51
|
Abstract
This chapter provides an overview of the growth factors active in bone regeneration and healing. Both normal and impaired bone healing are discussed, with a focus on the spatiotemporal activity of the various growth factors known to be involved in the healing response. The review highlights the activities of most important growth factors impacting bone regeneration, with a particular emphasis on those being pursued for clinical translation or which have already been marketed as components of bone regenerative materials. Current approaches the use of bone grafts in clinical settings of bone repair (including bone grafts) are summarized, and carrier systems (scaffolds) for bone tissue engineering via localized growth factor delivery are reviewed. The chapter concludes with a consideration of how bone repair might be improved in the future.
Collapse
|
52
|
|
53
|
Liebig BE, Kisiday JD, Bahney CS, Ehrhart NP, Goodrich LR. The platelet-rich plasma and mesenchymal stem cell milieu: A review of therapeutic effects on bone healing. J Orthop Res 2020; 38:2539-2550. [PMID: 32589800 PMCID: PMC8354210 DOI: 10.1002/jor.24786] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 02/04/2023]
Abstract
Platelet-rich plasma is autologous plasma that contains concentrated platelets compared to whole blood. It is relatively inexpensive to produce, can be easily isolated from whole blood, and can be administered while the patient is in the operating room. Further, because platelet-rich plasma is an autologous therapy, there is minimal risk for adverse reactions to the patient. Platelet-rich plasma has been used to promote bone regeneration due to its abundance of concentrated growth factors that are essential to wound healing. In this review, we summarize the methods for producing platelet-rich plasma and the history of its use in bone regeneration. We also summarize the growth factor profiles derived from platelet-rich plasma, with emphasis on those factors that play a direct role in promoting bone repair within the local fracture environment. In addition, we discuss the potential advantages of combining platelet-rich plasma with mesenchymal stem cells, a multipotent cell type often obtained from bone marrow or fat, to improve craniofacial and long bone regeneration. We detail what is currently known about how platelet-rich plasma influences mesenchymal stem cells in vitro, and then highlight the clinical outcomes of administering platelet-rich plasma and mesenchymal stem cells as a combination therapy to promote bone regeneration in vivo.
Collapse
Affiliation(s)
- Bethany E. Liebig
- Department of Clinical Sciences, Orthopaedic Research Center, Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - John D. Kisiday
- Department of Clinical Sciences, Orthopaedic Research Center, Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Chelsea S. Bahney
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado
| | - Nicole P. Ehrhart
- Department of Clinical Sciences, Flint Animal Cancer Center, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Laurie R. Goodrich
- Department of Clinical Sciences, Orthopaedic Research Center, Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
54
|
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: 30] [Impact Index Per Article: 7.5] [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]
|
55
|
Sun K, Lin H, Tang Y, Xiang S, Xue J, Yin W, Tan J, Peng H, Alexander PG, Tuan RS, Wang B. Injectable BMP-2 gene-activated scaffold for the repair of cranial bone defect in mice. Stem Cells Transl Med 2020; 9:1631-1642. [PMID: 32785966 PMCID: PMC7695643 DOI: 10.1002/sctm.19-0315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 06/22/2020] [Accepted: 07/05/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering using adult human mesenchymal stem cells (MSCs) seeded within biomaterial scaffolds has shown the potential to enhance bone healing. Recently, we have developed an injectable, biodegradable methacrylated gelatin‐based hydrogel, which was especially effective in producing scaffolds in situ and allowed the delivery of high viable stem cells and gene vehicles. The well‐demonstrated benefits of recombinant adeno‐associated viral (rAAV) vector, including long‐term gene transfer efficiency and relative safety, combination of gene and cell therapies has been developed in both basic and translational research to support future bone tissue regeneration clinical trials. In this study, we have critically assessed the applicability of single‐step visible light (VL) photocrosslinking fabrication of gelatin scaffold to deliver rAAV encoding human bone morphogenetic protein‐2 (BMP‐2) gene to address the need for sustained BMP‐2 presence localized within scaffolds for the repair of cranial bone defect in mouse model. In this method, rAAV‐BMP‐2 and human bone marrow‐derived MSCs (hBMSCs) were simultaneously included into gelatin scaffolds during scaffold formation by VL illumination. We demonstrated that the subsequent release of rAAV‐BMP‐2 constructs from the scaffold matrix, which resulted in efficient in situ expression of BMP‐2 gene by hBMSCs seeded within the scaffolds, and thus induced their osteogenic differentiation without the supplement of exogenous BMP‐2. The reparative capacity of this novel stem cell‐seeded and gene‐activated scaffolds was further confirmed in the cranial defect in the severe combined immunodeficiency mice, revealed by imaging, histology, and immunohistochemistry at 6 weeks after cranial defect treatment.
Collapse
Affiliation(s)
- Kai Sun
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shiqi Xiang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jingwen Xue
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Weifeng Yin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jian Tan
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Peter G Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rocky S Tuan
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
56
|
Rukavina P, Koch F, Wehrle M, Tröndle K, Björn Stark G, Koltay P, Zimmermann S, Zengerle R, Lampert F, Strassburg S, Finkenzeller G, Simunovic F. In vivo evaluation of bioprinted prevascularized bone tissue. Biotechnol Bioeng 2020; 117:3902-3911. [PMID: 32749669 DOI: 10.1002/bit.27527] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/16/2020] [Accepted: 08/01/2020] [Indexed: 12/21/2022]
Abstract
Bioprinting can be considered as a progression of the classical tissue engineering approach, in which cells are randomly seeded into scaffolds. Bioprinting offers the advantage that cells can be placed with high spatial fidelity within three-dimensional tissue constructs. A decisive factor to be addressed for bioprinting approaches of artificial tissues is that almost all tissues of the human body depend on a functioning vascular system for the supply of oxygen and nutrients. In this study, we have generated cuboid prevascularized bone tissue constructs by bioprinting human adipose-derived mesenchymal stem cells (ASCs) and human umbilical vein endothelial cells (HUVECs) by extrusion-based bioprinting and drop-on-demand (DoD) bioprinting, respectively. The computer-generated print design could be verified in vitro after printing. After subcutaneous implantation of bioprinted constructs in immunodeficient mice, blood vessel formation with human microvessels of different calibers could be detected arising from bioprinted HUVECs and stabilization of human blood vessels by mouse pericytes was observed. In addition, bioprinted ASCs were able to synthesize a calcified bone matrix as an indicator of ectopic bone formation. These results indicate that the combined bioprinting of ASCs and HUVECs represents a promising strategy to produce prevascularized artificial bone tissue for prospective applications in the treatment of critical-sized bone defects.
Collapse
Affiliation(s)
- Patrick Rukavina
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fritz Koch
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Maximilian Wehrle
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kevin Tröndle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - G Björn Stark
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Koltay
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.,Hahn-Schickard, Freiburg, Germany.,Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.,Hahn-Schickard, Freiburg, Germany
| | - Florian Lampert
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sandra Strassburg
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günter Finkenzeller
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Filip Simunovic
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
57
|
Zafar MS, Amin F, Fareed MA, Ghabbani H, Riaz S, Khurshid Z, Kumar N. Biomimetic Aspects of Restorative Dentistry Biomaterials. Biomimetics (Basel) 2020; 5:E34. [PMID: 32679703 PMCID: PMC7557867 DOI: 10.3390/biomimetics5030034] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Biomimetic has emerged as a multi-disciplinary science in several biomedical subjects in recent decades, including biomaterials and dentistry. In restorative dentistry, biomimetic approaches have been applied for a range of applications, such as restoring tooth defects using bioinspired peptides to achieve remineralization, bioactive and biomimetic biomaterials, and tissue engineering for regeneration. Advancements in the modern adhesive restorative materials, understanding of biomaterial-tissue interaction at the nano and microscale further enhanced the restorative materials' properties (such as color, morphology, and strength) to mimic natural teeth. In addition, the tissue-engineering approaches resulted in regeneration of lost or damaged dental tissues mimicking their natural counterpart. The aim of the present article is to review various biomimetic approaches used to replace lost or damaged dental tissues using restorative biomaterials and tissue-engineering techniques. In addition, tooth structure, and various biomimetic properties of dental restorative materials and tissue-engineering scaffold materials, are discussed.
Collapse
Affiliation(s)
- Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah, Al Munawwarah 41311, Saudi Arabia;
- Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan
| | - Faiza Amin
- Science of Dental Materials Department, Dow Dental College, Dow University of Health Sciences, Karachi 74200, Pakistan;
| | - Muhmmad Amber Fareed
- Adult Restorative Dentistry, Dental Biomaterials and Prosthodontics Oman Dental College, Muscat 116, Sultanate of Oman;
| | - Hani Ghabbani
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah, Al Munawwarah 41311, Saudi Arabia;
| | - Samiya Riaz
- School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Zohaib Khurshid
- Department of Prosthodontics and Dental Implantology, College of Dentistry, King Faisal University, Al-Ahsa 31982, Saudia Arabia;
| | - Naresh Kumar
- Department of Science of Dental Materials, Dow University of Health Sciences, Karachi 74200, Pakistan;
| |
Collapse
|
58
|
Huang K, Gu Z, Wu J. Tofu-Incorporated Hydrogels for Potential Bone Regeneration. ACS Biomater Sci Eng 2020; 6:3037-3045. [DOI: 10.1021/acsbiomaterials.9b01997] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Keqing Huang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, P. R. China
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, 518057, P. R. China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, 518057, P. R. China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, P. R. China
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, 518057, P. R. China
| |
Collapse
|
59
|
Safarova Y, Umbayev B, Hortelano G, Askarova S. Mesenchymal stem cells modifications for enhanced bone targeting and bone regeneration. Regen Med 2020; 15:1579-1594. [PMID: 32297546 DOI: 10.2217/rme-2019-0081] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In pathological bone conditions (e.g., osteoporotic fractures or critical size bone defects), increasing the pool of osteoblast progenitor cells is a promising therapeutic approach to facilitate bone healing. Since mesenchymal stem cells (MSCs) give rise to the osteogenic lineage, a number of clinical trials investigated the potential of MSCs transplantation for bone regeneration. However, the engraftment of transplanted cells is often hindered by insufficient oxygen and nutrients supply and the tendency of MSCs to home to different sites of the body. In this review, we discuss various approaches of MSCs transplantation for bone regeneration including scaffold and hydrogel constructs, genetic modifications and surface engineering of the cell membrane aimed to improve homing and increase cell viability, proliferation and differentiation.
