151
|
BMP-2 Grafted nHA/PLGA Hybrid Nanofiber Scaffold Stimulates Osteoblastic Cells Growth. BIOMED RESEARCH INTERNATIONAL 2015; 2015:281909. [PMID: 26539477 PMCID: PMC4619782 DOI: 10.1155/2015/281909] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/22/2015] [Indexed: 02/05/2023]
Abstract
Biomaterials play a pivotal role in regenerative medicine, which aims to regenerate and replace lost/degenerated tissues or organs. Natural bone is a hierarchical structure, comprised of various cells having specific functions that are regulated by sophisticated mechanisms. However, the regulation of the normal functions in damaged or injured cells is disrupted. In order to address this problem, we attempted to artificially generate a scaffold for mimicking the characteristics of the extracellular matrix at the nanoscale level to trigger osteoblastic cell growth. For this purpose, we have chemically grafted bone morphogenetic protein (BMP-2) onto the surface of L-glutamic acid modified hydroxyapatite incorporated into the PLGA nanofiber matrix. After extensive characterization using various spectroscopic techniques, the BMP-g-nHA/PLGA hybrid nanofiber scaffolds were subjected to various in vitro cytocompatibility tests. The results indicated that BMP-2 on BMP-g-nHA/PLGA hybrid nanofiber scaffolds greatly stimulated osteoblastic cells growth, contrary to the nHA/PLGA and pristine PLGA nanofiber scaffold, which are used as control. These results suggest that BMP-g-nHA/PLGA hybrid nanofiber scaffold can be used as a nanodrug carrier for the controlled and targeted delivery of BMP-2, which will open new possibilities for enhancing bone tissue regeneration and will help in the treatment of various bone-related diseases in the future.
Collapse
|
152
|
Denry I, Kuhn LT. Design and characterization of calcium phosphate ceramic scaffolds for bone tissue engineering. Dent Mater 2015; 32:43-53. [PMID: 26423007 DOI: 10.1016/j.dental.2015.09.008] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Our goal is to review design strategies for the fabrication of calcium phosphate ceramic scaffolds (CPS), in light of their transient role in bone tissue engineering and associated requirements for effective bone regeneration. METHODS We examine the various design options available to meet mechanical and biological requirements of CPS and later focus on the importance of proper characterization of CPS in terms of architecture, mechanical properties and time-sensitive properties such as biodegradability. Finally, relationships between in vitro versus in vivo testing are addressed, with an attempt to highlight reliable performance predictors. RESULTS A combinatory design strategy should be used with CPS, taking into consideration 3D architecture, adequate surface chemistry and topography, all of which are needed to promote bone formation. CPS represent the media of choice for delivery of osteogenic factors and anti-infectives. Non-osteoblast mediated mineral deposition can confound in vitro osteogenesis testing of CPS and therefore the expression of a variety of proteins or genes including collagen type I, bone sialoprotein and osteocalcin should be confirmed in addition to increased mineral content. CONCLUSIONS CPS are a superior scaffold material for bone regeneration because they actively promote osteogenesis. Biodegradability of CPS via calcium and phosphate release represents a unique asset. Structural control of CPS at the macro, micro and nanoscale and their combination with cells and polymeric materials is likely to lead to significant developments in bone tissue engineering.
Collapse
Affiliation(s)
- Isabelle Denry
- Department of Prosthodontics, University of Iowa College of Dentistry, 801 Newton Road, Iowa City, IA 52242-1010, USA.
| | - Liisa T Kuhn
- Department of Reconstructive Sciences, UConn Health, 263 Farmington Avenue, MC 1615, Farmington, CT 06030-1615, USA
| |
Collapse
|
153
|
Grenho L, Salgado CL, Fernandes MH, Monteiro FJ, Ferraz MP. Antibacterial activity and biocompatibility of three-dimensional nanostructured porous granules of hydroxyapatite and zinc oxide nanoparticles--an in vitro and in vivo study. NANOTECHNOLOGY 2015; 26:315101. [PMID: 26180062 DOI: 10.1088/0957-4484/26/31/315101] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Ceramic scaffolds are widely studied in the bone tissue engineering field due to their potential in regenerative medicine. However, adhesion of microorganisms on biomaterials with subsequent formation of antibiotic-resistant biofilms is a critical factor in implant-related infections. Therefore, new strategies are needed to address this problem. In the present study, three-dimensional and interconnected porous granules of nanostructured hydroxyapatite (nanoHA) incorporated with different amounts of zinc oxide (ZnO) nanoparticles were produced using a simple polymer sponge replication method. As in vitro experiments, granules were exposed to Staphylococcus aureus and Staphylococcus epidermidis and, after 24 h, the planktonic and sessile populations were assessed. Cytocompatibility towards osteoblast-like cells (MG63 cell line) was also evaluated for a period of 1 and 3 days, through resazurin assay and imaging flow cytometry analysis. As in vivo experiments, nanoHA porous granules with and without ZnO nanoparticles were implanted into the subcutaneous tissue in rats and their inflammatory response after 3, 7 and 30 days was examined, as well as their antibacterial activity after 1 and 3 days of S. aureus inoculation. The developed composites proved to be especially effective at reducing bacterial activity in vitro and in vivo for a weight percentage of 2% ZnO, with a low cell growth inhibition in vitro and no differences in the connective tissue growth and inflammatory response in vivo. Altogether, these results suggest that nanoHA-ZnO porous granules have a great potential to be used in orthopaedic and dental applications as a template for bone regeneration and, simultaneously, to restrain biomaterial-associated infections.
