMCID: PMC7428335 DOI: 10.1126/sciadv.abb5093] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/02/2020] [Indexed: 05/16/2023]
Abstract
Therapeutic growth factor delivery typically requires supraphysiological dosages, which can cause undesirable off-target effects. The aim of this study was to 3D bioprint implants containing spatiotemporally defined patterns of growth factors optimized for coupled angiogenesis and osteogenesis. Using nanoparticle functionalized bioinks, it was possible to print implants with distinct growth factor patterns and release profiles spanning from days to weeks. The extent of angiogenesis in vivo depended on the spatial presentation of vascular endothelial growth factor (VEGF). Higher levels of vessel invasion were observed in implants containing a spatial gradient of VEGF compared to those homogenously loaded with the same total amount of protein. Printed implants containing a gradient of VEGF, coupled with spatially defined BMP-2 localization and release kinetics, accelerated large bone defect healing with little heterotopic bone formation. This demonstrates the potential of growth factor printing, a putative point of care therapy, for tightly controlled tissue regeneration.
Collapse
Affiliation(s)
- Fiona E. Freeman
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Pierluca Pitacco
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Lieke H. A. van Dommelen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Jessica Nulty
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - David C. Browe
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Jung-Youn Shin
- Departments of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Eben Alsberg
- Departments of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Departments of Biomedical Engineering, Pharmacology, Orthopaedics, and Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Kumabe Y, Fukui T, Takahara S, Kuroiwa Y, Arakura M, Oe K, Oda T, Sawauchi K, Matsushita T, Matsumoto T, Hayashi S, Kuroda R, Niikura T. Percutaneous CO2 Treatment Accelerates Bone Generation During Distraction Osteogenesis in Rabbits. Clin Orthop Relat Res 2020; 478:1922-1935. [PMID: 32732577 PMCID: PMC7371043 DOI: 10.1097/corr.0000000000001288] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 04/14/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Distraction osteogenesis has been broadly used to treat various structural bone deformities and defects. However, prolonged healing time remains a major problem. Various approaches including the use of low-intensity pulsed ultrasound, parathyroid hormone, and bone morphogenetic proteins (BMPs) have been studied to shorten the treatment period with limited success. Our previous studies of rats have reported that the transcutaneous application of CO2 accelerates fracture repair and bone-defect healing in rats by promoting angiogenesis, blood flow, and endochondral ossification. This therapy may also accelerate bone generation during distraction osteogenesis, but, to our knowledge, no study investigating CO2 therapy on distraction osteogenesis has been reported. QUESTIONS/PURPOSES We aimed to investigate the effect of transcutaneous CO2 during distraction osteogenesis in rabbits, which are the most suitable animal as a distraction osteogenesis model for a lengthener in terms of limb size. We asked: Does transcutaneous CO2 during distraction osteogenesis alter (1) radiographic bone density in the distraction gap during healing; (2) callus parameters, including callus bone mineral content, volumetric bone mineral density, and bone volume fraction; (3) the newly formed bone area, cartilage area, and angiogenesis, as well as the expression of interleukin-6 (IL-6), BMP-2, BMP-7, hypoxia-inducible factor (HIF) -1α, and vascular endothelial growth factor (VEGF); and (4) three-point bend biomechanical strength, stiffness, and energy? METHODS Forty 24-week-old female New Zealand white rabbits were used according to a research protocol approved by our institutional ethical committee. A distraction osteogenesis rabbit tibia model was created as previously described. Briefly, an external lengthener was applied to the right tibia, and a transverse osteotomy was performed at the mid-shaft. The osteotomy stumps were connected by adjusting the fixator to make no gap. After a 7-day latency phase, distraction was continued at 1 mm per day for 10 days. Beginning the day after the osteotomy, a 20-minute transcutaneous application of CO2 on the operated leg using a CO2 absorption-enhancing hydrogel was performed five times per week in the CO2 group (n = 20). Sham treatment with air was administered in the control group (n = 20). Animals were euthanized immediately after the distraction period (n = 10), 2 weeks (n = 10), and 4 weeks (n = 20) after completion of distraction. We performed bone density quantification on the plain radiographs to evaluate consolidation in the distraction gap with image analyzing software. Callus parameters were measured with micro-CT to assess callus microstructure. The newly formed bone area and cartilage area were measured histologically with safranin O/fast green staining to assess the progress of ossification. We also performed immunohistochemical staining of endothelial cells with fluorescein-labeled isolectin B4 and examined capillary density to evaluate angiogenesis. Gene expressions in newly generated callus were analyzed by real-time polymerase chain reaction. Biomechanical strength, stiffness, and energy were determined from a three-point bend test to assess the mechanical strength of the callus. RESULTS Radiographs showed higher pixel values in the distracted area in the CO2 group than the control group at Week 4 of the consolidation phase (0.98 ± 0.11 [95% confidence interval 0.89 to 1.06] versus 1.19 ± 0.23 [95% CI 1.05 to 1.34]; p = 0.013). Micro-CT demonstrated that bone volume fraction in the CO2 group was higher than that in the control group at Week 4 (5.56 ± 3.21 % [95% CI 4.32 to 6.12 %] versus 11.90 ± 3.33 % [95% CI 9.63 to 14.25 %]; p = 0.035). There were no differences in any other parameters (that is, callus bone mineral content at Weeks 2 and 4; volumetric bone mineral density at Weeks 2 and 4; bone volume fraction at Week 2). At Week 2, rabbits in the CO2 group had a larger cartilage area compared with those in the control group (2.09 ± 1.34 mm [95% CI 1.26 to 2.92 mm] versus 5.10 ± 3.91 mm [95% CI 2.68 to 7.52 mm]; p = 0.011). More newly formed bone was observed in the CO2 group than the control group at Week 4 (68.31 ± 16.32 mm [95% CI 58.19 to 78.44 mm] versus 96.26 ± 19.37 mm [95% CI 84.25 to 108.26 mm]; p < 0.001). There were no differences in any other parameters (cartilage area at Weeks 0 and 4; newly formed bone area at Weeks 0 and 2). Immunohistochemical isolectin B4 staining showed greater capillary densities in rabbits in the CO2 group than the control group in the distraction area at Week 0 and surrounding tissue at Weeks 0 and 2 (distraction area at Week 0, 286.54 ± 61.55 /mm [95% CI 232.58 to 340.49] versus 410.24 ± 55.29 /mm [95% CI 361.78 to 458.71]; p < 0.001; surrounding tissue at Week 0 395.09 ± 68.16/mm [95% CI 335.34 to 454.83] versus 589.75 ± 174.42/mm [95% CI 436.86 to 742.64]; p = 0.003; at Week 2 271.22 ± 169.42 /mm [95% CI 122.71 to 419.73] versus 508.46 ± 49.06/mm [95% CI 465.45 to 551.47]; p < 0.001 respectively). There was no difference in the distraction area at Week 2. The expressions of BMP -2 at Week 2, HIF1-α at Week 2 and VEGF at Week 0 and 2 were greater in the CO2 group than in the control group (BMP -2 at Week 2 3.84 ± 0.83 fold [95% CI 3.11 to 4.58] versus 7.32 ± 1.63 fold [95% CI 5.88 to 8.75]; p < 0.001; HIF1-α at Week 2, 10.49 ± 2.93 fold [95% CI 7.91 to 13.06] versus 20.74 ± 11.01 fold [95% CI 11.09 to 30.40]; p < 0.001; VEGF at Week 0 4.80 ± 1.56 fold [95% CI 3.43 to 6.18] versus 11.36 ± 4.82 fold [95% CI 7.13 to 15.59]; p < 0.001; at Week 2 31.52 ± 8.26 fold [95% CI 24.27 to 38.76] versus 51.05 ± 15.52 fold [95% CI 37.44 to 64.66]; p = 0.034, respectively). There were no differences in any other parameters (BMP-2 at Week 0 and 4; BMP -7 at Weeks 0, 2 and 4; HIF-1α at Weeks 0 and 4; IL-6 at Weeks 0, 2 and 4; VEGF at Week 4). In the biomechanical assessment, ultimate stress and failure energy were greater in the CO2 group than in the control group at Week 4 (ultimate stress 259.96 ± 74.33 N [95% CI 167.66 to 352.25] versus 422.45 ± 99.32 N [95% CI 299.13 to 545.77]; p < 0.001, failure energy 311.32 ± 99.01 Nmm [95% CI 188.37 to 434.25] versus 954.97 ± 484.39 Nmm [95% CI 353.51 to 1556.42]; p = 0.003, respectively). There was no difference in stiffness (216.77 ± 143.39 N/mm [95% CI 38.73 to 394.81] versus 223.68 ± 122.17 N/mm [95% CI 71.99 to 375.37]; p = 0.92). CONCLUSION Transcutaneous application of CO2 accelerated bone generation in a distraction osteogenesis model of rabbit tibias. As demonstrated in previous studies, CO2 treatment might affect bone regeneration in distraction osteogenesis by promoting angiogenesis, blood flow, and endochondral ossification. CLINICAL RELEVANCE The use of the transcutaneous application of CO2 may open new possibilities for shortening healing time in patients with distraction osteogenesis. However, a deeper insight into the mechanism of CO2 in the local tissue is required before it can be used in future clinical practice.