Collapse
Affiliation(s)
- Yuliya Safarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan.,School of Engineering & Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Bauyrzhan Umbayev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Gonzalo Hortelano
- School of Sciences & Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Sholpan Askarova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
60
|
The Use of Platelet-Rich Plasma for the Treatment of Osteonecrosis of the Femoral Head: A Systematic Review. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2642439. [PMID: 32219128 PMCID: PMC7081027 DOI: 10.1155/2020/2642439] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/20/2020] [Indexed: 12/18/2022]
Abstract
Background As a pathological process, osteonecrosis of the femoral head (ONFH) is characterized by the avascularity of the femoral head, cellular necrosis, microfracture, and the collapse of the articular surface. Currently, critical treatment for early-stage ONFH is limited to core decompression. However, the efficacy of core decompression remains controversial. To improve the core decompression efficacy, regenerative techniques such as the use of platelet-rich plasma (PRP) were proposed for early-stage ONFH. As a type of autologous plasma containing concentrations of platelets greater than the baseline, PRP plays an important role in tissue repair, regeneration, and the differentiation of mesenchymal stem cells (MSCs). In this review, we present a comprehensive overview of the operation modes, mechanism, and efficacy of PRP for early-stage ONFH treatment. Methods We searched for relevant studies in the PubMed, Web of Science, and Embase databases. By searching these electronic databases, the identification of either clinical or experimental studies evaluating PRP, MSC, core decompression, and ONFH was our goal. Results Seventeen studies of PRP and avascular necrosis of the femoral head were evaluated in our review. Ten studies related to the possible mechanism of PRP for treating ONFH were reviewed. Seven studies of the operation modes of PRP in treating ONFH were identified. We reviewed the efficacy of PRP in treating ONFH systematically and made an attempt to compare the PRP operation modes in 7 studies and other operation modes in past studies for early-stage ONFH treatment. Conclusion PRP treats ONFH mainly through three mechanisms: inducing angiogenesis and osteogenesis to accelerate bone healing, inhibiting inflammatory reactions in necrotic lesions, and preventing apoptosis induced by glucocorticoids. In addition, as an adjunctive therapy for core decompression, the use of PRP is recommended to improve the treatment of early-stage ONFH patients, especially when combined with stem cells and bone grafts, by inducing osteogenic activity and stimulating the differentiation of stem cells in necrotic lesions.
Collapse
|
61
|
Sheehy EJ, Lemoine M, Clarke D, Gonzalez Vazquez A, O’Brien FJ. The Incorporation of Marine Coral Microparticles into Collagen-Based Scaffolds Promotes Osteogenesis of Human Mesenchymal Stromal Cells via Calcium Ion Signalling. Mar Drugs 2020; 18:md18020074. [PMID: 31979233 PMCID: PMC7073845 DOI: 10.3390/md18020074] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/08/2020] [Accepted: 01/21/2020] [Indexed: 01/24/2023] Open
Abstract
Composite biomaterial scaffolds consisting of natural polymers and bioceramics may offer an alternative to autologous grafts for applications such as bone repair. Herein, we sought to investigate the possibility of incorporating marine coral microparticles into a collagen-based scaffold, a process which we hypothesised would enhance the mechanical properties of the scaffold as well its capacity to promote osteogenesis of human mesenchymal stromal cells. Cryomilling and sieving were utilised to achieve coral microparticles of mean diameters 14 µm and 64 µm which were separately incorporated into collagen-based slurries and freeze-dried to form porous scaffolds. X-ray diffraction and Fourier transform infrared spectroscopy determined the coral microparticles to be comprised of calcium carbonate whereas collagen/coral composite scaffolds were shown to have a crystalline calcium ethanoate structure. Crosslinked collagen/coral scaffolds demonstrated enhanced compressive properties when compared to collagen only scaffolds and also promoted more robust osteogenic differentiation of mesenchymal stromal cells, as indicated by increased expression of bone morphogenetic protein 2 at the gene level, and enhanced alkaline phosphatase activity and calcium accumulation at the protein level. Only subtle differences were observed when comparing the effect of coral microparticles of different sizes, with improved osteogenesis occurring as a result of calcium ion signalling delivered from collagen/coral composite scaffolds. These scaffolds, fabricated from entirely natural sources, therefore show promise as novel biomaterials for tissue engineering applications such as bone regeneration.
Collapse
Affiliation(s)
- Eamon J. Sheehy
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (E.J.S.); (M.L.)
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Mark Lemoine
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (E.J.S.); (M.L.)
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Declan Clarke
- Zoan Biomed Ltd., An Luslann, Kylebroughlan, Moycullen, H91 TXV5 Co Galway, Ireland;
| | - Arlyng Gonzalez Vazquez
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (E.J.S.); (M.L.)
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
- Correspondence: (A.G.V.); (F.J.O.); Tel.: +353-1-402-8506 (A.G.V.); +353-1-402-2149 (F.J.O.)
| | - Fergal J. O’Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (E.J.S.); (M.L.)
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
- Correspondence: (A.G.V.); (F.J.O.); Tel.: +353-1-402-8506 (A.G.V.); +353-1-402-2149 (F.J.O.)
| |
Collapse
|
62
|
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: 7.3] [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
|
63
|
Xu G, Yamamoto N, Nojima T, Hayashi K, Takeuchi A, Miwa S, Igarashi K, Tsuchiya H. The process of bone regeneration from devitalization to revitalization after pedicle freezing with immunohistochemical and histological examination in rabbits. Cryobiology 2019; 92:130-137. [PMID: 31875528 DOI: 10.1016/j.cryobiol.2019.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/13/2019] [Accepted: 12/20/2019] [Indexed: 10/25/2022]
Abstract
The pedicle freezing procedure by liquid nitrogen is a method for the reconstruction of tumor-bearing bone after malignant tumor resection. However, the regenerative mechanism of bone after the pedicle freezing procedure is unclear. We investigated the complete process from devitalization to revitalization of bone after the pedicle freezing procedure in 13 rabbits. After osteotomy the 5 mm distal femurs were immersed in liquid nitrogen, and the specimens were divided into frozen area and sub-frozen area. The bilateral femurs were harvested for evaluation of bone regeneration by histological and immunohistochemical examination (VEGF, CD31, BMP-2 and Runx2) from 1 week to 52 weeks. The diameter of operating femurs was compared with contralateral femurs from 6 weeks to 52 weeks. No viable cells could be found from 1 to 8 weeks in the frozen area, and a mean 1.83 cm necrotic range were detected in the sub-frozen area. The periosteal reaction, massive fibrous tissue and immature bone matrix invaded from the normal area to the necrotic area from 12 weeks. Subsequently, the necrotic bone was gradually replaced by newly formed bone by creeping substitution, with endochondral and intramembrane bone formation. The diameter of frozen femurs was significantly larger than the contralateral femur at the same period from 8 weeks to 52 weeks (P < 0.01). All immunohistochemical factors were positively expressed in both areas at different time points. The active osteoblasts and microvessel migrated from marrow cavity and periosteum into dead bone. This study suggested that the frozen bone not only provides a scaffold but also possesses excellent osteoinductive properties.
Collapse
Affiliation(s)
- Gang Xu
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Norio Yamamoto
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan.
| | - Takayuki Nojima
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan; Section of Diagnostic Pathology, Kanazawa University Hospital, Kanazawa, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Akihiko Takeuchi
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Shinji Miwa
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Kentaro Igarashi
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Kanazawa University School of Medicine, Kanazawa, Japan
| |
Collapse
|
64
|
Magnesium Enhances Osteogenesis of BMSCs by Tuning Osteoimmunomodulation. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7908205. [PMID: 31828131 PMCID: PMC6885163 DOI: 10.1155/2019/7908205] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/17/2019] [Indexed: 11/18/2022]
Abstract
In the process of bone tissue engineering, the osteoimmunomodulatory property of biomaterials is very important for osteogenic differentiation of stem cells, which determines the outcome of bone regeneration. Magnesium (Mg) is a biodegradable, biocompatible metal that has osteoconductive properties and has been regarded as a promising bone biomaterial. However, the high degradation rate of Mg leads to excessive inflammation, thereby restricting its application in bone tissue engineering. Importantly, different coatings or magnesium alloys have been utilized to lower the rate of degradation. In fact, a prior study proved that β-TCP coating of Mg scaffolds can modulate the osteoimmunomodulatory properties of Mg-based biomaterials and create a favorable immune microenvironment for osteogenesis. However, the osteoimmunomodulatory properties of Mg ions themselves have not been explored yet. In this study, the osteoimmunomodulatory properties of Mg ions with involvement of macrophages and bone marrow stem cells (BMSCs) were systematically investigated. Microscale Mg ions (100 mg/L) were found to possess osteoimmunomodulatory properties that favor bone formation. Specifically, microscale Mg ions induced M2 phenotype changes of macrophages and the release of anti-inflammatory cytokines by inhibiting the TLR-NF-κB signaling pathway. Microscale Mg ions also stimulated the expression of osteoinductive molecules in macrophages while Mg ions/macrophage-conditioned medium promoted osteogenesis of BMSCs through the BMP/SMAD signaling pathway. These findings indicate that manipulating Mg ion concentration can endow the Mg biomaterial with favorable osteoimmunomodulatory properties, thereby providing fundamental evidence for improving and modifying the effect of Mg-based bone biomaterials.