Collapse
Affiliation(s)
- L Grenho
- Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e Materiais, Universidade do Porto, Rua Dr Roberto Frias, s/n 4200-465 Porto, Portugal. i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal. INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
| | | | | | | | | |
Collapse
|
154
|
Ansari S, Phark JH, Duarte S, Paulino da Silva M, Sharifzadeh N, Moshaverinia A, Zadeh HH. Biomechanical analysis of engineered bone with anti-BMP2 antibody immobilized on different scaffolds. J Biomed Mater Res B Appl Biomater 2015; 104:1465-73. [PMID: 26252572 DOI: 10.1002/jbm.b.33492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/16/2015] [Accepted: 07/18/2015] [Indexed: 11/09/2022]
Abstract
Recently we have demonstrated the ability of monoclonal antibodies (mAb) specific for bone morphogenetic protein (BMP)-2 immobilized on different scaffolds to mediate bone formation, a process referred to as Antibody Mediated Osseous Regeneration (AMOR). One of the key properties of regenerated bone is its biomechanical strength, in particular in load-bearing areas. This study sought to test the hypothesis that the biomechanical strength of regenerated bone depends of the mode of regeneration, as well as the scaffold used. Four different scaffolds, namely titanium granules (Ti), alginate hydrogel, anorganic bovine bone mineral (ABBM), and absorbable collagen sponge (ACS) were functionalized with anti-BMP-2 or isotype control mAb and implanted into rat critical-size calvarial defects. The morphology, density and strength of the regenerated bone were evaluated after 8 weeks. Results demonstrated that scaffolds functionalized with anti-BMP-2 mAb exhibited varying degrees of bone volume and density. Ti and ABBM achieved the highest bone volume, density, and strength of bone. When anti-BMP-2 mAb was immobilized on Ti or ABBM, the strength of the regenerated bone were 80% and 77% of native bone respectively, compared with 60% of native bone in sites implanted with rh-BMP-2. Control interventions with isotype mAb did not promote considerable bone regeneration and exhibited significantly lower mechanical properties. SEM analysis showed specimens immobilized with anti-BMP-2 mAb formed new bone with organized structure bridging the crack areas. Altogether, the present data demonstrated that the morphological and mechanical properties of bone bioengineered through AMOR could approximate that of native bone, when appropriate scaffolds are used. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 104B: 1465-1473, 2016.
Collapse
Affiliation(s)
- Sahar Ansari
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Jin-Ho Phark
- Division of Restorative Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Sillas Duarte
- Division of Restorative Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Maike Paulino da Silva
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, Sao Paulo, Brazil
| | - Navid Sharifzadeh
- Division of Periodontology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA
| | - Homayoun H Zadeh
- Division of Periodontology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA.
| |
Collapse
|
155
|
Ansari S, Freire M, Choi MG, Tavari A, Almohaimeed M, Moshaverinia A, Zadeh HH. Effects of the orientation of anti-BMP2 monoclonal antibody immobilized on scaffold in antibody-mediated osseous regeneration. J Biomater Appl 2015; 30:558-67. [PMID: 26184354 DOI: 10.1177/0885328215594704] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recently, we have shown that anti-BMP2 monoclonal antibodies (mAbs) can trap endogenous osteogenic BMP ligands, which can in turn mediate osteodifferentiation of progenitor cells. The effectiveness of this strategy requires the availability of the anti-BMP-2 monoclonal antibodies antigen-binding sites for anti-BMP-2 monoclonal antibodies to bind to the scaffold through a domain that will leave its antigen-binding region exposed and available for binding to an osteogenic ligand. We examined whether antibodies bound to a scaffold by passive adsorption versus through Protein G as a linker will exhibit differences in mediating bone formation. In vitro anti-BMP-2 monoclonal antibodies was immobilized on absorbable collagen sponge (ACS) with Protein G as a linker to bind the antibody through its Fc region and implanted into rat calvarial defects. The biomechanical strength of bone regenerated by absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies immune complex was compared to ACS/anti-BMP-2 monoclonal antibodies or ACS/Protein G/isotype mAb control group. Results demonstrated higher binding of anti-BMP-2 monoclonal antibodies/BMPs to C2C12 cells, when the mAb was initially attached to recombinant Protein G or Protein G-coupled microbeads. After eight weeks, micro-CT and histomorphometric analyses revealed increased bone formation within defects implanted with absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies compared with defects implanted with absorbable collagen sponge/anti-BMP-2 monoclonal antibodies (p < 0.05). Confocal laser scanning microscopy (CLSM) confirmed increased BMP-2, -4, and -7 detection in sites implanted with absorbable collagen sponge/Protein G/anti-BMP-2 monoclonal antibodies in vivo. Biomechanical analysis revealed the regenerated bone in sites with Protein G/anti-BMP-2 monoclonal antibodies had higher mechanical strength in comparison to anti-BMP-2 monoclonal antibodies. The negative control group, Protein G/isotype mAb, did not promote bone regeneration and exhibited significantly lower mechanical properties (p < 0.05). Altogether, our results demonstrated that application of Protein G as a linker to adsorb anti-BMP-2 monoclonal antibodies onto the scaffold was accompanied by increased in vitro binding of the anti-BMP-2 mAb/BMP immune complex to BMP-receptor positive cell, as well as increased volume and strength of de novo bone formation in vivo.