Collapse
Affiliation(s)
- Yohei Kumabe
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoaki Fukui
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shunsuke Takahara
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu Kuroiwa
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Michio Arakura
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Oe
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takahiro Oda
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichi Sawauchi
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takehiko Matsushita
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takahiro Niikura
- Y. Kumabe, T. Fukui, S. Takahara, Y. Kuroiwa, M. Arakura, K. Oe, T. Oda, K. Sawauchi, T. Matsushita, T. Matsumoto, S. Hayashi, R. Kuroda, T. Niikura, Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
14
|
Dashtimoghadam E, Fahimipour F, Tongas N, Tayebi L. Microfluidic fabrication of microcarriers with sequential delivery of VEGF and BMP-2 for bone regeneration. Sci Rep 2020; 10:11764. [PMID: 32678204 PMCID: PMC7366644 DOI: 10.1038/s41598-020-68221-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Wound instability and poor functional vascularization in bone tissue engineering lead to lack of tissue integration and ultimate failure of engineered grafts. In order to harness the regenerative potential of growth factors and stimulate bone healing, present study aims to design multifunctional cell therapy microcarriers with the capability of sequential delivery of essential growth factors, bone morphogenetic protein 2 (BMP-2) and vascular endothelial growth factor (VEGF). An on-chip double emulsion method was implemented to generate monodisperse VEGF encapsulated microcarriers. Bio-inspired poly(3,4-dihydroxyphenethylamine) (PDA) was then functionalized to the microcarriers surface for BMP-2 conjugation. The microcarriers were seeded with mesenchymal stem cells (MSCs) using a dynamic culture technique for cells expansion. Finally, the microcarriers were incorporated into an injectable alginate-RGD hydrogel laden with endothelial cells (ECs) for further analysis. The DNA and calcium content, as well as ALP activity of the construct were analyzed. The confocal fluorescent microscopy was employed to monitor the MSCs and tunneling structure of ECs. Eventually, the capability of developed microcarriers for bone tissue formation was examined in vivo. Microfluidic platform generated monodisperse VEGF-loaded PLGA microcarriers with size-dependent release patterns. Microcarriers generated with the on-chip technique showed more sustained VEGF release profiles compared to the conventional bulk mixing method. The PDA functionalization of microcarriers surface not only provided immobilization of BMP-2 with prolonged bioavailability, but also enhanced the attachment and proliferation of MSCs. Dynamic culturing of microcarriers showcased their great potential to boost MSCs population required for stem cell therapy of bone defects. ALP activity and calcium content analysis of MSCs-laden microcarriers loaded into injectable hydrogels revealed their capability of tunneling formation, vascular cell growth and osteogenic differentiation. The in vivo histology and real-time polymerase chain reaction analysis revealed that transplantation of MSC-laden microcarriers supports ectopic bone formation in the rat model. The presented approach to design bioactive microcarriers offer sustained sequential delivery of bone ECM chemical cues and offer an ideal stabilized 3D microenvironment for patient-specific cell therapy applications. The proposed methodology is readily expandable to integrate other cells and cytokines in a tuned spatiotemporal manner for personalized regenerative medicine.
Collapse
Affiliation(s)
| | - Farahnaz Fahimipour
- Marquette University School of Dentistry, Milwaukee, WI, USA
- Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nikita Tongas
- Marquette University School of Dentistry, Milwaukee, WI, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA.
| |
Collapse
|
15
|
Skeletal unloading reduces cluster of differentiation (CD) 38 expression in the bone marrow and osteoblasts of mice. J Orthop Sci 2020; 25:331-337. [PMID: 31072650 DOI: 10.1016/j.jos.2019.03.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/05/2019] [Accepted: 03/25/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Mechanical unloading induces bone loss in human weight-loaded bones. The findings of recent studies have revealed that cluster of differentiation 38 knockout mice display bone loss similar to that observed in osteoporosis. This study aimed to determine whether the expression of cluster of differentiation 38 is implicated in skeletal unloading and reloading. METHODS Eight-week-old male C57BL/6J mice were assigned to control, tail-suspension, or reloading after tail-suspension groups. In the tail-suspension group, tail suspension elevated the hind limbs for 1 week. The bilateral femurs and tibias from the groups were evaluated for cluster of differentiation 38 immunocytochemistry, and the cluster of differentiation 38 messenger ribonucleic acid levels and the expression of cluster of differentiation 38 and other cell-surface antigens were evaluated using quantitative real-time polymerase chain reaction and flow cytometric analyses. RESULTS In the tail-suspension group, the alkaline phosphatase reactivity, cluster of differentiation 38 immunoreactivity in the bone marrow and osteoblasts, and the expression of cluster of differentiation 38 messenger ribonucleic acid and that of other cell-surface antigens were significantly lower than those in the control group. In the reloading after tail-suspension group, the level of cluster of differentiation 38 expression was restored to the same level as that in the control group. CONCLUSIONS Cluster of differentiation 38 expression declined after skeletal unloading and recovered to normal levels after reloading. In the bone marrow, cluster of differentiation 38 expression plays a crucial role in bone formation in response to mechanical stress.
Collapse
|
16
|
Casanova MR, Oliveira C, Fernandes EM, Reis RL, Silva TH, Martins A, Neves NM. Spatial immobilization of endogenous growth factors to control vascularization in bone tissue engineering. Biomater Sci 2020; 8:2577-2589. [DOI: 10.1039/d0bm00087f] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
An engineered biofunctional system comprises endogenous BMP-2 and VEGF bound in a parallel pattern. It successfully enabled obtaining the spatial osteogenic and angiogenic differentiation of human hBM-MSCs under basal culture conditions.
Collapse
Affiliation(s)
- Marta R. Casanova
- 3B's Research Group
- I3Bs – Research Institute on Biomaterials
- Biodegradables and Biomimetics of University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco/Guimarães
| | - Catarina Oliveira
- 3B's Research Group
- I3Bs – Research Institute on Biomaterials
- Biodegradables and Biomimetics of University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco/Guimarães
| | - Emanuel M. Fernandes
- 3B's Research Group
- I3Bs – Research Institute on Biomaterials
- Biodegradables and Biomimetics of University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco/Guimarães
| | - Rui L. Reis
- 3B's Research Group
- I3Bs – Research Institute on Biomaterials
- Biodegradables and Biomimetics of University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco/Guimarães
| | - Tiago H. Silva
- 3B's Research Group
- I3Bs – Research Institute on Biomaterials
- Biodegradables and Biomimetics of University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco/Guimarães
| | - Albino Martins
- 3B's Research Group
- I3Bs – Research Institute on Biomaterials
- Biodegradables and Biomimetics of University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco/Guimarães
| | - Nuno M. Neves
- 3B's Research Group
- I3Bs – Research Institute on Biomaterials
- Biodegradables and Biomimetics of University of Minho
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- 4805-017 Barco/Guimarães
| |
Collapse
|
17
|
Junka R, Quevada K, Yu X. Acellular polycaprolactone scaffolds laden with fibroblast/endothelial cell-derived extracellular matrix for bone regeneration. J Biomed Mater Res A 2019; 108:351-364. [PMID: 31618528 DOI: 10.1002/jbm.a.36821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 10/03/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022]
Abstract
Inconsistencies in graft osteoconduction and osteoinduction present a clinical challenge in regeneration of large bone defects. Deposition of decellularized extracellular matrix (dECM) on tissue engineered scaffolds offers an alternative approach that can enhance these properties by mimicking bone's molecular complexity and direct infiltrating cells to repair damaged bone. However, dECMs derived from homogenous cell populations do not adequately simulate the heterogeneous and vascularized microenvironment of the bone. In this study, successive culture and decellularization of fibroblasts and endothelial cells (ECs) grown on polycaprolactone microfibers was used to develop a bioactive scaffold with heterogeneous dECM mimicking endothelial basement membrane. These scaffolds had greater amount of protein and minimally increased nucleic acid content than scaffolds with homogenous culture dECM. Coomassie Blue and antibody staining revealed extensive tube formation by ECs on fibroblast dECM. Fibroblast/endothelial dECM significantly enhanced osteoblast attachment, alkaline phosphatase activity, and osteocalcin- and osteopontin-positive extracellular mineral deposits. We demonstrated that the osteoconduction of dECMs can be tailored with the appropriate combination of cells to accelerate osteoblast mineral secretion. The overall concept can be expanded to generate increasingly more complex tissue constructs for regeneration of bone defects and other vascularized tissues.