Collapse
|
65
|
Juhl O, Zhao N, Merife AB, Cohen D, Friedman M, Zhang Y, Schwartz Z, Wang Y, Donahue H. Aptamer-Functionalized Fibrin Hydrogel Improves Vascular Endothelial Growth Factor Release Kinetics and Enhances Angiogenesis and Osteogenesis in Critically Sized Cranial Defects. ACS Biomater Sci Eng 2019; 5:6152-6160. [PMID: 32190730 PMCID: PMC7079287 DOI: 10.1021/acsbiomaterials.9b01175] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An aging population, decreased activity levels and increased combat injuries, have led to an increase in critical sized bone defects. As more defects are treated using allografts, which is the current standard of care, the deficiencies of allografts are becoming more evident. Allografts lack the angiogenic potential to induce sufficient vasculogenesis to counteract the hypoxic environment associated with critical sized bone defects. In this study, aptamer-functionalized fibrin hydrogels (AFH), engineered to release vascular endothelial growth factor (VEGF), were evaluated for their material properties, growth factor release kinetics, and angiogenic and osteogenic potential in vivo. Aptamer functionalization to native fibrin did not result in significant changes in biocompatibility or hydrogel gelation. However, aptamer functionalization of fibrin did improve the release kinetics of VEGF from AFH and, when compared to FH, reduced the diffusivity and extended the release of VEGF several days longer. VEGF released from AFH, in vivo, increased vascularization to a greater degree, relative to VEGF released from FH, in a murine critical-sized cranial defect. Defects treated with AFH loaded with VEGF, relative to nonhydrogel loaded controls, showed a nominal increase in osteogenesis. Together, these data suggest that AFH more efficiently incorporates and retains VEGF in vitro and in vivo, which then enhances angiogenesis and osteogenesis to a greater extent in vivo than FH.
Collapse
Affiliation(s)
- Otto Juhl
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Nan Zhao
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Anna-Blessing Merife
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - David Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Michael Friedman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Yue Zhang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Henry Donahue
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| |
Collapse
|
66
|
Raftery RM, Walsh DP, Blokpoel Ferreras L, Mencía Castaño I, Chen G, LeMoine M, Osman G, Shakesheff KM, Dixon JE, O'Brien FJ. Highly versatile cell-penetrating peptide loaded scaffold for efficient and localised gene delivery to multiple cell types: From development to application in tissue engineering. Biomaterials 2019; 216:119277. [PMID: 31252371 DOI: 10.1016/j.biomaterials.2019.119277] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/10/2023]
Abstract
Gene therapy has recently come of age with seven viral vector-based therapies gaining regulatory approval in recent years. In tissue engineering, non-viral vectors are preferred over viral vectors, however, lower transfection efficiencies and difficulties with delivery remain major limitations hampering clinical translation. This study describes the development of a novel multi-domain cell-penetrating peptide, GET, designed to enhance cell interaction and intracellular translocation of nucleic acids; combined with a series of porous collagen-based scaffolds with proven regenerative potential for different indications. GET was capable of transfecting cell types from all three germ layers, including stem cells, with an efficiency comparable to Lipofectamine® 3000, without inducing cytotoxicity. When implanted in vivo, GET gene-activated scaffolds allowed for host cell infiltration, transfection localized to the implantation site and sustained, but transient, changes in gene expression - demonstrating both the efficacy and safety of the approach. Finally, GET carrying osteogenic (pBMP-2) and angiogenic (pVEGF) genes were incorporated into collagen-hydroxyapatite scaffolds and with a single 2 μg dose of therapeutic pDNA, induced complete repair of critical-sized bone defects within 4 weeks. GET represents an exciting development in gene therapy and by combining it with a scaffold-based delivery system offers tissue engineering solutions for a myriad of regenerative indications.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - David P Walsh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland; Translational Research in Nanomedical Devices, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Lia Blokpoel Ferreras
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Irene Mencía Castaño
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mark LeMoine
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Gizem Osman
- Centre for Biomedical Sciences, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Kevin M Shakesheff
- Centre for Biomedical Sciences, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - James E Dixon
- Centre for Biomedical Sciences, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.
| |
Collapse
|
67
|
Huang K, Hou J, Gu Z, Wu J. Egg-White-/Eggshell-Based Biomimetic Hybrid Hydrogels for Bone Regeneration. ACS Biomater Sci Eng 2019; 5:5384-5391. [PMID: 33464059 DOI: 10.1021/acsbiomaterials.9b00990] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Keqing Huang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, P. R. China
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen 518057, P. R. China
| | - Jingyi Hou
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107# Yan Jiang Road West, Guangzhou, Guangdong 510120, P. R. China
| | - Zhipeng Gu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, P. R. China
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen 518057, P. R. China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, P. R. China
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen 518057, P. R. China
| |
Collapse
|
68
|
Park S, Arai Y, Kim BJ, Bello A, Ashraf S, Park H, Park KS, Lee SH. Suppression of SPRY4 Promotes Osteogenic Differentiation and Bone Formation of Mesenchymal Stem Cell. Tissue Eng Part A 2019; 25:1646-1657. [PMID: 30982407 DOI: 10.1089/ten.tea.2019.0056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The directed differentiation of human adipose-derived stem cells (hASCs) into different cell types has shown great therapeutic potential in treating various diseases. To maximize the therapeutic potentials, researchers have tried manipulating master transcriptional genes that promote efficient differentiation of mesenchymal stem cells (MSCs) such as the MAPK/ERK signaling pathway. Sprouty (SPRY) is a family of proteins that are known to inhibit the MAPK/ERK signaling pathway. Although the role of some SPRY isoforms in MSC differentiation is known, the function of SPRY4 isoform has not been fully elucidated. In the present study, the role of SPRY4 in the multilineage differentiation of hASCs has been elucidated. To investigate the role of SPRY4 in hASC differentiation and tissue regeneration, we performed a transient knockdown of SPRY expression via a small interfering RNA (siSPRY4). Western blot and quantitative polymerase chain reaction results revealed that the treatment of siSPRY4 before induction of differentiation had no significant effect on adipogenic, but reduced chondrogenic, differentiation of hASCs. Interestingly, SPRY4 transient knockdown had a significant effect on the osteogenic differentiation as indicated by the increased messenger RNA (mRNA) and protein expression of osteogenic markers such as alkaline phosphatase (ALP; 2.3-fold) and osteopontin (OPN; 3.5-fold) and increased calcium deposition measured via Alizarin red staining (3.3-fold). Moreover, in vivo tissue regeneration of siSPRY4-treated hASCs in ectopic bone formation and calvarial defect mouse models showed higher bone volume (5.24-fold) and trabecular number (4.59-fold) assessed via histological and microcomputed tomography analyses. We also determined that the enhanced osteogenic differentiation in SPRY4-treated hASCs was due to the induction of ERK1/2 phosphorylation. Taken together, our results suggest that the regulation of SPRY4 through MAPK signaling is a potentially critical aspect on the osteogenic differentiation of hASCs and for bone tissue regeneration, and thus, may be utilized as a potent technique in the development of effective bone therapeutics. Impact Statement This study tried to expand our current understanding of the osteogenic differentiation of mesenchymal stem cells. The transient downregulation of the SPRY4 expression via small interfering RNA (siRNA) showed significant enhancement of the osteogenic differentiation of adipose-derived stem cells via the induction of ERK 1/2 phosphorylation. This suggests the possible mechanism to maximize the potential of stem cell as therapeutics and has a great potential in treating various bone-related diseases.
Collapse
Affiliation(s)
- Sunghyun Park
- Department of Medical Biotechnology, Dongguk University, Goyang-si, Republic of Korea.,Department of Biomedical Science, CHA University, Seongnam-si, Republic of Korea
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University, Goyang-si, Republic of Korea
| | - Byoung Ju Kim
- Department of Medical Biotechnology, Dongguk University, Goyang-si, Republic of Korea
| | - Alvin Bello
- Department of Medical Biotechnology, Dongguk University, Goyang-si, Republic of Korea.,Department of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Sajjad Ashraf
- Department of Biomedical Science, CHA University, Seongnam-si, Republic of Korea
| | - Hansoo Park
- Department of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kyung-Soon Park
- Department of Biomedical Science, CHA University, Seongnam-si, Republic of Korea
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University, Goyang-si, Republic of Korea
| |
Collapse
|
69
|
Walsh DP, Raftery RM, Chen G, Heise A, O'Brien FJ, Cryan SA. Rapid healing of a critical-sized bone defect using a collagen-hydroxyapatite scaffold to facilitate low dose, combinatorial growth factor delivery. J Tissue Eng Regen Med 2019; 13:1843-1853. [PMID: 31306563 DOI: 10.1002/term.2934] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/02/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
The healing of large, critically sized bone defects remains an unmet clinical need in modern orthopaedic medicine. The tissue engineering field is increasingly using biomaterial scaffolds as 3D templates to guide the regenerative process, which can be further augmented via the incorporation of recombinant growth factors. Typically, this necessitates supraphysiological doses of growth factor to facilitate an adequate therapeutic response. Herein, we describe a cell-free, biomaterial implant which is functionalised with a low dose, combinatorial growth factor therapy that is capable of rapidly regenerating vascularised bone tissue within a critical-sized rodent calvarial defect. Specifically, we demonstrate that the dual delivery of the growth factors bone morphogenetic protein-2 (osteogenic) and vascular endothelial growth factor (angiogenic) at a low dose (5 μg/scaffold) on an osteoconductive collagen-hydroxyapatite scaffold is highly effective in healing these critical-sized bone defects. The high affinity between the hydroxyapatite component of this biomimetic scaffold and the growth factors functions to sequester them locally at the defect site. Using this growth factor-loaded scaffold, we show complete bridging of a critical-sized calvarial defect in all specimens at a very early time point of 4 weeks, with a 28-fold increase in new bone volume and seven-fold increase in new bone area compared with a growth factor-free scaffold. Overall, this study demonstrates that a collagen-hydroxyapatite scaffold can be used to locally harness the synergistic relationship between osteogenic and angiogenic growth factors to rapidly regenerate bone tissue without the need for more complex controlled delivery vehicles or high total growth factor doses.