Collapse
Affiliation(s)
- Sahar Ansari
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Marcelo Freire
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Moon G Choi
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Azadeh Tavari
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Mohammad Almohaimeed
- Dental Research Center (DRC), Tissue Engineering and Biomaterials Research Unit (TEBRU), College of Dentistry, Qassim University, Qassim, Saudi Arabia
| | - Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Homayoun H Zadeh
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
156
|
Yu X, Suárez-González D, Khalil AS, Murphy WL. How does the pathophysiological context influence delivery of bone growth factors? Adv Drug Deliv Rev 2015; 84:68-84. [PMID: 25453269 PMCID: PMC4401584 DOI: 10.1016/j.addr.2014.10.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/29/2014] [Accepted: 10/07/2014] [Indexed: 02/08/2023]
Abstract
"Orthobiologics" represents an important category of therapeutics for the regeneration of bone defects caused by injuries or diseases, and bone growth factors are a particularly rapidly growing sub-category. Clinical application of bone growth factors has accelerated in the last two decades with the introduction of BMPs into clinical bone repair. Optimal use of growth factor-mediated treatments heavily relies on controlled delivery, which can substantially influence the local growth factor dose, release kinetics, and biological activity. The characteristics of the surrounding environment, or "context", during delivery can dictate growth factor loading efficiency, release and biological activity. This review discusses the influence of the surrounding environment on therapeutic delivery of bone growth factors. We specifically focus on pathophysiological components, including soluble components and cells, and how they can actively influence the therapeutic delivery and perhaps efficacy of bone growth factors.
Collapse
Affiliation(s)
- Xiaohua Yu
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Darilis Suárez-González
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
157
|
Brooks BD, Sinclair KD, Grainger DW, Brooks AE. A resorbable antibiotic-eluting polymer composite bone void filler for perioperative infection prevention in a rabbit radial defect model. PLoS One 2015; 10:e0118696. [PMID: 25815727 PMCID: PMC4376868 DOI: 10.1371/journal.pone.0118696] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 01/23/2015] [Indexed: 02/04/2023] Open
Abstract
Nearly 1.3 million total joint replacement procedures are performed in the United States annually, with numbers projected to rise exponentially in the coming decades. Although finite infection rates for these procedures remain consistently low, device-related infections represent a significant cause of implant failure, requiring secondary or revision procedures. Revision procedures manifest several-fold higher infection recurrence rates. Importantly, many revision surgeries, infected or not, require bone void fillers to support the host bone and provide a sufficient tissue bed for new hardware placement. Antibiotic-eluting bone void fillers (ABVF), providing both osteoconductive and antimicrobial properties, represent one approach for reducing rates of orthopedic device-related infections. Using a solvent-free, molten-cast process, a polymer-controlled antibiotic-eluting calcium carbonate hydroxyapatite (HAP) ceramic composite BVF (ABVF) was fabricated, characterized, and evaluated in vivo using a bacterial challenge in a rabbit radial defect window model. ABVF loaded with tobramycin eliminated the infectious burden in rabbits challenged with a clinically relevant strain of Staphylococcus aureus (inoculum as high as 10⁷ CFU). Histological, microbiological, and radiographic methods were used to detail the effects of ABVF on microbial challenge to host bone after 8 weeks in vivo. In contrast to the HAP/BVF controls, which provided no antibiotic protection and required euthanasia 3 weeks post-operatively, tobramycin-releasing ABVF animals showed no signs of infection (clinical, microbiological, or radiographic) when euthanized at the 8-week study endpoint. ABVF sites did exhibit fibrous encapsulation around the implant at 8 weeks. Local antibiotic release from ABVF to orthopedic sites requiring bone void fillers eliminated the periprosthetic bacterial challenge in this 8-week in vivo study, confirming previous in vitro results.