Collapse
Affiliation(s)
- Radoslaw Junka
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Kristian Quevada
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Xiaojun Yu
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| |
Collapse
|
18
|
Gaihre B, Unagolla JM, Liu J, Ebraheim NA, Jayasuriya AC. Thermoresponsive Injectable Microparticle-Gel Composites with Recombinant BMP-9 and VEGF Enhance Bone Formation in Rats. ACS Biomater Sci Eng 2019; 5:4587-4600. [PMID: 33448832 PMCID: PMC10742348 DOI: 10.1021/acsbiomaterials.9b00082] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone morphogenetic protein-9 (BMP-9) has been shown to be the most osteogenic BMP. Most of these experiments, however, involve an adenovirus-transfection strategy. Here, we used the scaffold-based strategy to study the bone forming ability of recombinant BMP-9 combined with vascular endothelial growth factor (VEGF). A robust, injectable, multicomponent-releasing scaffold in the form of a composite gel was developed by combining chitosan microparticles (MPs) with thermosensitive gel (MPs-gel). The MPs acted as the carriers for BMP-9 and the gel was loaded with VEGF. The developed gel consisted of hydrophobic chains of methyl cellulose (MC) and the cross-linked structures of alginate (Alg) and calcium. Gelation was achieved at physiological temperature and thus facilitated the injection and localization of MPs enabling an increased efficacy of incorporated growth factors at the target site. A release profile of incorporated growth factors over a two-week period showed higher release of VEGF at each time point compared to that of BMP-9. Human mesenchymal stem cells (hMSCs) encapsulated within the MPs-gel maintained their viability. BMP-9 enhanced the proliferation of hMSCs along the surface of MPs. Furthermore, BMP-9 potently induced the osteogenic differentiation of encapsulated hMSCs elucidated by the increased alkaline phosphatase (ALP) activity and the higher expression of ALP, collagen 1, and osteocalcin genes. In addition, in vivo experiments demonstrated that MPs-gel with the combination of BMP-9-VEGF could significantly enhance both subcutaneous and cranial bone formation (p < 0.05). Taken together, the results here strongly suggest that BMP-9-VEGF incorporated MPs-gel holds promise as an injectable bone tissue engineering platform.
Collapse
Affiliation(s)
- Bipin Gaihre
- Department of Bioengineering, The University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Janitha M. Unagolla
- Department of Bioengineering, The University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Jiayong Liu
- Department of Orthopaedic Surgery, University of Toledo Medical Center, 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| | - Nabil A. Ebraheim
- Department of Orthopaedic Surgery, University of Toledo Medical Center, 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| | - Ambalangodage C. Jayasuriya
- Department of Bioengineering, The University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
- Department of Orthopaedic Surgery, University of Toledo Medical Center, 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| |
Collapse
|
19
|
Sharma A, Desando G, Petretta M, Chawla S, Bartolotti I, Manferdini C, Paolella F, Gabusi E, Trucco D, Ghosh S, Lisignoli G. Investigating the Role of Sustained Calcium Release in Silk-Gelatin-Based Three-Dimensional Bioprinted Constructs for Enhancing the Osteogenic Differentiation of Human Bone Marrow Derived Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2019; 5:1518-1533. [DOI: 10.1021/acsbiomaterials.8b01631] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Aarushi Sharma
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology, New Delhi 110016, India
| | - Giovanna Desando
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio RAMSES, Bologna 40136, Italy
| | - Mauro Petretta
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio RAMSES, Bologna 40136, Italy
- RegenHu Ltd, Villaz St. Pierre CH-1690, Switzerland
| | - Shikha Chawla
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology, New Delhi 110016, India
| | | | - Cristina Manferdini
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Francesca Paolella
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Elena Gabusi
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Diego Trucco
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology, New Delhi 110016, India
| | - Gina Lisignoli
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| |
Collapse
|
20
|
Sequential releasing of VEGF and BMP-2 in hydroxyapatite collagen scaffolds for bone tissue engineering: Design and characterization. Int J Biol Macromol 2019; 123:622-628. [DOI: 10.1016/j.ijbiomac.2018.11.099] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/27/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
|
21
|
Wagner ER, Parry J, Dadsetan M, Bravo D, Riester SM, Van Wijnen AJ, Yaszemski MJ, Kakar S. VEGF-mediated angiogenesis and vascularization of a fumarate-crosslinked polycaprolactone (PCLF) scaffold. Connect Tissue Res 2018. [PMID: 29513041 DOI: 10.1080/03008207.2018.1424145] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Revascularization of natural and synthetic scaffolds is a critical part of the scaffold's incorporation and tissue ingrowth. Our goals were to create a biocompatible polymer scaffold with 3D-printing technology, capable of sustaining vascularization and tissue ingrowth. METHODS We synthesized biodegradable polycaprolactone fumarate (PCLF) scaffolds to allow tissue ingrowth via large interconnected pores. The scaffolds were prepared with Poly(lactic-co-glycolic acid)(PLGA) microspheres seeded with or without different growth factors including VEGF,FGF-2, and/or BMP-2. Scaffolds were implanted into the subcutaneous tissues of rats before undergoing histologic and microCT angiographic analysis. RESULTS At harvest after 12 weeks, scaffolds had tissue infiltrating into their pores without signs of scar tissue formation, fibrous capsule formation, or immune responses against PCLF. Histology for M1/M2 macrophage phenotypes confirmed that there were no overt signs of immune responses. Both microCT angiography and histologic analysis demonstrated marked tissue and vessel ingrowth throughout the pores traversing the body of the scaffolds. Scaffolds seeded with microspheres containing VEGF or VEGF with either BMP-2 or FGF-2 had significantly higher vascular ingrowth and vessel penetration than controls. All VEGF-augmented scaffolds were positive for Factor-VIII and exhibited collagen tissue infiltration throughout the pores. Furthermore, scaffolds with VEGF and BMP-2 had high levels of mineral deposition throughout the scaffold that are attributable to BMP-2. CONCLUSIONS PCLF polymer scaffold can be utilized as a framework for vascular ingrowth and regeneration of multiple types of tissues. This novel scaffold material has promise in tissue regeneration across all types of tissues from soft tissue to bone.
Collapse
Affiliation(s)
- Eric R Wagner
- a Mayo Clinic , Department of Orthopedic Surgery , Rochester , MN
| | - Joshua Parry
- a Mayo Clinic , Department of Orthopedic Surgery , Rochester , MN
| | - Mahrokh Dadsetan
- a Mayo Clinic , Department of Orthopedic Surgery , Rochester , MN
| | - Dalibel Bravo
- a Mayo Clinic , Department of Orthopedic Surgery , Rochester , MN
| | - Scott M Riester
- a Mayo Clinic , Department of Orthopedic Surgery , Rochester , MN
| | | | | | - Sanjeev Kakar
- a Mayo Clinic , Department of Orthopedic Surgery , Rochester , MN
| |
Collapse
|
22
|
Cao L, Kong X, Lin S, Zhang S, Wang J, Liu C, Jiang X. Synergistic effects of dual growth factor delivery from composite hydrogels incorporating 2-N,6-O-sulphated chitosan on bone regeneration. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1-S17. [PMID: 30231646 DOI: 10.1080/21691401.2018.1488721] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A promising strategy to accelerate bone generation is to deliver a combination of certain growth factors to the integration site via a controlled spatial and temporal delivery mode. Here, a composite hydrogel incorporating poly(lactide-co-glycolide) (PLGA) microspheres was accordingly prepared to load and deliver the osteogenic rhBMP-2 and angiogenic rhVEGF165 in the required manner. In addition, 2-N,6-O-sulphated chitosan (26SCS), which is a synergetic factor of growth factors, was incorporated in the composite hydrogel as well. The system showed a similar release behaviour of the two growth factors regardless of 26SCS inclusion. RhBMP-2 loaded in PLGA microspheres showed a sustained release over a period of 2 weeks, whereas rhVEGF165 loaded in hydrogel eluted almost completely from the hydrogel over the first 16 days. Both growth factors retained their efficacy, as quantified with relevant in vitro assays. Moreover, an enhanced cell response was achieved upon the delivery of dual growth factors, compared to that obtained with a single factor. Furthermore, in the presence of 26SCS, the system revealed significantly upregulated alkaline phosphatase activity, human umbilical vein endothelial cell proliferation, sprouting, nitric oxide secretion, and angiogenic gene expression. This study highlighted that the composite hydrogel incorporated with 26SCS appears to constitute a promising approach to deliver multiple growth factors. From our findings, we could also conclude that rhBMP-2 can promote angiogenesis and that the mechanism is worthy of further study in subsequent research.