Collapse
Affiliation(s)
- David P Walsh
- Drug Delivery and Advanced Materials Team, School of Pharmacy, RCSI, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI, TCD, Dublin, Ireland
| | - Rosanne M Raftery
- Drug Delivery and Advanced Materials Team, School of Pharmacy, RCSI, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI, TCD, Dublin, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), RCSI, Dublin, Ireland
| | - Andreas Heise
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI, TCD, Dublin, Ireland
- Department of Chemistry, RCSI, Dublin, Ireland
- Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway, Ireland
| | - Fergal J O'Brien
- Drug Delivery and Advanced Materials Team, School of Pharmacy, RCSI, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI, TCD, Dublin, Ireland
- Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway, Ireland
| | - Sally-Ann Cryan
- Drug Delivery and Advanced Materials Team, School of Pharmacy, RCSI, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI, TCD, Dublin, Ireland
- Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway, Ireland
| |
Collapse
|
70
|
Abstract
The objective of this study was to test the association of the variation in a 360 bp region in exon 2 of the ovine bone morphogenetic protein 4 (BMP4) gene with growth performance (birth weight, pre-weaning average daily gain, weaning weight, post-weaning average daily gain and marketing weight) and body conformational traits (height at withers, height at hips, body length, heart girth, thigh circumference, body mass index, skeletal muscle index, body index and relative body index) in 242 Barki lambs using polymerase chain reaction-single strand conformational polymorphism (PCR-SSCP). Two variants (A and B) and three genotypes (AA, AB and BB) were detected. The BMP4 genotype significantly affected (p < .05 or p < .01) post-weaning daily gain, marketing weight, height at hips, thigh circumference, body mass index and skeletal muscle index. The results provided valuable information indicating selection for the BMP4 genotype might increase growth and muscularity in Barki lambs.
Collapse
Affiliation(s)
- Adel H M Ibrahim
- Department of Animal Breeding, Desert Research Center, Cairo, Egypt
| |
Collapse
|
71
|
Ji L, Song Z, Zeng F, Hu M, Chen S, Qin Z, Xia D. [Research progress on controlled release of various growth factors in bone regeneration]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:750-755. [PMID: 31198005 PMCID: PMC8355764 DOI: 10.7507/1002-1892.201901116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/28/2019] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To summarize the research progress of controlled release of angiogenic factors and osteogenic factors in bone tissue engineering. METHODS The domestic and abroad literature on the controlled release structure of growth factors during bone regeneration in recent years was extensively reviewed and summarized. RESULTS The sustained-release structure includes direct binding, microsphere-three-dimensional scaffold structure, core-shell structure, layer self-assembly, hydrogel, and gene carrier. A sustained-release system composed of different sustained-release structures combined with different growth factors can promote bone regeneration and angiogenesis. CONCLUSION Due to its controllability and persistence, the growth factor sustained-release system has become a research hotspot in bone tissue engineering and has broad application prospects.
Collapse
Affiliation(s)
- Lin Ji
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Ziwei Song
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Fuhai Zeng
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Ming Hu
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Siqi Chen
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Zhongjie Qin
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Delin Xia
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000,
| |
Collapse
|
72
|
Scibetta AC, Morris ER, Liebowitz AB, Gao X, Lu A, Philippon MJ, Huard J. Characterization of the chondrogenic and osteogenic potential of male and female human muscle-derived stem cells: Implication for stem cell therapy. J Orthop Res 2019; 37:1339-1349. [PMID: 30667562 DOI: 10.1002/jor.24231] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/02/2019] [Indexed: 02/04/2023]
Abstract
People of all backgrounds are susceptible to bone and cartilage damage, and these injuries can be debilitating. Current treatments for bone and cartilage injuries are less than optimal, and we are interested in developing new approaches to treat these diseases, specifically using human muscle-derived stem cells (hMDSCs). Our lab previously demonstrated that sex differences exist between male and female murine MDSCs; thus, this paper sought to investigate whether sex differences also exist in hMDSCs. In the present study, we characterized the chondrogenic and osteogenic sex differences of hMDSCs in vitro and in vivo. We performed in vitro osteogenic and chondrogenic differentiation using hMDSC pellet cultures. As demonstrated by microCT, histology, and immunohistochemistry, male hMDSCs were more chondrogenic and osteogenic than their female counterparts in vitro. No differences were observed based on the sex of hMDSCs in osteogenic and chondrogenic gene expression and cell surface markers. For our in vivo study, we transduced hMDSCs with lenti-BMP2/GFP and transplanted these cells into critical-sized calvarial defects in mice. MicroCT results revealed that male hMDSCs regenerated more bone at 2 weeks and demonstrated higher bone density at 4 and 6 weeks than female hMDSCs. Histology demonstrated that both male and female hMDSCs regenerated functional bone. Clinical relevance: These studies reinforce that stem cells isolated from male and female patients differ in function, and we should disclose the sex of cells used in future studies. Considering sex differences of hMDSCs may help to improve cell-based therapies for autologous cell treatment of bone and cartilage damage. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1339-1349, 2019.
Collapse
Affiliation(s)
| | | | | | - Xueqin Gao
- Steadman Philippon Research Institute, Vail, Colorado.,Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Aiping Lu
- Steadman Philippon Research Institute, Vail, Colorado.,Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | | | - Johnny Huard
- Steadman Philippon Research Institute, Vail, Colorado.,Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
73
|
Huard J. Stem cells, blood vessels, and angiogenesis as major determinants for musculoskeletal tissue repair. J Orthop Res 2019; 37:1212-1220. [PMID: 29786150 DOI: 10.1002/jor.24058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/21/2018] [Indexed: 02/04/2023]
Abstract
This manuscript summarizes 20 years of research from my laboratories at the University of Pittsburgh and more recently, at the University of Texas Health Science Center at Houston and the Steadman Philippon Research Institute in Vail, Colorado. The discovery of muscle-derived stem cells (MDSCs) did not arise from a deliberate search to find a novel population of muscle cells with high regenerative potential, but instead was conceived in response to setbacks encountered while working in muscle cell transplantation for Duchenne muscular dystrophy (DMD). DMD is a devastating inherited X-linked muscle disease characterized by progressive muscle weakness due to lack of dystrophin expression in muscle fiber sarcolemma.1 Although the transplantation of normal myoblasts into dystrophin-deficient muscle can restore dystrophin, this approach has been hindered by limited survival (less than 1%) of the injected cells.1 The fact that 99% of the cells were not surviving implantation was seen as a major weakness with this technology by most. My research team decided to investigate which cells represent the 1% of the cells surviving post-implantation. We have subsequently confirmed that the few cells which exhibit high survival post-implantation also display stem cell characteristics, and were termed "muscle-derived stem cells" or MDSCs. Herein, I will describe the origin of these MDSCs, the mechanisms of MDSC action during tissue repair, and finally the development of therapeutic strategies to improve regeneration and repair of musculoskeletal tissues. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1212-1220, 2019.
Collapse
Affiliation(s)
- Johnny Huard
- Department of Orthopaedic Surgery, and The Brown Foundation Institute of Molecular Medicine Center for Tissue Engineering and Aging Research, McGovern Medical School, The University of Texas Health Science Center, 1881 East Road, 3SCR6.3618, Houston, Texas, 77054.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, 181 W. Meadow Drive, Suite 1000, Vail, Colorado, 81657
| |
Collapse
|
74
|
Kawakami Y, Hambright WS, Takayama K, Mu X, Lu A, Cummins JH, Matsumoto T, Yurube T, Kuroda R, Kurosaka M, Fu FH, Robbins PD, Niedernhofer LJ, Huard J. Rapamycin Rescues Age-Related Changes in Muscle-Derived Stem/Progenitor Cells from Progeroid Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:64-76. [PMID: 31312666 PMCID: PMC6610712 DOI: 10.1016/j.omtm.2019.05.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022]
Abstract
Aging-related loss of adult stem cell function contributes to impaired tissue regeneration. Mice deficient in zinc metalloproteinase STE24 (Zmpste24−/−) exhibit premature age-related musculoskeletal pathologies similar to those observed in children with Hutchinson-Gilford progeria syndrome (HGPS). We have reported that muscle-derived stem/progenitor cells (MDSPCs) isolated from Zmpste24−/− mice are defective in their proliferation and differentiation capabilities in culture and during tissue regeneration. The mechanistic target of rapamycin complex 1 (mTORC1) regulates cell growth, and inhibition of the mTORC1 pathway extends the lifespan of several animal species. We therefore hypothesized that inhibition of mTORC1 signaling would rescue the differentiation defects observed in progeroid MDSPCs. MDSPCs were isolated from Zmpste24−/− mice, and the effects of mTORC1 on MDSPC differentiation and function were examined. We found that mTORC1 signaling was increased in senescent Zmpste24−/− MDSPCs, along with impaired chondrogenic, osteogenic, and myogenic differentiation capacity versus wild-type MDSPCs. Interestingly, we observed that mTORC1 inhibition with rapamycin improved myogenic and chondrogenic differentiation and reduced levels of apoptosis and senescence in Zmpste24−/− MDSPCs. Our results demonstrate that age-related adult stem/progenitor cell dysfunction contributes to impaired regenerative capacities and that mTORC1 inhibition may represent a potential therapeutic strategy for improving differentiation capacities of senescent stem and muscle progenitor cells.