Collapse
Affiliation(s)
- Benjamin D Brooks
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah, 84112-5820, United States of America
| | - Kristofer D Sinclair
- Elute Inc., 417 Wakara Way, Suite 3510, Salt Lake City, Utah, 84108, United States of America
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah, 84112-5820, United States of America; Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112-5820, United States of America
| | - Amanda E Brooks
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah, 84112-5820, United States of America
| |
Collapse
|
158
|
Wen C, Yan H, Fu S, Qian Y, Wang D, Wang C. Allogeneic adipose-derived stem cells regenerate bone in a critical-sized ulna segmental defect. Exp Biol Med (Maywood) 2015; 241:1401-9. [PMID: 25819682 DOI: 10.1177/1535370215576298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 01/28/2015] [Indexed: 12/17/2022] Open
Abstract
Adipose-derived stem cells (ASCs) with multilineage potential can be induced into osteoblasts, adipocytes and chondrocytes. ASCs as seed cell are widely used in the field of tissue engineering, but most studies either use autologous cells as the source or an immunodeficient animal as the host. In our present study, we explored the feasibility of applying allogeneic ASCs and demineralized bone matrix (DBM) scaffolds for repairing tubular bone defects without using immunosuppressive therapy. Allogeneic ASCs were expanded and seeded on DBM scaffolds and induced to differentiate along the osteogenic lineage. Eight Sprague-Dawley (SD) rats were used in this study and bilateral critical-sized defects (8 mm) of the ulna were created and divided into two groups: with ASC-DBM constructs or DBM alone. The systemic immune response and the extent of bone healing were evaluated post-operatively. Twenty-four weeks after implantation, digital radiography (DR) testing showed that new bones had formed in the experimental group. By contrast, no bone tissue formation was observed in the control group. This study demonstrated that allogeneic ASCs could promote bone regeneration and repair tubular bone defects combined with DBM by histologically typical bone without systemic immune response.
Collapse
Affiliation(s)
- Congji Wen
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China Department of Plastic Surgery, Yancheng First Peoples' Hospital, 16 Yue He Road, People's Republic of China. 224000
| | - Hai Yan
- Department of Orthepedics, Nantong Rich Hospital, No. 2000 Lake Avenue, Nantong, 226010, People's Republic of China
| | - Shibo Fu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China
| | - Yunliang Qian
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China
| | - Danru Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China
| | - Chen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China
| |
Collapse
|
159
|
Velasco MA, Narváez-Tovar CA, Garzón-Alvarado DA. Design, materials, and mechanobiology of biodegradable scaffolds for bone tissue engineering. BIOMED RESEARCH INTERNATIONAL 2015; 2015:729076. [PMID: 25883972 PMCID: PMC4391163 DOI: 10.1155/2015/729076] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/27/2015] [Indexed: 11/22/2022]
Abstract
A review about design, manufacture, and mechanobiology of biodegradable scaffolds for bone tissue engineering is given. First, fundamental aspects about bone tissue engineering and considerations related to scaffold design are established. Second, issues related to scaffold biomaterials and manufacturing processes are discussed. Finally, mechanobiology of bone tissue and computational models developed for simulating how bone healing occurs inside a scaffold are described.
Collapse
Affiliation(s)
- Marco A. Velasco
- Studies and Applications in Mechanical Engineering Research Group (GEAMEC), Universidad Santo Tomás, Bogotá, Colombia
| | - Carlos A. Narváez-Tovar
- Studies and Applications in Mechanical Engineering Research Group (GEAMEC), Universidad Santo Tomás, Bogotá, Colombia
- Biomimetics Laboratory and Numerical Methods and Modeling Research Group (GNUM), Instituto de Biotecnología (IBUN), Universidad Nacional de Colombia, Bogotá, Colombia
| | - Diego A. Garzón-Alvarado
- Biomimetics Laboratory and Numerical Methods and Modeling Research Group (GNUM), Instituto de Biotecnología (IBUN), Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
160
|
Zhang J, He F, Zhang W, Zhang M, Yang H, Luo ZP. Mechanical force enhanced bony formation in defect implanted with calcium sulphate cement. Bone Res 2015; 3:14048. [PMID: 26273532 PMCID: PMC4472145 DOI: 10.1038/boneres.2014.48] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 08/24/2014] [Accepted: 10/25/2014] [Indexed: 11/13/2022] Open
Abstract