Collapse
Affiliation(s)
- Lingyan Cao
- a Department of Prosthodontics , Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, PR China.,b Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , National Clinical Research Center of Stomatology , Shanghai , PR China
| | - Xiangjun Kong
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Shuxian Lin
- a Department of Prosthodontics , Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, PR China.,b Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , National Clinical Research Center of Stomatology , Shanghai , PR China
| | - Shuang Zhang
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Jing Wang
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Changsheng Liu
- c Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China.,d Key Laboratory for Ultrafine Materials of Ministry of Education , East China University of Science and Technology , Shanghai , PR China
| | - Xinquan Jiang
- a Department of Prosthodontics , Ninth People's Hospital, Shanghai JiaoTong University School of Medicine , Shanghai, PR China.,b Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , National Clinical Research Center of Stomatology , Shanghai , PR China
| |
Collapse
|
23
|
Inflammatory-Driven Angiogenesis in Bone Augmentation with Bovine Hydroxyapatite, B-Tricalcium Phosphate, and Bioglasses: A Comparative Study. J Immunol Res 2018; 2018:9349207. [PMID: 30298138 PMCID: PMC6157209 DOI: 10.1155/2018/9349207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/12/2018] [Accepted: 07/25/2018] [Indexed: 01/30/2023] Open
Abstract
Introduction The clinical use of bioactive materials for bone augmentation has remained a challenge because of predictability and effectiveness concerns, as well as increased costs. The purpose of this study was to analyse the ability to integrate bone substitutes by evaluating the immunohistochemical expression of the platelet endothelial cell adhesion molecules, vascular endothelial growth factor, collagen IV, laminin, and osteonectin, in the vicinity of bone grafts, enabling tissue revascularization and appearance of bone lamellae. There is a lack of in vivo studies of inflammatory-driven angiogenesis in bone engineering using various grafts. Methods The study was performed in animal experimental model on the standardized monocortical defects in the tibia of 20 New Zealand rabbits. The defects were augmented with three types of bone substituents. The used bone substituents were beta-tricalcium phosphate, bovine hydroxyapatite, and bioactive glasses. After a period of 6 months, bone fragments were harvested for histopathologic examination. Endothelial cell analysis was done by analysing vascularization with PECAM/CD31 and VEGF and fibrosis with collagen IV, laminin, and osteonectin stains. Statistical analysis was realized by descriptive analysis which was completed with the kurtosis and skewness as well as the Kruskal-Wallis and Mann-Whitney statistical tests. Results The discoveries show that the amount of bone that is formed around beta-tricalcium phosphate and bovine hydroxyapatite is clearly superior to the bioactive glasses. Both the lumen diameter and the number of vessels were slightly increased in favor of beta-tricalcium phosphate. Conclusion We can conclude that bone substitutes as bovine bone and beta-tricalcium phosphate have significant increased angiogenesis (and subsequent improved osteogenesis) compared to the bioactive glass. In our study, significant angiogenesis is linked with a greater tissue formation, indicating that in bone engineering with the allografts we used, inflammation has more benefic effects, the catabolic action being exceeded by the tissue formation.
Collapse
|
24
|
Abstract
The use of quantitative real-time reverse transcriptase PCR (qRT2-PCR) for the identification of differentially regulated genes is a powerful technology. The protocol presented here uses qRT2-PCR gene arrays to investigate the regulation of 84 angiogenic related genes in human primary alveolar osteoblasts following treatment with the bisphosphonate, zoledronic acid (ZA), and geranylgeraniol (GGOH). GGOH has potential as a therapeutic agent for Bisphosphate-Related Osteonecrosis of the Jaw (BRONJ), a serious side-effect resulting from the treatment for metastatic cancer (Zafar et al., J Oral Pathol Med 43:711-721, 2014; Ruggiero, Ann NY Acad Sci 1218:38-46, 2011). The isolation of the primary osteoblast cells follows the methods previously described (Dillon et al., Methods Mol Biol 816:3-18, 2012) with a new RNA extraction technique described fully. The method highlights the importance of obtaining high-quality RNA which is DNA-free. Relative levels of gene expression are normalized against selected housekeeping genes (HKG) and a number of examples of how fold regulation (2-∆∆Cq) and gene expression level (2-∆Cq) data can be presented are given.
Collapse
Affiliation(s)
- Dawn E Coates
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, 310 Great King Street, Dunedin, 9016, New Zealand.
| | - Sobia Zafar
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, 310 Great King Street, Dunedin, 9016, New Zealand
| | - Trudy J Milne
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, 310 Great King Street, Dunedin, 9016, New Zealand
| |
Collapse
|
25
|
Translating the role of osteogenic-angiogenic coupling in bone formation: Highly efficient chitosan-pDNA activated scaffolds can accelerate bone regeneration in critical-sized bone defects. Biomaterials 2017; 149:116-127. [DOI: 10.1016/j.biomaterials.2017.09.036] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/12/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
|
26
|
Seo DU, Kim SG, Oh JS, Lim SC. Comparative Study on Early Osseointegration of Implants According to Various Drilling Speeds in the Mandible of Dogs. IMPLANT DENT 2017; 26:841-847. [PMID: 29068799 DOI: 10.1097/id.0000000000000673] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE This study evaluated the effect of drilling speed on early bone healing in the mandible of dogs. MATERIAL AND METHODS Six dogs were selected, and mandibular premolars and molars were extracted. After 2 months, 3 hydroxyapatite-surfaced fixtures were implanted with drilling speeds of 50, 800, and 1200 rpm on the right side first and then on the left side after 2 weeks. Implant stability quotient (ISQ) was measured on insertion, after 2 and 4 weeks. RESULTS Based on the ISQ measurement, the 1200-rpm group showed a higher value than the 50-rpm group at 2 weeks and 4 weeks (P < 0.05). New bone formation around the implant was highest for the 800-rpm group at 2 weeks and the 1200-rpm group at 4 weeks. The bone-implant contact of the superior half of the alveolar bone was highest for the 800-rpm group at 2 weeks and the 1200-rpm group at 4 weeks. There was no statistically significant difference. CONCLUSION This study suggests that 50, 800, and 1200 rpm are drilling speeds which can expect favorable outcome, yet, higher drilling speed presented overall the best biological responses.
Collapse
Affiliation(s)
- Dong-Uk Seo
- Graduate Student, Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju, Korea
| | - Su-Gwan Kim
- Professor, Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju, Republic of Korea
| | - Ji-Su Oh
- Associate Professor, Department of Oral and Maxillofacial Surgery, School of Dentistry, Chosun University, Gwangju, Korea
| | - Sung-Chul Lim
- Professor, Department of Pathology, School of Medicine, Chosun University, Gwangju, Korea
| |
Collapse
|
27
|
Bai Y, Bai L, Zhou J, Chen H, Zhang L. Sequential delivery of VEGF, FGF-2 and PDGF from the polymeric system enhance HUVECs angiogenesis in vitro and CAM angiogenesis. Cell Immunol 2017; 323:19-32. [PMID: 29111157 DOI: 10.1016/j.cellimm.2017.10.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/17/2017] [Accepted: 10/21/2017] [Indexed: 12/17/2022]
Abstract
Angiogenesis is an organized series of events, beginning with vessel destabilization, followed by endothelial cell re-organization, and ending with vessel maturation. The formation of a mature vascular network requires precise spatial and temporal regulation of a large number of angiogenic factors, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor-2 (FGF-2) and platelet-derived growth factor (PDGF). VEGF aids in vascular permeability and endothelial cell recruitment, FGF-2 activates endothelial cell proliferation and migration while PDGF stimulates vascular stability. Accordingly, VEGF may inhibit vessel stabilization while PDGF may inhibit endothelial cell recruitment. Therefore, a new polymeric system was prepared by the supercritical carbon dioxide foaming technology, which realized sequential delivery of two or more growth factors with the controlled dose and rate. Increased release of VEGF (71.10%) and FGF-2 (69.76%) compared to PDGF (43.17%) was observed for the first 7 days. Thereafter, up till 21 days, an increased rate of release of BMP-2 compared to VEGF 165 was observed. The effects of PDGF-PLAms/VEGF-FGF-2-PLGA scaffolds on angiogenesis were investigated by human umbilical vein endothelial cells (HUVECs) angiogenic differentiation in vitro and chorioallantoic membrane (CAM) angiogenesis in vivo. Sequential delivery of VEGF, FGF-2 and PDGF from structural polymer scaffolds with distinct kinetics resulted in significant angiogenic differentiation of HUVECs and rapid formation of mature vascular networks in chorioallantoic membrane. This study reported a composite scaffold with distinct release kinetics, and these results clearly indicated the importance of sequential delivery of multiple growth factors in tissue regeneration and engineering.
Collapse
Affiliation(s)
- Yan Bai
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China; Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, PR China.
| | - Lijuan Bai
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Zhou
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Huali Chen
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Liangke Zhang
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
28
|
Wang Q, Zhang Y, Li B, Chen L. Controlled dual delivery of low doses of BMP-2 and VEGF in a silk fibroin-nanohydroxyapatite scaffold for vascularized bone regeneration. J Mater Chem B 2017; 5:6963-6972. [PMID: 32264345 DOI: 10.1039/c7tb00949f] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The controlled co-release of osteoinductive and angiogenic factors is an efficient approach to promote vascularized bone regeneration, and a suitable controlled release system can largely reduce the usage of these factors to avoid cost and safety problems. In this study, a cell-free vascularized bone tissue engineering system based on a silk fibroin (SF)/nanohydroxyapatite (nHAp) scaffold was developed, in which very low doses of osteoinductive and angiogenic factors, bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF), were embedded and released in a controlled manner to facilitate bone formation and vascularization, respectively. BMP-2 and VEGF were adsorbed onto SF microspheres (diameter of 1.5 ± 0.3 μm) that were prepared using a co-flow capillary device, and these microspheres were subsequently incorporated within the SF/nHAp scaffolds to provide controlled release. BMP-2 and VEGF were incorporated into SF microspheres via chemical covalent bonding and physical adsorption, respectively, leading to their controlled and sustained release from the SF/nHAp scaffolds. The rapid initial release of VEGF mimicked its expression at the early bone healing stage and promoted angiogenesis, and the relatively slow and sustained release of BMP-2 facilitated osteogenic differentiation both in vitro and in vivo, and the bone completely bridged the rat calvarial defects after 12 weeks of implantation. Overall, our findings suggest that the controlled dual release of very low doses of BMP-2 (300 ng per scaffold) and VEGF (20 ng per scaffold) from SF/nHAp scaffolds results in a synergistic effect on vascularized bone regeneration; this controlled release system can largely reduce the usage of BMP-2 as compared to other systems.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China.