Collapse
Affiliation(s)
- Yohei Kawakami
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - William S Hambright
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Koji Takayama
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Xiaodong Mu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Aiping Lu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - James H Cummins
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takashi Yurube
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Freddie H Fu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA
| | - Johnny Huard
- Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
75
|
Interleukin-10 Does Not Augment Osseous Regeneration in the Scarred Calvarial Defect Achieved with Low-Dose Biopatterned BMP2. Plast Reconstr Surg 2019; 143:1215e-1223e. [PMID: 31136482 DOI: 10.1097/prs.0000000000005640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Large calvarial defects represent a major reconstructive challenge, as they do not heal spontaneously. Infection causes inflammation and scarring, further reducing the healing capacity of the calvaria. Bone morphogenetic protein-2 (BMP2) has been shown to stimulate osteogenesis but has significant side effects in high doses. BMP2 has not been tested in combination with antiinflammatory cytokines such as interleukin-10. METHODS Sixteen New Zealand White rabbits underwent 15 × 15-mm flap calvarectomies. The flap was incubated in Staphylococcus aureus and replaced, and infection and scarring were allowed to develop. The flap was subsequently removed and the wound débrided. A 15 × 15-mm square of acellular dermal matrix biopatterned with low-dose BMP2, interleukin-10, or a combination was implanted. Computed tomographic scans were taken over 42 days. Rabbits were then killed and histology was performed. RESULTS Defects treated with BMP2 showed significantly (p < 0.05) greater osseous regeneration than untreated controls. Interleukin-10 did not significantly augment the healing achieved with BMP2, and interleukin-10 alone did not significantly increase healing compared with controls. Histology showed evidence of bone formation in defects treated with BMP2. Untreated controls and defects treated with interleukin-10 alone showed only fibrous tissue in the defect site. CONCLUSIONS Low-dose BMP2 delivered directly to the scarred calvarial defect augments bony healing. Interleukin-10 at the dose applied did not significantly augment healing alone or in combination with BMP2. Healing had not finished at 42 days and analysis at later time points or the use of higher doses of BMP2 may yield greater healing.
Collapse
|
76
|
Pizzicannella J, Gugliandolo A, Orsini T, Fontana A, Ventrella A, Mazzon E, Bramanti P, Diomede F, Trubiani O. Engineered Extracellular Vesicles From Human Periodontal-Ligament Stem Cells Increase VEGF/VEGFR2 Expression During Bone Regeneration. Front Physiol 2019; 10:512. [PMID: 31114512 PMCID: PMC6503111 DOI: 10.3389/fphys.2019.00512] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/11/2019] [Indexed: 01/15/2023] Open
Abstract
Bone regeneration represents still a challenge, in particular for calvarium defects. Recently, the development of biomaterials with the addiction of stem cells is giving promising results for the treatment of bone defects. In particular, it was demonstrated that scaffolds enriched with mesenchymal stem cells (MSCs) and/or their derivatives, such as conditioned medium (CM) and extracellular vesicles (EVs), may improve bone regeneration. Moreover, given the deep link between osteogenesis and angiogenesis, a successful approach must also take into consideration the development of vascularization. In this work we evaluated the bone regeneration capacity of a collagen membrane (3D-COL) enriched with human periodontal-ligament stem cells (hPDLSCs) and CM or EVs or EVs engineered with polyethylenimine (PEI-EVs) in rats subjected to a calvarial defect. We evaluated also their capacity to induce angiogenic factors. At first, in vitro results showed an increased expression of osteogenic markers in hPDLSCs cultured with the 3D-COL and PEI-EVs, associated also with the increased protein levels of Vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2). The increased expression of these proteins was confirmed also in vivo in rats implanted with the 3D-COL enriched with hPDLSCs and PEI-EVs. Moreover, histological examination evidenced in this group of rats the activation of bone regeneration and of the vascularization process. Also MicroCT imaging with morphometric analysis confirmed in rats transplanted with 3D-COL enriched with hPDLSCs and PEI-EVs an important regenerative process and a better integration level. All together, these results evidenced that the 3D-COL enriched with hPDLSCs and PEI-EVs may promote bone regeneration of calvaria defects, associated also with an increased vascularization.
Collapse
Affiliation(s)
- Jacopo Pizzicannella
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | - Tiziana Orsini
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Antonella Fontana
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Alessia Ventrella
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | | | - Francesca Diomede
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Oriana Trubiani
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
77
|
Sharma S, Xue Y, Xing Z, Yassin MA, Sun Y, Lorens JB, Finne-Wistrand A, Sapkota D, Mustafa K. Adenoviral mediated mono delivery of BMP2 is superior to the combined delivery of BMP2 and VEGFA in bone regeneration in a critical-sized rat calvarial bone defect. Bone Rep 2019; 10:100205. [PMID: 31193299 PMCID: PMC6525280 DOI: 10.1016/j.bonr.2019.100205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/11/2019] [Accepted: 04/10/2019] [Indexed: 01/30/2023] Open
Abstract
Apart from osteogenesis, neovascularization of the defect area is an important determinant for successful bone healing. Accordingly, several studies have employed the combined delivery of VEGFA and BMP2 for bone regeneration. Nevertheless, the outcomes of these studies are highly variable. The aim of our study was to compare the effectiveness of adenoviral mediated delivery of BMP2 alone and in combination with VEGFA in rat bone marrow stromal cells (rBMSC) seeded on a poly(LLA-co-CL) scaffold in angiogenesis and osteogenesis using a critical-sized rat calvarial defect model. Both mono delivery of BMP2 and the combined delivery of a lower ratio of VEGFA and BMP2 (1:4) led to up-regulation of osteogenic genes (Alpl and Runx2) and increased calcium deposition in vitro, compared with the GFP control. Micro computed tomography (microCT) analysis of the rat calvarial defect at 8 weeks showed that the mono delivery of BMP2 (43.37 ± 3.55% defect closure) was the most effective in healing the bone defect, followed by the combined delivery of BMP2 and VEGFA (27.86 ± 2.89%) and other controls. Histological and molecular analyses supported the microCT findings. Analysis of the angiogenesis, however, showed that both mono delivery of BMP2 and combined delivery of BMP2 and VEGFA had similar angiogenic effect in the calvarial defects. Examination of the key genes related to host response against the adenoviral vectors showed that the current model system was not associated with adverse immune response. Overall, the results show that the mono delivery of BMP2 was superior to the combined delivery of BMP2 and VEGFA in healing the critical-sized rat calvarial bone defect. These findings underscore the importance of appropriate growth factor combination for the successful outcome in bone regeneration.
Collapse
Affiliation(s)
- Sunita Sharma
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Ying Xue
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Zhe Xing
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Mohammed A Yassin
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yang Sun
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - James B Lorens
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Dipak Sapkota
- Department of Oral Biology, Faculty of Dentistry, 0316 Oslo, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
78
|
Kim I, Lee SS, Kim SHL, Bae S, Lee H, Hwang NS. Osteogenic Effects of VEGF-Overexpressed Human Adipose-Derived Stem Cells with Whitlockite Reinforced Cryogel for Bone Regeneration. Macromol Biosci 2019; 19:e1800460. [PMID: 30821921 DOI: 10.1002/mabi.201800460] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Bone is a vascularized tissue that is comprised of collagen fibers and calcium phosphate crystals such as hydroxyapatite (HAp) and whitlockite (WH). HAp and WH are known to elicit bone regeneration by stimulating osteoblast activities and osteogenic commitment of stem cells. In addition, vascular endothelial growth factor (VEGF) is shown to promote osteogenesis and angiogenesis which is considered as an essential process in bone repair by providing nutrients. In this study, VEGF-secreting human adipose-derived stem cells (VEGF-ADSCs) are developed by transducing ADSCs with VEGF-encoded lentivirus. Additionally, WH-reinforced gelatin/heparin cryogels (WH-C) are fabricated by loading WH into gelatin/heparin cryogels. VEGF-ADSC secrete tenfold more VEGF than ADSC and show increased VEGF secretion with cell growth. Also, incorporation of WH into cryogels provides a mineralized environment with ions secreted from WH. When the VEGF-ADSCs are seeded on WH-C, sustained release of VEGF is observed due to the specific affinity of VEGF to heparin. Finally, the synergistic effect of VEGF-ADSC and WH on osteogenesis is successfully confirmed by alkaline phosphatase and real-time polymerase chain reaction analysis. In vivo bone formation is demonstrated via implantation of VEGF-ADSC seeded WH-C into mouse calvarial bone defect model, resulted in enhanced bone development with the highest bone volume/total volume.