To improve the osteogenic property of bone repairing materials and to accelerate bone healing are major tasks in bone biomaterials research. The objective of this study was to investigate if the mechanical force could be used to accelerate bone formation in a bony defect in vivo. The calcium sulfate cement was implanted into the left distal femoral epiphyses surgically in 16 rats. The half of rats were subjected to external mechanical force via treadmill exercise, the exercise started at day 7 postoperatively for 30 consecutive days and at a constant speed 8 m·min−1 for 45 min·day−1, while the rest served as a control. The rats were scanned four times longitudinally after surgery using microcomputed tomography and newly formed bone was evaluated. After sacrificing, the femurs had biomechanical test of three-point bending and histological analysis. The results showed that bone healing under mechanical force were better than the control with residual defect areas of 0.64±0.19 mm2 and 1.78±0.39 mm2 (P<0.001), and the ultimate loads to failure under mechanical force were 69.56±4.74 N, stronger than the control with ultimate loads to failure of 59.17±7.48 N (P=0.039). This suggests that the mechanical force might be used to improve new bone formation and potentially offer a clinical strategy to accelerate bone healing.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Orthopedic Surgery, the 1st Affiliated Hospital, and Orthopedic Institute, Soochow University , Suzhou, China
| | - Fan He
- Department of Orthopedic Surgery, the 1st Affiliated Hospital, and Orthopedic Institute, Soochow University , Suzhou, China
| | - Wen Zhang
- Department of Orthopedic Surgery, the 1st Affiliated Hospital, and Orthopedic Institute, Soochow University , Suzhou, China
| | - Meng Zhang
- Department of Orthopedic Surgery, the 1st Affiliated Hospital, and Orthopedic Institute, Soochow University , Suzhou, China
| | - Huilin Yang
- Department of Orthopedic Surgery, the 1st Affiliated Hospital, and Orthopedic Institute, Soochow University , Suzhou, China
| | - Zong-Ping Luo
- Department of Orthopedic Surgery, the 1st Affiliated Hospital, and Orthopedic Institute, Soochow University , Suzhou, China
| |
Collapse
|
161
|
Wu L, Yuan Y, Hao F, Yang Z, Zhang J, Yu M. The Effects of SiC Foams on Cell Proliferation and Differentiation in Primary Osteoblasts. J HARD TISSUE BIOL 2015. [DOI: 10.2485/jhtb.24.37] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Lin Wu
- Department of Prosthodontics, School of Stomatology, China Medical University
| | - Yue Yuan
- Department of Prosthodontics, School of Stomatology, China Medical University
| | - Fengyu Hao
- Department of Prosthodontics, School of Stomatology, China Medical University
| | - Zhenming Yang
- Materials Fabrication and Processing Division, Institute of Metal Research, Chinese Academy of Sciences
| | - Jinsong Zhang
- Materials Fabrication and Processing Division, Institute of Metal Research, Chinese Academy of Sciences
| | - Meng Yu
- Key Laboratory of Transgenetic Animal Research, Liaoning Province, Department of Laboratory Animal of China Medical University, China Medical University
| |
Collapse
|
162
|
Schlaubitz S, Derkaoui SM, Marosa L, Miraux S, Renard M, Catros S, Le Visage C, Letourneur D, Amédée J, Fricain JC. Pullulan/dextran/nHA macroporous composite beads for bone repair in a femoral condyle defect in rats. PLoS One 2014; 9:e110251. [PMID: 25330002 PMCID: PMC4203774 DOI: 10.1371/journal.pone.0110251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/15/2014] [Indexed: 11/29/2022] Open
Abstract
The repair of bone defects is of particular interest for orthopedic, oral, maxillofacial, and dental surgery. Bone loss requiring reconstruction is conventionally addressed through bone grafting. Depending on the size and the location of the defect, this method has limits and risks. Biomaterials can offer an alternative and have features supporting bone repair. Here, we propose to evaluate the cellular penetration and bone formation of new macroporous beads based on pullulan/dextran that has been supplemented with nanocrystalline hydroxyapatite in a rat model. Cross-linked beads of 300–500 µm diameters were used in a lateral femoral condyle defect and analyzed by magnetic resonance imaging, micro-computed tomography, and histology in comparison to the empty defects 15, 30, and 70 days after implantation. Inflammation was absent for both conditions. For empty defects, cellularisation and mineralization started from the periphery of the defect. For the defects containing beads, cellular structures filling out the spaces between the scaffolds with increasing interconnectivity and trabecular-like organization were observed over time. The analysis of calcified sections showed increased mineralization over time for both conditions, but was more pronounced for the samples containing beads. Bone Mineral Density and Bone Mineral Content were both significantly higher at day 70 for the beads in comparison to empty defects as well as compared with earlier time points. Analysis of newly formed tissue around the beads showed an increase of osteoid tissue, measured as percentage of the defect surface. This study suggests that the use of beads for the repair of small size defects in bone may be expanded on to meet the clinical need for a ready-to-use fill-up material that can favor bone formation and mineralization, as well as promote vessel ingrowth into the defect site.