| | | | | | | |
Collapse
|
29
|
Liang C, Lin X, Wang SL, Guo LH, Wang XY, Li J. Osteogenic potential of three different autogenous bone particles harvested during implant surgery. Oral Dis 2017. [PMID: 28644543 DOI: 10.1111/odi.12704] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The aim was to compare the osteoblast activity and osteogenic potential of autogenous bone particles harvested using three different techniques and determine the most advantageous method of collecting autogenous bone particles. SUBJECTS AND METHODS Bone particles were harvested from 20 patients during dental implant surgery using bone scraping, low-speed drilling and bone trap filtering. After the osteoblasts were cultured, cell proliferation, migration, mineralization, transcription of osteogenesis-related genes, secretion of osteogenesis-related proteins and osteoinductive protein content in the bone particle matrix were evaluated. RESULTS Osteoblast activity and osteogenic potential were higher in bone samples harvested by scraper or low-speed drilling than in samples harvested by bone trap filter. Although these parameters were slightly lower in the low-speed drilling group than in the scraper group, significant differences were found only in bone Gla protein levels. However, the levels of osteoinductive proteins in the bone particle matrix were significantly higher in the low-speed drilling group than in the scraper group. CONCLUSIONS Low-speed drilling is a recommendable and effective technique for collecting autogenous bone particles. In implant operations, low-speed drilling can be considered the first-line option, and if the quantity of harvested bone is insufficient, bone shavings obtained by the scraper may be considered.
Collapse
Affiliation(s)
- C Liang
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - X Lin
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - S-L Wang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - L-H Guo
- Department of Stomatology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - X-Y Wang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - J Li
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Synergistic Effects of Vascular Endothelial Growth Factor on Bone Morphogenetic Proteins Induced Bone Formation In Vivo: Influencing Factors and Future Research Directions. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2869572. [PMID: 28070506 PMCID: PMC5187461 DOI: 10.1155/2016/2869572] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/16/2016] [Accepted: 10/24/2016] [Indexed: 02/08/2023]
Abstract
Vascular endothelial growth factor (VEGF) and bone morphogenetic proteins (BMPs), as key mediators in angiogenesis and osteogenesis, are used in a combined delivery manner as a novel strategy in bone tissue engineering. VEGF has the potential to enhance BMPs induced bone formation. Both gene delivery and material-based delivery systems were incorporated in previous studies to investigate the synergistic effects of VEGF and BMPs. However, their results were controversial due to variation of methods incorporated in different studies. Factors influencing the synergistic effects of VEGF on BMPs induced bone formation were identified and analyzed in this review to reduce confusion on this issue. The potential mechanisms and directions of future studies were also proposed here. Further investigating mechanisms of the synergistic effects and optimizing these influencing factors will help to generate more effective bone regeneration.
Collapse
|
31
|
Fu X, Tan J, Sun CG, Leng HJ, Xu YS, Song CL. Intraosseous Injection of Simvastatin in Poloxamer 407 Hydrogel Improves Pedicle-Screw Fixation in Ovariectomized Minipigs. J Bone Joint Surg Am 2016; 98:1924-1932. [PMID: 27852910 DOI: 10.2106/jbjs.15.00937] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Osteoporosis leads to poor osseointegration and reduces implant stability. Statins have been found to stimulate bone formation, but the bioavailability from oral administration is low. Local application may be more effective at augmenting bone formation and enhancing implant stability. This study was performed to evaluate the efficacy of an intraosseous injection of simvastatin in thermosensitive poloxamer 407 hydrogel to enhance pedicle-screw fixation in calcium-restricted ovariectomized minipigs. METHODS Nine mature female Guangxi Bama minipigs underwent bilateral ovariectomy and were fed a calcium-restricted diet for 18 months. Simvastatin (0, 0.5, or 1 mg) in thermosensitive poloxamer 407 hydrogel was injected into the lumbar vertebrae (L4-L6), and titanium alloy pedicle screws were implanted. Bone mineral density (BMD) measurements of the lumbar vertebrae were determined by dual x-ray absorptiometry (DXA) before and 3 months after treatment. The lumbar vertebrae were harvested and analyzed with use of microcomputed tomography, biomechanical pull-out testing, histological analysis, and Western blot analysis for bone morphogenetic protein (BMP)-2 and vascular endothelial growth factor (VEGF) expression. RESULTS Evaluation over a 3-month study period demonstrated that the BMD of the vertebrae injected with 0.5 and 1.0 mg of simvastatin had increased by 31.25% and 31.09%, respectively, compared with vehicle-only injection (p ≤ 0.00014 for both) and increased by 32.12% and 28.16%, respectively, compared with the pre-treatment levels (p < 0.0001 for both). A single injection of simvastatin in poloxamer 407 increased trabecular volume fraction, thickness, and number and decreased trabecular separation (p ≤ 0.002). The bone formation and mineral apposition rates significantly increased (p ≤ 0.023). The percentage of osseointegration in the simvastatin 0.5 and 1-mg groups was 46.54% and 42.63% greater, respectively, than that in the vehicle-only group (p ≤ 0.006), and the maximum pull-out strength was 45.75% and 51.53% greater, respectively, than in the vehicle-only group (p ≤ 0.0005). BMP-2 and VEGF expressions were higher than for the vehicle-only injection. CONCLUSIONS A single intraosseous injection of simvastatin in thermosensitive poloxamer 407 hydrogel stimulated bone formation, increased BMD, improved bone microstructure, promoted osseointegration, and significantly enhanced the stability of pedicle screws in calcium-restricted ovariectomized minipigs. CLINICAL RELEVANCE These results provide rationale for evaluating intraosseous injection of simvastatin in poloxamer 407 to enhance implant fixation in patients with osteoporosis.
Collapse
Affiliation(s)
- X Fu
- Departments of Orthopedics (X.F., J.T., C.G.S., H.J.L., and C.L.S.) and Neurology (Y.S.X.), Peking University Third Hospital, Beijing, People's Republic of China
| | - J Tan
- Departments of Orthopedics (X.F., J.T., C.G.S., H.J.L., and C.L.S.) and Neurology (Y.S.X.), Peking University Third Hospital, Beijing, People's Republic of China
| | - C G Sun
- Departments of Orthopedics (X.F., J.T., C.G.S., H.J.L., and C.L.S.) and Neurology (Y.S.X.), Peking University Third Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Spinal Diseases, Beijing, People's Republic of China
| | - H J Leng
- Departments of Orthopedics (X.F., J.T., C.G.S., H.J.L., and C.L.S.) and Neurology (Y.S.X.), Peking University Third Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Spinal Diseases, Beijing, People's Republic of China
| | - Y S Xu
- Departments of Orthopedics (X.F., J.T., C.G.S., H.J.L., and C.L.S.) and Neurology (Y.S.X.), Peking University Third Hospital, Beijing, People's Republic of China
| | - C L Song
- Departments of Orthopedics (X.F., J.T., C.G.S., H.J.L., and C.L.S.) and Neurology (Y.S.X.), Peking University Third Hospital, Beijing, People's Republic of China .,Beijing Key Laboratory of Spinal Diseases, Beijing, People's Republic of China
| |
Collapse
|
32
|
Zhang W, Zhang X, Ling J, Wei X, Jian Y. Osteo-/odontogenic differentiation of BMP2 and VEGF gene-co-transfected human stem cells from apical papilla. Mol Med Rep 2016; 13:3747-54. [PMID: 26986020 PMCID: PMC4838134 DOI: 10.3892/mmr.2016.4993] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 02/04/2016] [Indexed: 12/13/2022] Open
Abstract
Stem cells from apical papilla (SCAP) possess clear osteo-/odontogenic differentiation capabilities, and are regarded as the major cellular source for root dentin development. Bone morphogenetic protein 2 (BMP2) and vascular endothelial growth factor (VEGF) serve pivotal roles in the modulation of tooth development and dentin formation. However, the synergistic effects of BMP2 and VEGF on osteo-/odontogenic differentiation of SCAP remain unclear. The current study aimed to investigate the proliferative and osteo-/odontogenic differentiating capabilities of BMP2 and VEGF gene-co-transfected SCAP (SCAP-BMP2-VEGF) in vitro. The basic characteristics of the isolated SCAP were identified by the induction of multipotent differentiation and by flow cytometry. Lentiviral vector-mediated gene transfection was conducted with SCAP in order to construct blank vector-transfected SCAP (SCAP-green fluorescent protein), BMP2 gene-transfected SCAP (SCAP-BMP2), VEGF gene-transfected SCAP (SCAP-VEGF) and SCAP-BMP2-VEGF. The Cell Counting Kit 8 assay was used to analyze the proliferative capacities of the four groups of cells. The expression of osteo-/odontogenic genes and proteins in the cells were evaluated by reverse transcription-quantitative polymerase chain reaction and western blotting. The mineralized nodules formed by the four group cells were visualized by alkaline phosphatase (ALP) staining. Among the four groups of cells, SCAP-VEGF was demonstrated to exhibit increased proliferation, and SCAP-BMP2-VEGF exhibited reduced proliferation during eight days observation. SCAP-BMP2-VEGF exhibited significantly increased expression levels of ALP, osteocalcin, dentin sialophosphoprotein, dentin matrix acidic phosphoprotein gene 1 and dentin sialoprotein than the other three groups at the majority of the time points. Furthermore, the SCAP-BMP2-VEGF group exhibited a significantly greater number of ALP-positive mineralized nodules than the other groups following 16 days culture in vitro. In conclusion, lentiviral vector-mediated BMP2 and VEGF gene co-transfection significantly activated the osteo-/odontogenic differentiation of human SCAP.