Collapse
Affiliation(s)
- Inseon Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seunghun S Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Hyun L Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sunghoon Bae
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hoyon Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea.,Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.,BioMax Institute of Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
79
|
Reconstruction of a Calvarial Wound Complicated by Infection: Comparing the Effects of Biopatterned Bone Morphogenetic Protein 2 and Vascular Endothelial Growth Factor. J Craniofac Surg 2019; 30:260-264. [PMID: 30339591 DOI: 10.1097/scs.0000000000004779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP2) bioprinted on biological matrix induces osseous regeneration in large calvarial defects in rabbits, both uncomplicated and scarred. Healing in unfavorable defects scarred from previous infection is decreased due in part to the lack of vascularity. This impedes the access of mesenchymal stem cells, key to osseous regeneration and the efficacy of BMP2, to the wound bed. The authors hypothesized that bioprinted vascular endothelial growth factor (VEGF) would augment the osseous regeneration achieved with low dose biopatterned BMP2 alone. Thirteen New Zealand white rabbits underwent subtotal calvariectomy using a dental cutting burr. Care was taken to preserve the underlying dura. A 15 mm × 15 mm flap of bone was cut away and incubated in a 1 × 108 cfu/mL planktonic solution of S aureus before reimplantation. After 2 weeks of subsequent infection the flap was removed and the surgical wound debrided followed by 10 days of antibiotic treatment. On postoperative day 42 the calvarial defects were treated with acellular dermal matrix bioprinted with nothing (control), VEGF, BMP2, BMP2/VEGF combined. Bone growth was analyzed with serial CT and postmortem histology. Defects treated with BMP2 (BMP2 alone and BMP2/VEGF combination) showed significantly greater healing than control and VEGF treated defect (P < 0.5). Vascular endothelial growth factor treated defect demonstrated less healing than control and VEGF/BMP2 combination treatments achieved less healing than BMP2 alone though these differences were nonsignificant. Low dose BMP2-patterned acellular dermal matrix improves healing of scarred calvarial defects. Vascular endothelial growth factor at the doses applied in this study failed to increase healing.
Collapse
|
80
|
Biz C, Crimi A, Fantoni I, Pozzuoli A, Ruggieri P. Muscle stem cells: what's new in orthopedics? ACTA BIO-MEDICA : ATENEI PARMENSIS 2019; 90:8-13. [PMID: 30714993 PMCID: PMC6503412 DOI: 10.23750/abm.v90i1-s.8078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIM OF THE WORK Adult stem cells were studied as a source of potentially useful development for tissue engineering and repair techniques. The aim of this review is to clarify the actual and possible uses of muscle stem cells in orthopedics. METHODS A selection of studies was made to obtain a homogeneous and up to date overview on the muscle stem cells applications. RESULTS In recent years muscle was studied as a good source of adult stem cells that can differentiate into different cell lineages. Muscle stem cells are a heterogeneous population of cells, which demonstrated in vitro a great potential for the regeneration and repair of muscle, bone and cartilage tissue. Among muscle stem cells, satellite stem cells are the most known progenitor cells: they can differentiate in osteoblasts, adipocytes, chondrocytes and myocytes. CONCLUSIONS Although muscle stem cells are a promising field of research, more pre-clinical studies in animal models are still needed to determine the safety and efficiency of the transplant procedures in humans.
Collapse
Affiliation(s)
- Carlo Biz
- Orthopaedic Clinic, Department of Surgery, Oncology and Gastroenterology DiSCOG, University of Padua, Padova, Italy.
| | | | | | | | | |
Collapse
|
81
|
Liu T, Li B, Zheng XF, Jiang SD, Zhou ZZ, Xu WN, Zheng HL, Wang CD, Zhang XL, Jiang LS. Chordin-Like 1 Improves Osteogenesis of Bone Marrow Mesenchymal Stem Cells Through Enhancing BMP4-SMAD Pathway. Front Endocrinol (Lausanne) 2019; 10:360. [PMID: 31249554 PMCID: PMC6582276 DOI: 10.3389/fendo.2019.00360] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/21/2019] [Indexed: 11/13/2022] Open
Abstract
Chordin-like 1 (CHRDL1) is a secreted glycoprotein with repeated cysteine-rich domains, which can bind to BMPs family ligands. Although it has been reported to play important roles in several systems, the exact roles of CHRDL1 on human bone mesenchymal stem cells (hBMSCs) osteogenesis remain to be explored. The present study aimed to investigate the roles of CHRDL1 on the osteogenic differentiation of hBMSCs and the underlying molecular mechanisms. We found that CHRDL1 was upregulated during hBMSCs osteogenesis, and rhBMP-4 administration could enhance CHRDL1 mRNA expression in a dose and time dependent manner. Knockdown of CHRDL1 did not affect hBMSCs proliferation, but inhibited the BMP-4-dependent osteogenic differentiation, showing decreased mRNA expression levels of osteogenic markers and reduced mineralization. On the contrary, overexpression of CHRDL1 enhanced BMP-4 induced osteogenic differentiation of hBMSCs. Moreover, in vivo experiments by transplanting CHRDL1 gene modified hBMSCs into nude mice defective femur models displayed higher new bone formation in CHRDL1 overexpression groups, but lower new bone formation in CHRDL1 knockdown groups, compared with control groups. In consistent with the bone formation rate, there were increased CHRDL1 protein expression in new bone formation regions of defective femur in CHRDL1 overexpression groups, while reduced CHRDL1 protein expression in CHRDL1 knockdown groups compared with control groups. These indicate that CHRDL1 can promote osteoblast differentiation in vivo. Furthermore, the mechanisms study showed that CHRDL1 improved BMP-4 induced phosphorylation of SMAD1/5/9 during osteogenic differentiation of hBMSCs. Besides, promotion of osteogenic differentiation and the activation of SMAD phosphorylation by CHRDL1 can be blocked by BMP receptor type I inhibitor LDN-193189. In conclusion, our results suggested that CHRDL1 can promote hBMSCs osteogenic differentiation through enhancing the activation of BMP-4-SMAD1/5/9 pathway.
Collapse
Affiliation(s)
- Tao Liu
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Li
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin-Feng Zheng
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng-Dan Jiang
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Zhu Zhou
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ning Xu
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huo-Liang Zheng
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuan-Dong Wang
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Ling Zhang
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiao-Ling Zhang
| | - Lei-Sheng Jiang
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lei-Sheng Jiang
| |
Collapse
|
82
|
Shahzadi L, Chaudhry AA, Aleem AR, Malik MH, Ijaz K, Akhtar H, Alvi F, Khan AF, Rehman IU, Yar M. Development of K-doped ZnO nanoparticles encapsulated crosslinked chitosan based new membranes to stimulate angiogenesis in tissue engineered skin grafts. Int J Biol Macromol 2018; 120:721-728. [DOI: 10.1016/j.ijbiomac.2018.08.103] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 08/10/2018] [Accepted: 08/21/2018] [Indexed: 11/30/2022]
|
83
|
Lu X, Li K, Xie Y, Qi S, Shen Q, Yu J, Huang L, Zheng X. Improved osteogenesis of boron incorporated calcium silicate coatings via immunomodulatory effects. J Biomed Mater Res A 2018; 107:12-24. [PMID: 29781148 DOI: 10.1002/jbm.a.36456] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/02/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023]
Abstract
Osteoimmunology has revealed the importance of a favorable immune response for successful biomaterial-mediated osteogenesis. Boron-incorporated calcium silicate (Ca11 Si4 B2 O22 , B-CS) coating has been reported as a potential candidate for improving osteogenesis in orthopedic applications in vitro. However, relatively little is known about its effects on the immune response and subsequent osteogenesis. In this work, the immunomodulatory properties of the B-CS coating and its specific mechanism of action were explored. We found that the B-CS coating decreased M1 polarization and converted macrophages to the M2 phenotype via restraining the toll-like receptor signaling pathway, thus inducing a significant reduction in pro-inflammatory cytokines and an increase in anti-inflammatory cytokines. Moreover, the B-CS coating inhibited osteoclastogenesis and osteoclastic activities by downregulating osteoclastogenic genes and inhibiting the RANKL/RANK system. BMP2 and VEGF were also significantly upregulated by macrophages and bone mesenchymal stem cells, leading to activation of the BMP2 signaling pathway and subsequent upregulation of osteogenesis-associated genes, finally promoting osteogenic differentiation. These findings show that the B-CS coating could be a promising coating material for hip and knee implants. Furthermore, incorporation of the element boron into bioceramic coatings could be a good strategy in the design of bone biomaterials with beneficial immune responses. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 12-24, 2019.
Collapse
Affiliation(s)
- Xiang Lu
- Key Laboratory of Inorganic Coating Materials, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, People's Republic of China.,University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, People's Republic of China
| | - Kai Li
- Key Laboratory of Inorganic Coating Materials, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, People's Republic of China
| | - Youtao Xie
- Key Laboratory of Inorganic Coating Materials, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, People's Republic of China
| | - Shengcai Qi
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China
| | - Qingyi Shen
- Department of Prosthodontics, Shanghai Stomatological Disease Center, Shanghai, 200031, People's Republic of China
| | - Jiangming Yu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Liping Huang
- Key Laboratory of Inorganic Coating Materials, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, People's Republic of China
| | - Xuebin Zheng
- Key Laboratory of Inorganic Coating Materials, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, People's Republic of China
| |
Collapse
|
84
|
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: 19] [Impact Index Per Article: 3.2] [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
|
85
|
Ding D, Zhu Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1041-1060. [DOI: 10.1016/j.msec.2017.12.036] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/18/2017] [Accepted: 12/30/2017] [Indexed: 01/06/2023]
|
86
|
Controllable and durable release of BMP-2-loaded 3D porous sulfonated polyetheretherketone (PEEK) for osteogenic activity enhancement. Colloids Surf B Biointerfaces 2018; 171:668-674. [DOI: 10.1016/j.colsurfb.2018.08.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 01/07/2023]
|
87
|
Wang X, Wang G, Zingales S, Zhao B. Biomaterials Enabled Cell-Free Strategies for Endogenous Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:463-481. [PMID: 29897021 DOI: 10.1089/ten.teb.2018.0012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Repairing bone defects poses a major orthopedic challenge because current treatments are constrained by the limited regenerative capacity of human bone tissue. Novel therapeutic strategies, such as stem cell therapy and tissue engineering, have the potential to enhance bone healing and regeneration, and hence may improve quality of life for millions of people. However, the ex vivo expansion of stem cells and their in vivo delivery pose technical difficulties that hamper clinical translation and commercial development. A promising alternative to cell delivery-based strategies is to stimulate or augment the inherent self-repair mechanisms of the patient to promote endogenous restoration of the lost/damaged bone. There is growing evidence indicating that increasing the endogenous regenerative potency of bone tissues for therapeutics will require the design and development of new generations of biomedical devices that provide key signaling molecules to instruct cell recruitment and manipulate cell fate for in situ tissue regeneration. Currently, a broad range of biomaterial-based deployment technologies are becoming available, which allow for controlled spatial presentation of biological cues required for endogenous bone regeneration. This article aims to explore the proposed concepts and biomaterial-enabled strategies involved in the design of cell-free endogenous techniques in bone regenerative medicine.