Collapse
Affiliation(s)
- Silke Schlaubitz
- CIC 1401, University hospital of Bordeaux/Inserm, Bordeaux, France
| | - Sidi Mohammed Derkaoui
- U1148, LVTS/Inserm, Paris, France
- Près Sorbonne Paris Cité, University of Paris Nord and University Paris Diderot, Paris, France
| | - Lydia Marosa
- U1026 Tissue Bioengineering, University of Bordeaux/Inserm, Bordeaux, France
| | | | - Martine Renard
- CIC 1401, University hospital of Bordeaux/Inserm, Bordeaux, France
| | - Sylvain Catros
- U1026 Tissue Bioengineering, University of Bordeaux/Inserm, Bordeaux, France
- Dental School, University of Bordeaux, Bordeaux, France
| | - Catherine Le Visage
- U1148, LVTS/Inserm, Paris, France
- Près Sorbonne Paris Cité, University of Paris Nord and University Paris Diderot, Paris, France
| | - Didier Letourneur
- U1148, LVTS/Inserm, Paris, France
- Près Sorbonne Paris Cité, University of Paris Nord and University Paris Diderot, Paris, France
| | - Joëlle Amédée
- U1026 Tissue Bioengineering, University of Bordeaux/Inserm, Bordeaux, France
| | - Jean-Christophe Fricain
- U1026 Tissue Bioengineering, University of Bordeaux/Inserm, Bordeaux, France
- Dental School, University of Bordeaux, Bordeaux, France
- * E-mail:
| |
Collapse
|
163
|
Immobilization of murine anti-BMP-2 monoclonal antibody on various biomaterials for bone tissue engineering. BIOMED RESEARCH INTERNATIONAL 2014; 2014:940860. [PMID: 25147826 PMCID: PMC4132312 DOI: 10.1155/2014/940860] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/02/2014] [Indexed: 11/18/2022]
Abstract
Biomaterials are widely used as scaffolds for tissue engineering. We have developed a strategy for bone tissue engineering that entails application of immobilized anti-BMP-2 monoclonal antibodies (mAbs) to capture endogenous BMPs in vivo and promote antibody-mediated osseous regeneration (AMOR). The purpose of the current study was to compare the efficacy of immobilization of a specific murine anti-BMP-2 mAb on three different types of biomaterials and to evaluate their suitability as scaffolds for AMOR. Anti-BMP-2 mAb or isotype control mAb was immobilized on titanium (Ti) microbeads, alginate hydrogel, and ACS. The treated biomaterials were surgically implanted in rat critical-sized calvarial defects. After 8 weeks, de novo bone formation was assessed using micro-CT and histomorphometric analyses. Results showed de novo bone regeneration with all three scaffolds with immobilized anti-BMP-2 mAb, but not isotype control mAb. Ti microbeads showed the highest volume of bone regeneration, followed by ACS. Alginate showed the lowest volume of bone. Localization of BMP-2, -4, and -7 antigens was detected on all 3 scaffolds with immobilized anti-BMP-2 mAb implanted in calvarial defects. Altogether, these data suggested a potential mechanism for bone regeneration through entrapment of endogenous BMP-2, -4, and -7 proteins leading to bone formation using different types of scaffolds via AMOR.
Collapse
|
164
|
Development and characterization of novel porous 3D alginate-cockle shell powder nanobiocomposite bone scaffold. BIOMED RESEARCH INTERNATIONAL 2014; 2014:146723. [PMID: 25110655 PMCID: PMC4109673 DOI: 10.1155/2014/146723] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/05/2014] [Accepted: 06/15/2014] [Indexed: 11/18/2022]
Abstract
A novel porous three-dimensional bone scaffold was developed using a natural polymer (alginate/Alg) in combination with a naturally obtained biomineral (nano cockle shell powder/nCP) through lyophilization techniques. The scaffold was developed in varying composition mixture of Alg-nCP and characterized using various evaluation techniques as well as preliminary in vitro studies on MG63 human osteoblast cells. Morphological observations using SEM revealed variations in structures with the use of different Alg-nCP composition ratios. All the developed scaffolds showed a porous structure with pore sizes ideal for facilitating new bone growth; however, not all combination mixtures showed subsequent favorable characteristics to be used for biological applications. Scaffolds produced using the combination mixture of 40% Alg and 60% nCP produced significantly promising results in terms of mechanical strength, degradation rate, and increased cell proliferation rates making it potentially the optimum composition mixture of Alg-nCP with future application prospects.
Collapse
|
165
|
Marques C, Ferreira JMF, Andronescu E, Ficai D, Sonmez M, Ficai A. Multifunctional materials for bone cancer treatment. Int J Nanomedicine 2014; 9:2713-25. [PMID: 24920907 PMCID: PMC4044993 DOI: 10.2147/ijn.s55943] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The purpose of this review is to present the most recent findings in bone tissue engineering. Special attention is given to multifunctional materials based on collagen and collagen-hydroxyapatite composites used for skin and bone cancer treatments. The multi-functionality of these materials was obtained by adding to the base regenerative grafts proper components, such as ferrites (magnetite being the most important representative), cytostatics (cisplatin, carboplatin, vincristine, methotrexate, paclitaxel, doxorubicin), silver nanoparticles, antibiotics (anthracyclines, geldanamycin), and/or analgesics (ibuprofen, fentanyl). The suitability of complex systems for the intended applications was systematically analyzed. The developmental possibilities of multifunctional materials with regenerative and curative roles (antitumoral as well as pain management) in the field of skin and bone cancer treatment are discussed. It is worth mentioning that better materials are likely to be developed by combining conventional and unconventional experimental strategies.