Collapse
Affiliation(s)
- Wen Zhang
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Xiaolei Zhang
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Junqi Ling
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Xi Wei
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Yutao Jian
- Guangdong Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
33
|
Curry AS, Pensa NW, Barlow AM, Bellis SL. Taking cues from the extracellular matrix to design bone-mimetic regenerative scaffolds. Matrix Biol 2016; 52-54:397-412. [PMID: 26940231 DOI: 10.1016/j.matbio.2016.02.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 12/30/2022]
Abstract
There is an ongoing need for effective materials that can replace autologous bone grafts in the clinical treatment of bone injuries and deficiencies. In recent years, research efforts have shifted away from a focus on inert biomaterials to favor scaffolds that mimic the biochemistry and structure of the native bone extracellular matrix (ECM). The expectation is that such scaffolds will integrate with host tissue and actively promote osseous healing. To further enhance the osteoinductivity of bone graft substitutes, ECM-mimetic scaffolds are being engineered with a range of growth factors (GFs). The technologies used to generate GF-modified scaffolds are often inspired by natural processes that regulate the association between endogenous ECMs and GFs. The purpose of this review is to summarize research centered on the development of regenerative scaffolds that replicate the fundamental collagen-hydroxyapatite structure of native bone ECM, and the functionalization of these scaffolds with GFs that stimulate critical events in osteogenesis.
Collapse
Affiliation(s)
- Andrew S Curry
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294, United States
| | - Nicholas W Pensa
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294, United States
| | - Abby M Barlow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294, United States
| | - Susan L Bellis
- Department of Biomedical Engineering, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294, United States; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294, United States.
| |
Collapse
|
34
|
Eğri S, Eczacıoğlu N. Sequential VEGF and BMP-2 releasing PLA-PEG-PLA scaffolds for bone tissue engineering: I. Design and in vitro tests. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:321-329. [DOI: 10.3109/21691401.2016.1147454] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sinan Eğri
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Gaziosmanpaşa University, Tokat, Turkey
| | - Numan Eczacıoğlu
- Department of Bioengineering, Faculty of Engineering, Karamanoğlu Mehmetbey University, Karaman, Turkey
| |
Collapse
|
35
|
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: 23] [Impact Index Per Article: 2.6] [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
|
36
|
Martino MM, Briquez PS, Maruyama K, Hubbell JA. Extracellular matrix-inspired growth factor delivery systems for bone regeneration. Adv Drug Deliv Rev 2015; 94:41-52. [PMID: 25895621 DOI: 10.1016/j.addr.2015.04.007] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 03/27/2015] [Accepted: 04/11/2015] [Indexed: 12/22/2022]
Abstract
Growth factors are very promising molecules to enhance bone regeneration. However, their translation to clinical use has been seriously limited, facing issues related to safety and cost-effectiveness. These problems derive from the vastly supra-physiological doses of growth factor used without optimized delivery systems. Therefore, these issues have motivated the development of new delivery systems allowing better control of the spatiotemporal release and signaling of growth factors. Because the extracellular matrix (ECM) naturally plays a fundamental role in coordinating growth factor activity in vivo, a number of novel delivery systems have been inspired by the growth factor regulatory function of the ECM. After introducing the role of growth factors during the bone regeneration process, this review exposes different issues that growth factor-based therapies have encountered in the clinic and highlights recent delivery approaches based on the natural interaction between growth factor and the ECM.
Collapse
Affiliation(s)
- Mikaël M Martino
- Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| | - Priscilla S Briquez
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Kenta Maruyama
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Jeffrey A Hubbell
- Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA; Materials Science Division, Argonne National Laboratory, Argonne, IL, USA.
| |
Collapse
|
37
|
Lee SH, Kim SW, Lee JI, Yoon HJ. The effect of platelet-rich fibrin on bone regeneration and angiogenesis in rabbit cranial defects. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-015-0031-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
38
|
Compton J, Fragomen A, Rozbruch SR. Skeletal Repair in Distraction Osteogenesis: Mechanisms and Enhancements. JBJS Rev 2015; 3:01874474-201508000-00002. [PMID: 27490473 DOI: 10.2106/jbjs.rvw.n.00107] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jocelyn Compton
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10031
| | - Austin Fragomen
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021
| | - S Robert Rozbruch
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021
| |
Collapse
|
39
|
Durand M, Collombet JM, Frasca S, Begot L, Lataillade JJ, Le Bousse-Kerdilès MC, Holy X. In vivo hypobaric hypoxia performed during the remodeling process accelerates bone healing in mice. Stem Cells Transl Med 2014; 3:958-68. [PMID: 24944208 DOI: 10.5966/sctm.2013-0209] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We investigated the effects of respiratory hypobaric hypoxia on femoral bone-defect repair in mice because hypoxia is believed to influence both mesenchymal stromal cell (MSC) and hematopoietic stem cell mobilization, a process involved in the bone-healing mechanism. To mimic conditions of non-weight-bearing limb immobilization in patients suffering from bone trauma, our hypoxic mouse model was further subjected to hind-limb unloading. A hole was drilled in the right femur of adult male C57/BL6J mice. Four days after surgery, mice were subjected to hind-limb unloading for 1 week. Seven days after surgery, mice were either housed for 4 days in a hypobaric room (FiO2 at 10%) or kept under normoxic conditions. Unsuspended control mice were housed in either hypobaric or normoxic conditions. Animals were sacrificed on postsurgery day 11 to allow for collection of both contralateral and lesioned femurs, blood, and spleen. As assessed by microtomography, delayed hypoxia enhanced bone-healing efficiency by increasing the closing of the cortical defect and the newly synthesized bone volume in the cavity by +55% and +35%, respectively. Proteome analysis and histomorphometric data suggested that bone-repair improvement likely results from the acceleration of the natural bone-healing process rather than from extended mobilization of MSC-derived osteoprogenitors. Hind-limb unloading had hardly any effect beyond delayed hypoxia-enhanced bone-healing efficiency.
Collapse
Affiliation(s)
- Marjorie Durand
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Jean-Marc Collombet
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Sophie Frasca
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Laurent Begot
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Jean-Jacques Lataillade
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Marie-Caroline Le Bousse-Kerdilès
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Xavier Holy
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| |
Collapse
|
40
|
Yu X, Khalil A, Dang PN, Alsberg E, Murphy WL. Multilayered Inorganic Microparticles for Tunable Dual Growth Factor Delivery. ADVANCED FUNCTIONAL MATERIALS 2014; 24:3082-3093. [PMID: 25342948 PMCID: PMC4204399 DOI: 10.1002/adfm.201302859] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
There is an increasing need to control the type, quantity, and timing of growth factors released during tissue healing. Sophisticated delivery systems offering the ability to deliver multiple growth factors with independently tunable kinetics are highly desirable. Here, a multilayered, mineral coated micro-particle (MCMs) platform that can serve as an adaptable dual growth factor delivery system is developed. Bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) are bound to the mineral coatings with high binding efficiencies of up to 80%. BMP-2 is firstly bound onto a 1st mineral coating layer; then VEGF is bound onto a 2nd mineral coating layer. The release of BMP-2 is sustained over a period of 50 days while the release of VEGF is a typical two-phase release with rapid release in the first 14 days and more sustained release for the following 36 days. Notably, the release behaviors of both growth factors can be independently tailored by changing the intrinsic properties of the mineral coatings. Furthermore, the release of BMP-2 can be tuned by changing the thickness of the 2nd layer. This injectable microparticle based delivery platform with tunable growth factor release has immense potential for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Xiaohua Yu
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Andrew Khalil
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Phuong Ngoc Dang
- Department of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- AO Foundation Collaborative Research Center, Clavadelerstrasse 8, Davos, 7270, Switzerland
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
- AO Foundation Collaborative Research Center, Clavadelerstrasse 8, Davos, 7270, Switzerland
- Department of Orthopedics and Rehabilitation, 1300 University Ave, University of Wisconsin, Madison, WI, 53705, USA
| |
Collapse
|
41
|
Calvo-Guirado JL, Delgado-Peña J, Maté-Sánchez JE, Mareque Bueno J, Delgado-Ruiz RA, Romanos GE. Novel hybrid drilling protocol: evaluation for the implant healing--thermal changes, crestal bone loss, and bone-to-implant contact. Clin Oral Implants Res 2014; 26:753-60. [PMID: 24502654 DOI: 10.1111/clr.12341] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2014] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To evaluate a new hybrid drilling protocol, by the analysis of thermal changes in vitro, and their effects in the crestal bone loss and bone-to-implant contact in vivo. MATERIALS AND METHODS Temperature changes during simulated osteotomies with a hybrid drilling technique (biologic plus simplified) (test) versus an incremental drilling technique (control) were investigated. One hundred and twenty random osteotomies were performed (60 by group) in pig ribs up to 3.75-mm-diameter drill to a depth of 10 mm. Thermal changes and time were recorded by paired thermocouples. In a parallel experiment, bilateral mandibular premolars P2, P3, P4, and first molar M1 were extracted from six dogs. After 2-month healing, implant sites were randomly prepared using either of the drilling techniques. Forty eight implants of 3.75 mm diameter and 10 mm length were inserted. The dogs were euthanized at 30 and 90 days, and crestal bone loss (CBL) and bone-to-implant contact (BIC) were evaluated. RESULTS The control group showed maximum temperatures of 35.3 °C ± 1.8 °C, ΔT of 10.4 °C, and a mean time of 100 s/procedure; meanwhile, the test group showed maximum temperatures of 36.7 °C ± 1.2 °C, ΔT of 8.1 °C, and a mean time of 240 s/procedure. After 30 days, CBL values for both groups (test: 1.168 ± 0.194 mm; control: 1.181 ± 0.113 mm) and BIC values (test: 43 ± 2.8%; control: 45 ± 1.3%) were similar, without significant differences (P > 0.05). After 90 days, CBL (test: 1.173 ± 0.187 mm; control: 1.205 ± 0.122 mm) and BIC (test: 64 ± 3.3%; control: 64 ± 2.4%) values were similar, without significant differences (P > 0.05). The BIC values were increased at 90 days in both groups compared with the 30-day period (P < 0.05). CONCLUSIONS Within the limitations of this study, the new hybrid protocol for the preparation of the implant bed without irrigation, increase the temperature similarly to the incremental conventional protocol, and requires twice the time for the completion of the drilling procedure in vitro. Crestal bone loss and bone-to-implant contact in the hybrid drilling protocol are comparable with the conventional drilling protocol and do not affect the osseointegration process in vivo.