Collapse
Affiliation(s)
- Xiaojing Wang
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| | - Guowei Wang
- 3 Department of Stomatology, Laoshan Branch of No. 401 Hospital of the Chinese Navy , Qingdao, Shandong, P.R. China
| | - Sarah Zingales
- 4 Department of Chemistry and Biochemistry, Georgia Southern University , Savannah, Georgia
| | - Baodong Zhao
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| |
Collapse
|
88
|
Abstract
PURPOSE OF REVIEW To assess the utilization and efficacy of platelet-rich plasma (PRP), for the treatment of articular cartilage injury, most commonly characterized by progressive pain and loss of joint function in the setting of osteoarthritis (OA). RECENT FINDINGS PRP modulates the inflammatory and catabolic environment through a locally applied concentrate of platelets, leukocytes, and growth factors. Clinically, PRP has been shown to be possibly a viable treatment adjuvant for a variety of inflammatory and degenerative conditions. Recent efforts have focused on optimizing delivery methods that enable platelets to slowly degranulate their biological constituents, which may promote healing and improve OA symptoms for a longer duration. There are various factors that affect the progression of OA within joints, including inhibition of inflammatory cytokines and altering the level of enzymatic expression. PRP therapy aims to mediate inflammatory and catabolic factors in a degenerative environment through the secretion of anti-inflammatory factors and chemotaxic effects. There are a growing number of studies that have demonstrated the clinical benefit of PRP for non-operative management of OA. Additional randomized controlled trials with long-term follow-up are needed in order to validate PRP's therapeutic efficacy in this setting. Additionally, continued basic research along with well-designed pre-clinical studies and reporting standards are necessary in order to clarify the effectiveness of PRP for cartilage repair and regeneration for future clinical applications.
Collapse
|
89
|
Orth M, Altmeyer M, Scheuer C, Braun B, Holstein J, Eglin D, D'Este M, Histing T, Laschke M, Pohlemann T, Menger M. Effects of locally applied adipose tissue-derived microvascular fragments by thermoresponsive hydrogel on bone healing. Acta Biomater 2018; 77:201-211. [PMID: 30030175 DOI: 10.1016/j.actbio.2018.07.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/13/2018] [Accepted: 07/16/2018] [Indexed: 12/17/2022]
Abstract
Insufficient vascularization is a major cause for the development of non-unions. To overcome this problem, adipose tissue-derived microvascular fragments (MVF) may serve as vascularization units. However, their application into bone defects needs a carrier system. Herein, we analyzed whether this is achieved by a thermoresponsive hydrogel (TRH). MVF were isolated from CD-1 mice and cultivated after incorporation into TRH, while non-incorporated MVF served as controls. Viability of MVF was assessed immunohistochemically over a 7-day period. Moreover, osteotomies were induced in femurs of CD-1 mice. The osteotomy gaps were filled with MVF-loaded TRH (TRH + MVF), unloaded TRH (TRH) or no material (control). Bone healing was evaluated 14 and 35 days postoperatively. MVF incorporated into TRH exhibited less apoptotic cells and showed a stable vessel morphology compared to controls. Micro-computed tomography revealed a reduced bone volume in TRH + MVF femurs. Histomorphometry showed less bone and more fibrous tissue after 35 days in TRH + MVF femurs compared to controls. Accordingly, TRH + MVF femurs exhibited a lower osseous bridging score and a reduced bending stiffness. Histology and Western blot analysis revealed an increased vascularization and CD31 expression, whereas vascular endothelial growth factor (VEGF) expression was reduced in TRH + MVF femurs. Furthermore, the callus of TRH + MVF femurs showed increased receptor activator of NF-κB ligand expression and higher numbers of osteoclasts. These findings indicate that TRH is an appropriate carrier system for MVF. Application of TRH + MVF increases the vascularization of bone defects. However, this impairs bone healing, most likely due to lower VEGF expression during the early course of bone healing. STATEMENT OF SIGNIFICANCE In the present study we analyzed for the first time the in vivo performance of a thermoresponsive hydrogel (TRH) as a delivery system for bioactive microvascular fragments (MVF). We found that TRH represents an appropriate carrier for MVF as vascularization units and maintains their viability. Application of MVF-loaded TRH impaired bone formation in an established murine model of bone healing, although vascularization was improved. This unexpected outcome was most likely due to a reduced VEGF expression in the early phase bone healing.
Collapse
|
90
|
Betz VM, Ren B, Messmer C, Jansson V, Betz OB, Müller PE. Bone morphogenetic protein-2 is a stronger inducer of osteogenesis within muscle tissue than heterodimeric bone morphogenetic protein-2/6 and -2/7: Implications for expedited gene-enhanced bone repair. J Gene Med 2018; 20:e3042. [DOI: 10.1002/jgm.3042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 11/09/2022] Open
Affiliation(s)
- Volker M. Betz
- Department of Gene Therapy; University of Ulm; Ulm Germany
- Center for Rehabilitation; RKU - University and Rehabilitation Hospitals Ulm; Ulm Germany
| | - Bin Ren
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
| | - Carolin Messmer
- Center for Rehabilitation; RKU - University and Rehabilitation Hospitals Ulm; Ulm Germany
| | - Volkmar Jansson
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
| | - Oliver B. Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA USA
| | - Peter E. Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
| |
Collapse
|
91
|
Betz VM, Kochanek S, Rammelt S, Müller PE, Betz OB, Messmer C. Recent advances in gene-enhanced bone tissue engineering. J Gene Med 2018; 20:e3018. [PMID: 29601661 DOI: 10.1002/jgm.3018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/18/2018] [Accepted: 03/18/2018] [Indexed: 12/13/2022] Open
Abstract
The loss of bone tissue represents a critical clinical condition that is frequently faced by surgeons. Substantial progress has been made in the area of bone research, providing insight into the biology of bone under physiological and pathological conditions, as well as tools for the stimulation of bone regeneration. The present review discusses recent advances in the field of gene-enhanced bone tissue engineering. Gene transfer strategies have emerged as highly effective tissue engineering approaches for supporting the repair of the musculoskeletal system. By contrast to treatment with recombinant proteins, genetically engineered cells can release growth factors at the site of injury over extended periods of time. Of particular interest are the expedited technologies that can be applied during a single surgical procedure in a cost-effective manner, allowing translation from bench to bedside. Several promising methods based on the intra-operative genetic manipulation of autologous cells or tissue fragments have been developed in preclinical studies. Moreover, gene therapy for bone regeneration has entered the clinical stage with clinical trials for the repair of alveolar bone. Current trends in gene-enhanced bone engineering are also discussed with respect to the movement of the field towards expedited, translational approaches. It is possible that gene-enhanced bone tissue engineering will become a clinical reality within the next few years.