Collapse
Affiliation(s)
- Catarina Marques
- Department of Materials and Ceramics Engineering, Centre for Research in Ceramics and Composite Materials, University of Aveiro, Aveiro, Portugal
| | - José MF Ferreira
- Department of Materials and Ceramics Engineering, Centre for Research in Ceramics and Composite Materials, University of Aveiro, Aveiro, Portugal
| | - Ecaterina Andronescu
- Faculty of Applied Chemistry and Material Science, University Politehnica of Bucharest, Bucharest, Romania
| | - Denisa Ficai
- Faculty of Applied Chemistry and Material Science, University Politehnica of Bucharest, Bucharest, Romania
| | - Maria Sonmez
- National Research and Development Institute for Textiles and Leather, Bucharest, Romania
| | - Anton Ficai
- Faculty of Applied Chemistry and Material Science, University Politehnica of Bucharest, Bucharest, Romania
| |
Collapse
|
166
|
Jin H, Zhang K, Qiao C, Yuan A, Li D, Zhao L, Shi C, Xu X, Ni S, Zheng C, Liu X, Yang B, Sun H. Efficiently engineered cell sheet using a complex of polyethylenimine-alginate nanocomposites plus bone morphogenetic protein 2 gene to promote new bone formation. Int J Nanomedicine 2014; 9:2179-90. [PMID: 24855355 PMCID: PMC4019610 DOI: 10.2147/ijn.s60937] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regeneration of large bone defects is a common clinical problem. Recently, stem cell sheet has been an emerging strategy in bone tissue engineering. To enhance the osteogenic potential of stem cell sheet, we fabricated bone morphogenetic protein 2 (BMP-2) gene-engineered cell sheet using a complex of polyethylenimine-alginate (PEI-al) nanocomposites plus human BMP-2 complementary(c)DNA plasmid, and studied its osteogenesis in vitro and in vivo. PEI-al nanocomposites carrying BMP-2 gene could efficiently transfect bone marrow mesenchymal stem cells. The cell sheet was made by culturing the cells in medium containing vitamin C for 10 days. Assays on the cell culture showed that the genetically engineered cells released the BMP-2 for at least 14 days. The expression of osteogenesis-related gene was increased, which demonstrated that released BMP-2 could effectively induce the cell sheet osteogenic differentiation in vitro. To further test the osteogenic potential of the cell sheet in vivo, enhanced green fluorescent protein or BMP-2-producing cell sheets were treated on the cranial bone defects. The results indicated that the BMP-2-producing cell sheet group was more efficient than other groups in promoting bone formation in the defect area. Our results suggested that PEI-al nanocomposites efficiently deliver the BMP-2 gene to bone marrow mesenchymal stem cells and that BMP-2 gene-engineered cell sheet is an effective way for promoting bone regeneration.
Collapse
Affiliation(s)
- Han Jin
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Kai Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, People's Republic of China
| | - Chunyan Qiao
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Anliang Yuan
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Daowei Li
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Liang Zhao
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Ce Shi
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Xiaowei Xu
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Shilei Ni
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Changyu Zheng
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M University Baylor College of Dentistry, Dallas, TX, USA
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, People's Republic of China
| | - Hongchen Sun
- Department of Pathology, School of Stomatology, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
167
|
Xia Z, Villa MM, Wei M. A Biomimetic Collagen-Apatite Scaffold with a Multi-Level Lamellar Structure for Bone Tissue Engineering. J Mater Chem B 2014; 2:1998-2007. [PMID: 24999428 PMCID: PMC4078891 DOI: 10.1039/c3tb21595d] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Collagen-apatite (Col-Ap) scaffolds have been widely employed for bone tissue engineering. We fabricated a Col-Ap scaffold with a unique multi-level lamellar structure consisting of co-aligned micro and macro pores. The basic building blocks of this scaffold are bone-like mineralized collagen fibers developed via a biomimetic self-assembly process in a collagen-containing modified simulated body fluid (m-SBF). This biomimetic method preserves the structural integrity and great tensile strength of collagen by reinforcing the collagen hydrogel with apatite nano-particles. Unidirectional aligned macro pores with a size of 63.8 to 344 μm are created by controlling the freezing rate and direction. The thickness of Col-Ap lamellae can be adjusted in the range 3.6 to 23 μm depending on the self-compression time. Furthermore, the multi-level lamellar structure has led to a twelve-fold increase in Young's modulus and a two-fold increase in the compression modulus along the aligned direction compared to a scaffold of the same composition with an isotropic equiaxed pore structure. Moreover, this novel lamellar scaffold supports the attachment and spreading of MC3T3-E1osteoblasts. Therefore, owing to the biomimetic composition, tunable structure, improved mechanical strength, and good biocompatibility of this novel scaffold, it has great potential to be used in bone tissue engineering applications.
Collapse
Affiliation(s)
- Z Xia
- Department of Materials Science and Engineering, University of Connecticut, 97 North Eagleville Road, U-3136, Storrs, CT, 06269
| | - M M Villa
- Department of Materials Science and Engineering, University of Connecticut, 97 North Eagleville Road, U-3136, Storrs, CT, 06269
| | - M Wei
- Department of Materials Science and Engineering, University of Connecticut, 97 North Eagleville Road, U-3136, Storrs, CT, 06269
| |
Collapse
|
168
|
Fabrication and characteristics of chitosan sponge as a tissue engineering scaffold. BIOMED RESEARCH INTERNATIONAL 2014; 2014:786892. [PMID: 24804246 PMCID: PMC3997083 DOI: 10.1155/2014/786892] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 03/10/2014] [Indexed: 01/13/2023]
Abstract
Cells, growth factors, and scaffolds are the three main factors required to create a tissue-engineered construct. After the appearance of bovine spongiform encephalopathy (BSE), considerable attention has therefore been focused on nonbovine materials. In this study, we examined the properties of a chitosan porous scaffold. A porous chitosan sponge was prepared by the controlled freezing and lyophilization of different concentrations of chitosan solutions. The materials were examined by scanning electron microscopy, and the porosity, tensile strength, and basic fibroblast growth factor (bFGF) release profiles from chitosan sponge were examined in vitro. The morphology of the chitosan scaffolds presented a typical microporous structure, with the pore size ranging from 50 to 200 μm. The porosity of chitosan scaffolds with different concentrations was approximately 75–85%. A decreasing tendency for porosity was observed as the concentration of the chitosan increased. The relationship between the tensile properties and chitosan concentration indicated that the ultimate tensile strength for the sponge increased with a higher concentration. The in vitro bFGF release study showed that the higher the concentration of chitosan solution became, the longer the releasing time of the bFGF from the chitosan sponge was.