Collapse
|
42
|
Rocha CA, Cestari TM, Vidotti HA, de Assis GF, Garlet GP, Taga R. Sintered anorganic bone graft increases autocrine expression of VEGF, MMP-2 and MMP-9 during repair of critical-size bone defects. J Mol Histol 2014; 45:447-61. [PMID: 24482159 DOI: 10.1007/s10735-014-9565-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/20/2014] [Indexed: 12/14/2022]
Abstract
This study aimed to evaluate morphometrically the bone formation and immunohistochemically the expression of vascular endothelial growth factor (VEGF) and metalloproteinase (MMP)-2 and -9 during the healing of critical-size defects treated with sintered anorganic bone (sAB). The 8-mm diameter full-thickness trephine defects created in the parietal bones of rats were filled with sAB (test group) or blood clot (CSD-control group). At 7, 14, 21, 30, 90 and 180 days postoperatively (n = 6/period) the volume of newly formed bone and total number of immunolabeled cells (Ntm) for each protein were determined. Bone formation was smaller and faster in the CSD-control group, stabilizing at 21 days (6.74 mm(3)). The peaks of VEGF, MMP-2 and MMP-9 occurred at 7 and 14 days in fibroblasts and osteoblasts, with mean reduction of 0.80 time at 21 days, keeping constant until 180 days. In the test group, sAB provided continuous bone formation between particles throughout all periods. The peak of MMP-2 was observed at 7-14 days in connective tissue cells and for VEGF and MMP-9 at 30 days in osteoblasts and osteocytes. Ntm for VEGF, MMP-2 and MMP-9 were in average, respectively, 3.70, 2.03 and 5.98 times higher than in the control group. At 180 days, newly formed bone (22.9 mm(3)) was 3.74 times greater in relation to control. The physical and chemical properties of sAB allow increased autocrine expression of VEGF, MMP-2 and MMP-9, favoring bone formation/remodeling with very good healing of cranial defects when compared to natural repair in the CSD-control.
Collapse
Affiliation(s)
- Caroline Andrade Rocha
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, SP, 17012-901, Brazil,
| | | | | | | | | | | |
Collapse
|
43
|
Yoon JS, Lee SH, Yoon HJ. The influence of platelet-rich fibrin on angiogenesis in guided bone regeneration using xenogenic bone substitutes: a study of rabbit cranial defects. J Craniomaxillofac Surg 2014; 42:1071-7. [PMID: 24530076 DOI: 10.1016/j.jcms.2014.01.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 11/18/2013] [Accepted: 01/07/2014] [Indexed: 11/30/2022] Open
Abstract
PURPOSE The purpose of this study was to investigate the influence of platelet-rich fibrin (PRF) on angiogenesis and osteogenesis in guided bone regeneration (GBR) using xenogenic bone in rabbit cranial defects. MATERIALS AND METHODS In each rabbit, 2 circular bone defects, one on either side of the midline, were prepared using a reamer drill. Each of the experimental sites received bovine bone with PRF, and each of the control sites received bovine bone alone. The animals were sacrificed at 1 week (n = 4), 2 weeks (n = 3) and 4 weeks (n = 3). Biopsy samples were examined histomorphometrically by light microscopy, and expression of vascular endothelial growth factor (VEGF) was determined by immunohistochemical staining. RESULTS At all experimental time points, immunostaining intensity for VEGF was consistently higher in the experimental group than in the control group. However, the differences between the control group and the experimental group were not statistically significant in the histomorphometrical and immunohistochemical examinations. CONCLUSIONS The results of this study suggest that PRF may increase the number of marrow cells. However, PRF along with xenogenic bone substitutes does not show a significant effect on bony regeneration. Further large-scale studies are needed to confirm our results.
Collapse
Affiliation(s)
- Jong-Suk Yoon
- Department of Oral and Maxillofacial Surgery, Yeouido St. Mary's Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Hwa Lee
- Department of Oral and Maxillofacial Surgery, Yeouido St. Mary's Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun-Joong Yoon
- Department of Oral and Maxillofacial Surgery, Yeouido St. Mary's Hospital, Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Rose L, Uludağ H. Realizing the potential of gene-based molecular therapies in bone repair. J Bone Miner Res 2013; 28:2245-62. [PMID: 23553878 DOI: 10.1002/jbmr.1944] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/13/2013] [Accepted: 03/19/2013] [Indexed: 12/17/2022]
Abstract
A better understanding of osteogenesis at genetic and biochemical levels is yielding new molecular entities that can modulate bone regeneration and potentially act as novel therapies in a clinical setting. These new entities are motivating alternative approaches for bone repair by utilizing DNA-derived expression systems, as well as RNA-based regulatory molecules controlling the fate of cells involved in osteogenesis. These sophisticated mediators of osteogenesis, however, pose unique delivery challenges that are not obvious in deployment of conventional therapeutic agents. Viral and nonviral delivery systems are actively pursued in preclinical animal models to realize the potential of the gene-based medicines. This article will summarize promising bone-inducing molecular agents on the horizon as well as provide a critical review of delivery systems employed for their administration. Special attention was paid to synthetic (nonviral) delivery systems because they are more likely to be adopted for clinical testing because of safety considerations. We present a comparative analysis of dose-response relationships, as well as pharmacokinetic and pharmacodynamic features of various approaches, with the purpose of clearly defining the current frontier in the field. We conclude with the authors' perspective on the future of gene-based therapy of bone defects, articulating promising research avenues to advance the field of clinical bone repair.
Collapse
Affiliation(s)
- Laura Rose
- Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
45
|
You W, Fan L, Duan D, Tian L, Dang X, Wang C, Wang K. Foxc2 over-expression in bone marrow mesenchymal stem cells stimulates osteogenic differentiation and inhibits adipogenic differentiation. Mol Cell Biochem 2013; 386:125-34. [DOI: 10.1007/s11010-013-1851-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/27/2013] [Indexed: 11/29/2022]
|
46
|
You W, Gao H, Fan L, Duan D, Wang C, Wang K. Foxc2 regulates osteogenesis and angiogenesis of bone marrow mesenchymal stem cells. BMC Musculoskelet Disord 2013; 14:199. [PMID: 23815774 PMCID: PMC3710500 DOI: 10.1186/1471-2474-14-199] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 06/28/2013] [Indexed: 02/07/2023] Open
Abstract
Background The Forkhead/Fox transcription factor Foxc2 is a critical regulator of osteogenesis and angiogenesis of cells. Bone marrow mesenchymal stem cells (BMSCs) have the capacity to differentiate into osteoblasts, chondrocytes, adipocytes, myocytes and fibroblasts. The present study investigates the role of Foxc2 overexpression in osteogenesis and angiogenesis of BMSCs in vitro. Methods BMSCs were isolated from SD rat femurs and tibias, and characterized by cell surface antigen identification and osteoblasts and adipocytes differentiation. The cells were transfected with lentiviral Foxc2 (Lv-Foxc2) or green fluorescent protein (Lv-GFP). Seventy hours later, Foxc2 expression was observed using real time-PCR and Western blot. The transfected cells were stained with Alizarin red S or alkaline phosphatase (ALP) after osteogenic induction. Meanwhile, the expression levels of osteocalcin (OCN), Runt-related transcription factor 2 (Runx2), vascular endothelial growth factor (VEGF) and platelet-derived growth factor-β (PDGF-β) were measured by real time-PCR, Western blot and immunostaining. Results Results of cell characterization showed that the cells were positive to CD44 (99.56%) and negative to CD34 (0.44%), and could differentiate into osteoblasts and adipocytes. Foxc2 overexpression not only increased the numbers of mineralized nodes and ALP activity, but also enhanced the expressions of Runx2, OCN, VEGF and PDGF-β in transfected BMSCs after osteogenic induction. The effects of Foxc2 on osteogenesis and angiogenesis were significantly different between Lv-Foxc2 transfected BMSCs and Lv-GFP transfected BMSCs (P<0.05). In addition, the MAPK-specific inhibitors, PD98059 and LY294002, blocked the Foxc2-induced regulation of BMSC differentiation. Conclusions Foxc2 gene is successfully transfected into BMSCs with stable and high expression. The overexpression of Foxc2 acts on BMSCs to stimulate osteogenesis and angiogenesis. The effect of Foxc2 on angiogenesis of the cells is mediated via activating PI3K and ERK.