Collapse
Affiliation(s)
- Volker M Betz
- Department of Gene Therapy, University of Ulm, Ulm, Germany.,Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| | | | - Stefan Rammelt
- University Center of Orthopedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany
| | - Peter E Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver B Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carolin Messmer
- Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| |
Collapse
|
92
|
Zhang C, Meng C, Guan D, Ma F. BMP2 and VEGF165 transfection to bone marrow stromal stem cells regulate osteogenic potential in vitro. Medicine (Baltimore) 2018; 97:e9787. [PMID: 29384874 PMCID: PMC5805446 DOI: 10.1097/md.0000000000009787] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An exogenous supply of bone morphogenetic protein 2 (BMP2) and vascular endothelial growth factors 165 (VEGF165) will synergize to promote bone regeneration in vivo. The aim of this study was to confirm the role of VEGF165 on the osteogenesis potential of bone mesenchymal stem cells (BMSCs) transduced by adenovirus vector containing BMP2 gene in vitro.Rabbit BMSCs were isolated and transfected with various adenovirus vectors: Ad-BMP2-VEGF165 (BMP2+VEGF165 group), Ad-BMP2 (BMP2 group), Ad-VEGF165 (VEGF165 group), and Ad-green fluorescent protein (GFP group). The multiplicity of infection was detected by GFP expression. Expression of BMP2 and VEGF165 was detected by Western blot and ELISA, and the osteogenic biological activity of BMP2 and VEGF165 by osteogenic assay. Meanwhile, the osteogenic biological activity of BMP2 and VEGF165 was evaluated by detection of Col I (collagen type I), OC (osteocalcin), and ALP (alkaline phosphatase) activity using OC staining, ALP activity assay, and real-time PCR assay.Expression of target genes and proteins reached peak values at 5 days and then gradually declined. The OC staining, ALP activity, and real-time PCR assay of ColI, OC, and ALP were all increased in cells transfected with Ad-BMP2-VEGF165, Ad-BMP2, Ad-VEGF165, and Ad-GFP. However, the osteogenic biological activity in cells transfected with Ad-BMP2 was higher compared to cells transfected with other vectors after transfection at 14 and 21 days. We also found that BMP2 +VEGF165 group showed more osteogenic activity effect than the VEGF165 or control group. Furthermore, osteogenic assays in VEGF165 showed that a slightly lower osteogenic effect when compared to controls at 21 days.VEGF165 might be a potent inhibitor of BMSCs differentiation into osteoblasts. The strategies to use BMP2 and VEGF165 in bone regeneration and the molecular mechanism of their interaction require further investigation.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Spine Surgery, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu
- Department of Orthopedics, Affiliated Hospital of Jining Medical University, Guhuai Road, Jining, Shandong
| | - Chunyang Meng
- Department of Orthopedics, Affiliated Hospital of Jining Medical University, Guhuai Road, Jining, Shandong
| | - Dafan Guan
- Department of Orthopedics, Ankang Central Hospital, Ankang, Shanxi, China
| | - Fengyu Ma
- Department of Orthopedics, Affiliated Hospital of Jining Medical University, Guhuai Road, Jining, Shandong
| |
Collapse
|
93
|
Kishan A, Walker T, Sears N, Wilems T, Cosgriff-Hernandez E. Winner of the society for biomaterials student award in the Ph.D. category for the annual meeting of the society for biomaterials, april 11-14, 2018, Atlanta, GA: Development of a bimodal, in situ crosslinking method to achieve multifactor release from el. J Biomed Mater Res A 2018; 106:1155-1164. [DOI: 10.1002/jbm.a.36342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/10/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Alysha Kishan
- Department of Biomedical Engineering; Texas A&M University; College Station Texas 77843
| | - Taneidra Walker
- Department of Biomedical Engineering; The University of Texas at Austin; Austin Texas 78712
| | - Nick Sears
- Department of Biomedical Engineering; Texas A&M University; College Station Texas 77843
| | - Thomas Wilems
- Department of Biomedical Engineering; The University of Texas at Austin; Austin Texas 78712
| | | |
Collapse
|
94
|
Matthias N, Hunt SD, Wu J, Lo J, Smith Callahan LA, Li Y, Huard J, Darabi R. Volumetric muscle loss injury repair using in situ fibrin gel cast seeded with muscle-derived stem cells (MDSCs). Stem Cell Res 2018; 27:65-73. [PMID: 29331939 PMCID: PMC5851454 DOI: 10.1016/j.scr.2018.01.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/02/2017] [Accepted: 01/05/2018] [Indexed: 12/22/2022] Open
Abstract
Volumetric muscle defect, caused by trauma or combat injuries, is a major health concern leading to severe morbidity. It is characterized by partial or full thickness loss of muscle and its bio-scaffold, resulting in extensive fibrosis and scar formation. Therefore, the ideal therapeutic option is to use stem cells combined with bio-scaffolds to restore muscle. For this purpose, muscle-derived stem cells (MDSCs) are a great candidate due to their unique multi-lineage differentiation potential. In this study, we evaluated the regeneration potential of MDSCs for muscle loss repair using a novel in situ fibrin gel casting. Muscle defect was created by a partial thickness wedge resection in the tibialis anterior (TA)muscles of NSG mice which created an average of 25% mass loss. If untreated, this defect leads to severe muscle fibrosis. Next, MDSCs were delivered using a novel in situ fibrin gel casting method. Our results demonstrated MDSCs are able to engraft and form new myofibers in the defect when casted along with fibrin gel. LacZ labeled MDSCs were able to differentiate efficiently into new myofibers and significantly increase muscle mass. This was also accompanied by significant reduction of fibrotic tissue in the engrafted muscles. Furthermore, transplanted cells also contributed to new vessel formation and satellite cell seeding. These results confirmed the therapeutic potential of MDSCs and feasibility of direct in situ casting of fibrin/MDSC mixture to repair muscle mass defects.
Collapse
Affiliation(s)
- Nadine Matthias
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Samuel D Hunt
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Jianbo Wu
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Jonathan Lo
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Laura A Smith Callahan
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States; The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States; Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States; Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Yong Li
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States; Department of Pediatric Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Johnny Huard
- Department of Orthopedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Radbod Darabi
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States; The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States.
| |
Collapse
|
95
|
A review of the cellular and molecular effects of extracorporeal shockwave therapy. Vet Comp Orthop Traumatol 2017; 29:99-107. [DOI: 10.3415/vcot-15-04-0057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/15/2015] [Indexed: 12/31/2022]
Abstract
SummaryExtracorporeal shockwave therapy (ESWT) is a novel therapeutic modality and its use in promoting connective tissue repair and analgesic effect has been advocated in the literature. It is convenient, cost-effective, and has negligible complications; it therefore bypasses many of the problems associated with surgical interventions. This paper reviews the proposed mechanisms of action in promoting tissue repair and regeneration as well as analysing its efficacy providing an analgesic effect in clinical applications. Further research will be required to not only identify the underlying mechanisms more precisely, but will also be critical for ensuring consistency across the literature so that the most beneficial treatment protocol can be developed. Extracorporeal shockwave therapy stands as a promising alternative modality in promoting tissue repair.
Collapse
|
96
|
Huang H, Cheng WX, Hu YP, Chen JH, Zheng ZT, Zhang P. Relationship between heterotopic ossification and traumatic brain injury: Why severe traumatic brain injury increases the risk of heterotopic ossification. J Orthop Translat 2017; 12:16-25. [PMID: 29662775 PMCID: PMC5866497 DOI: 10.1016/j.jot.2017.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/12/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023] Open
Abstract
Heterotopic ossification (HO) is a pathological phenomenon in which ectopic lamellar bone forms in soft tissues. HO involves many predisposing factors, including congenital and postnatal factors. Postnatal HO is usually induced by fracture, burn, neurological damage (brain injury and spinal cord injury) and joint replacement. Recent studies have found that patients who suffered from bone fracture combined with severe traumatic brain injury (S-TBI) are at a significantly increased risk for HO occurrence. Thus, considerable research focused on the influence of S-TBI on fracture healing and bone formation, as well as on the changes in various osteogenic factors with S-TBI occurrence. Brain damage promotes bone formation, but the exact mechanisms underlying bone formation and HO after S-TBI remain to be clarified. Hence, this article summarises the findings of previous studies on the relationship between S-TBI and HO and discusses the probable causes and mechanisms of HO caused by S-TBI. The translational potential of this article: A better understanding of the probable causes of traumatic brain injury-induced HO can provide new perspectives and ideas in preventing HO and may support to design more targeted therapies to reduce HO or enhance the bone formation.
Collapse
Affiliation(s)
- Huan Huang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wen-Xiang Cheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Ping Hu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian-Hai Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zheng-Tan Zheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Peng Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
97
|
Murphy MP, Quarto N, Longaker MT, Wan DC. * Calvarial Defects: Cell-Based Reconstructive Strategies in the Murine Model. Tissue Eng Part C Methods 2017; 23:971-981. [PMID: 28825366 DOI: 10.1089/ten.tec.2017.0230] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Calvarial defects pose a continued clinical dilemma for reconstruction. Advancements within the fields of stem cell biology and tissue engineering have enabled researchers to develop reconstructive strategies using animal models. We review the utility of various animal models and focus on the mouse, which has aided investigators in understanding cranial development and calvarial bone healing. The murine model has also been used to study regenerative approaches to critical-sized calvarial defects, and we discuss the application of stem cells such as bone marrow-derived mesenchymal stromal cells, adipose-derived stromal cells, muscle-derived stem cells, and pluripotent stem cells to address deficient bone in this animal. Finally, we highlight strategies to manipulate stem cells using various growth factors and inhibitors and ultimately how these factors may prove crucial in future advancements within calvarial reconstruction using native skeletal stem cells.
Collapse
Affiliation(s)
- Matthew P Murphy
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Natalina Quarto
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| |
Collapse
|
98
|
Vascular Endothelial Growth Factor Induction of Muscle-Derived Stem Cells Enhances Vascular Phenotype While Preserving Myogenic Potential. Ann Plast Surg 2017; 79:404-409. [DOI: 10.1097/sap.0000000000001147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
99
|
Xue J, Lin H, Bean A, Tang Y, Tan J, Tuan RS, Wang B. One-Step Fabrication of Bone Morphogenetic Protein-2 Gene-Activated Porous Poly-L-Lactide Scaffold for Bone Induction. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:50-59. [PMID: 29018836 PMCID: PMC5626914 DOI: 10.1016/j.omtm.2017.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 08/31/2017] [Indexed: 01/18/2023]
Abstract
Bone morphogenetic protein 2 (BMP2) is an efficacious inducer for the osteogenesis of mesenchymal stem cells (MSCs). Conventional applications of BMP2 have involved either the direct incorporation of BMP2 protein or ex vivo BMP2 gene transfer into stem cells prior to their transplantation. These approaches are able to promote bone formation to some extent; however, they are hampered by either the lack of stability and sustainability of BMP2 protein or the time-consuming and cost-prohibitive in vitro cell culture procedure. To overcome these limitations, we have developed a gene-activated poly-L-lactide acid (PLLA) scaffold with the encapsulation of recombinant adeno-associated viral (AAV) vector encoding a full-length cDNA of human BMP2 using an ice-based microparticle porogenization method that was recently developed. Results showed continuous release of AAV particles from the micropores of scaffolds for up to 1 week, subsequently transducing embedded human MSCs and producing functional BMP2. MSCs within scaffolds underwent efficacious osteogenesis, on the basis of osteoinductive gene expression and osteogenic differentiation, which resulted in robust new bone formation in vivo at 4 weeks. These findings show the potential of the technology toward developing clinical applications of a rapid, cost-effective, and potentially point-of-care approach for the repair of bone defects.
Collapse
Affiliation(s)
- Jingwen Xue
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Hang Lin
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Allison Bean
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Ying Tang
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Jian Tan
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
100
|
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: 65] [Impact Index Per Article: 9.3] [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
|