Collapse
Affiliation(s)
- Wulin You
- Department of Orthopedics, The Second Affilliated Hospital of Xi'an Jiaotong University, Xiwu Road, Xi'an, Shaanxi Province 710004, China
| | | | | | | | | | | |
Collapse
|
47
|
Strachna O, Torrecilla D, Reumann MK, Serganova I, Kim J, Gieschler S, Boskey AL, Blasberg RG, Mayer-Kuckuk P. Molecular imaging of expression of vascular endothelial growth factor a (VEGF a) in femoral bone grafts transplanted into living mice. Cell Transplant 2013; 23:901-12. [PMID: 23582187 PMCID: PMC5477423 DOI: 10.3727/096368912x667015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The biology of cells transplanted with bone grafts is incompletely understood. Focusing on the early angiogenic response postgrafting, we report a mouse femur graft model in which grafts were derived from mice transgenic for a firefly luciferase (FLuc) bioluminescence reporter gene driven by a promoter for the angiogenic signaling molecule vascular endothelial growth factor (VEGF). Upon transplantation into wild-type (wt) mice, in vivo bioluminescence imaging (BLI) permitted longitudinal visualization and measurements of VEGF promoter activity in the transplanted graft cells and demonstrated a lag period of 7 days posttransplantation prior to robust induction of the promoter. To determine cellular mediators of VEGF induction in graft bone, primary graft-derived osteoblastic cells (GDOsts) were characterized. In vitro BLI on GDOsts showed hypoxia-induced VEGF expression and that this induction depended on PI3K signaling and, to a lesser degree, on the MEK pathway. This transcriptional regulation correlated with VEGF protein production and was validated in GDOsts seeded on demineralized bone matrix (DBM), a bone graft substitute material. Together, combined imaging of VEGF expression in living animals and in live cells provided clues about the regulation of VEGF in cells post-bone grafting. These data are particularly significant toward the development of future smart bone graft substitutes.
Collapse
Affiliation(s)
- Olga Strachna
- Research Division, Hospital for Special Surgery, New York, NY, USA
| | | | | | - Inna Serganova
- Center for Molecular Imaging in Cancer, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Jihye Kim
- Research Division, Hospital for Special Surgery, New York, NY, USA
| | - Simone Gieschler
- Research Division, Hospital for Special Surgery, New York, NY, USA
| | - Adele L. Boskey
- Mineralized Tissue Laboratory, Hospital for Special Surgery, New York, NY, USA
| | - Ronald G. Blasberg
- Center for Molecular Imaging in Cancer, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Philipp Mayer-Kuckuk
- Bone Cell Biology and Imaging Laboratory, Department of Orthopedics, MRI, Technical University Munich, Munich, Germany
| |
Collapse
|
48
|
Cardemil C, Omar OM, Norlindh B, Wexell CL, Thomsen P. The effects of a systemic single dose of zoledronic acid on post-implantation bone remodelling and inflammation in an ovariectomised rat model. Biomaterials 2012. [PMID: 23182921 DOI: 10.1016/j.biomaterials.2012.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Bisphosphonates reverse the negative effects of ovariectomy on bone, but they have also been associated with adverse processes in human jawbone. The molecular events determining bone regeneration and implant integration in osteoporotic conditions, with and without bisphosphonate treatment, are unclear. In this study, ovariectomised rats, to which a single dose of saline (NaCl) or zoledronic acid (Zol) was administered, received titanium alloy implants in their tibiae and mandibles. An enzyme-linked immunosorbent assay, gene expression analysis and histomorphometry were performed. The results show that ovariectomy, per se, upregulated the expression of genes denoting bone formation in the tibia, bone remodelling in the mandible and apoptosis in the tibia and mandible. Zoledronic acid administration resulted in lower levels of a remodelling marker in serum and downregulated gene expression for inflammation, bone formation, angiogenesis and apoptosis, mainly in the mandible, after 28 d of healing. Histomorphometry revealed improved bone-to-implant contact in the tibia, while the opposite was observed in the mandible. The present data show that a systemic single dose of zoledronic acid, in ovariectomised animals, results in site-specific differences in the regulation of genes involved in bone healing and regeneration in association with implant installation. These events occur in parallel with site-specific differences in the rate of osseointegration, indicating diverse tissue responses in the tibia and mandible after zoledronic acid treatment. The zoledronic acid effect on gene expression, during the late phase of healing in the mandible, suggests negative effects by the anti-resorptive agent on osseointegration at that particular site.
Collapse
Affiliation(s)
- Carina Cardemil
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Box 412, SE-40530 Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
49
|
Zhang P, Hamamura K, Jiang C, Zhao L, Yokota H. Salubrinal promotes healing of surgical wounds in rat femurs. J Bone Miner Metab 2012; 30:568-79. [PMID: 22610062 DOI: 10.1007/s00774-012-0359-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 04/02/2012] [Indexed: 10/28/2022]
Abstract
Phosphorylation of eukaryotic initiation factor 2α (eIF2α), transiently activated by various cellular stresses, is known to alleviate stress-induced cellular damage. Here, we addressed a question: does elevation of eIF2α phosphorylation by salubrinal (a pharmacological inhibitor of eIF2α dephosphorylation) enhance healing of bone wounds? We hypothesized that salubrinal would accelerate a closure of surgically generated bone holes by modifying expression of stress-sensitive genes. To examine this hypothesis, we employed a rat wound model. Surgical wounds were generated on anterior and posterior femoral cortexes, and salubrinal was locally administered on the anterior side. The results showed that, compared to a contralateral control, the size of surgical wounds was reduced by 10.8 % (day 10) and 18.0 % (day 20) on the anterior side (both p < 0.001), and 4.1 % (day 10; p < 0.05) and 11.1 % (day 20; p < 0.001) on the posterior side. In addition, salubrinal locally elevated cortical thickness and increased BMD and BMC. Pharmacokinetic analysis revealed that subcutaneous injection of salubrinal transiently increased its concentration in plasma followed by a rapid decrease within 24 h, and its half-life in plasma was 1.2 h. Salubrinal altered the phosphorylation level of eIF2α as well as the mRNA levels of ATF3, ATF4, and CHOP, and suppressed cell death induced by stress to the endoplasmic reticulum. In summary, the results herein demonstrate that subcutaneous administration of salubrinal accelerates healing of surgically generated bone holes through the modulation of eIF2α phosphorylation.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, SL220, 723 West Michigan Street, Indianapolis, IN, 46202, USA
| | | | | | | | | |
Collapse
|
50
|
Yun YR, Jang JH, Jeon E, Kang W, Lee S, Won JE, Kim HW, Wall I. Administration of growth factors for bone regeneration. Regen Med 2012; 7:369-85. [DOI: 10.2217/rme.12.1] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Growth factors (GFs) such as BMPs, FGFs, VEGFs and IGFs have significant impacts on osteoblast behavior, and thus have been widely utilized for bone tissue regeneration. Recently, securing biological stability for a sustainable and controllable release to the target tissue has been a challenge to practical applications. This challenge has been addressed to some degree with the development of appropriate carrier materials and delivery systems. This review highlights the importance and roles of those GFs, as well as their proper administration for targeting bone regeneration. Additionally, the in vitro and in vivo performance of those GFs with or without the use of carrier systems in the repair and regeneration of bone tissue is systematically addressed. Moreover, some recent advances in the utility of the GFs, such as using fusion technology, are also reviewed.
Collapse
Affiliation(s)
- Ye-Rang Yun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Korea
| | - Jun Hyeog Jang
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Eunyi Jeon
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Wonmo Kang
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Sujin Lee
- Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea
| | - Jong-Eun Won
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Korea
- Department of Nanobiomedical Science & WCU Research Center, Dankook University Graduate School, Cheonan 330-714, Korea
| | - Hae Won Kim
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 330-714, Korea
| | - Ivan Wall
- Department of Nanobiomedical Science & WCU Research Center, Dankook University Graduate School, Cheonan 330-714, Korea
- Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| |
Collapse
|