Collapse
|
169
|
Faghihi F, Baghaban Eslaminejad M. The effect of nano-scale topography on osteogenic differentiation of mesenchymal stem cells. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:5-16. [DOI: 10.5507/bp.2013.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 02/14/2013] [Indexed: 01/08/2023] Open
|
170
|
Hu J, Zhou Y, Huang L, Liu J, Lu H. Effect of nano-hydroxyapatite coating on the osteoinductivity of porous biphasic calcium phosphate ceramics. BMC Musculoskelet Disord 2014; 15:114. [PMID: 24690170 PMCID: PMC3994218 DOI: 10.1186/1471-2474-15-114] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 03/05/2014] [Indexed: 12/14/2022] Open
Abstract
Background Porous biphasic calcium phosphate (BCP) ceramics exhibit good biocompatibility and bone conduction but are not inherently osteoinductive. To overcome this disadvantage, we coated conventional porous BCP ceramics with nano-hydroxyapatite (nHA). nHA was chosen as a coating material due to its high osteoinductive potential. Methods We used a hydrothermal deposition method to coat conventional porous BCP ceramics with nHA and assessed the effects of the coating on the physical and mechanical properties of the underlying BCP. Next, its effects on mesenchymal stem cell (MSC) attachment, proliferation, viability, and osteogenic differentiation were investigated. Results nHA formed a deposited layer on the BCP surface, and synthesized nHA had a rod-like shape with lengths ranging from ~50–200 nm and diameters from ~15–30 mm. The nHA coating did not significantly affect the density, porosity, flexural strength, or compressive strength of the underlying BCP (P > 0.1). Scanning electron microscopy showed MSC attachment to the scaffolds, with a healthy morphology and anchorage to nHA crystals via cytoplasmic processes. The densities of MSCs attached on BCP and nHA-coated BCP scaffolds were 62 ± 26 cells/mm2 and 63 ± 27 cells/mm2 (P > 0.1), respectively, after 1 day and 415 ± 62 cells/mm2 and 541 ± 35 cells/mm2 (P < 0.05) respectively, after 14 days. According to an MTT assay, MSC viability was higher on nHA-coated BCP scaffolds than on BCP scaffolds (P < 0.05). In addition, MSCs on nHA-coated BCP scaffolds produced more alkaline phosphatase, collagen type I, and osteocalcin than MSCs on BCP scaffolds (P < 0.05). Conclusions Our results demonstrate that BCP scaffolds coated with nHA were more conducive for MSC adhesion, proliferation, and osteogenic differentiation than conventional, uncoated BCP scaffolds, indicating that nHA coating can enhance the osteoinductive potential of BCP ceramics, making this material more suitable for applications in bone tissue engineering.
Collapse
Affiliation(s)
| | | | | | | | - Hongbin Lu
- Department of Sports Medicine, Research Center of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
171
|
Ariani MD, Matsuura A, Hirata I, Kubo T, Kato K, Akagawa Y. New development of carbonate apatite-chitosan scaffold based on lyophilization technique for bone tissue engineering. Dent Mater J 2014; 32:317-25. [PMID: 23538769 DOI: 10.4012/dmj.2012-257] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Carbonate apatite-chitosan scaffolds (CA-ChSs) were fabricated using the lyophilization technique. It was found that ChSs prepared with 200 mg chitosan powder (ChSs200) had well-structured three-dimensional architecture with high porosity and good retentive form without brittleness. In addition, it was shown that the number of osteoblast-like cells MC3T3-E1 proliferated on desalinated ChSs200 was larger than that on the non-desalinated ChSs200. CA-ChSs were fabricated by adding 100 mg carbonate apatite (CA) to 200 mg chitosan gels followed by freeze-drying (CA100ChSs200). SEM observation revealed that CA100ChSs200 had favorable three- dimensional porous structures. The number of living cells increased more rapidly on CA100ChSs200 prepared with different amounts of CA than on ChSs. ALP activity significantly increased after day 14 and reached a plateau after day 21 in ChSs200 and CA100ChSs200. It was concluded that newly developed CA100ChSs200 may be a possible scaffold material for tissue engineering.
Collapse
Affiliation(s)
- Maretaningtias Dwi Ariani
- Department of Advanced Prosthodontics, Hiroshima University Graduate School of Biomedical Sciences, Minami-ku, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
172
|
Asafo-Adjei TA, Dziubla TD, Puleo DA. Synthesis and Characterization of a Poly(ethylene glycol)-Poly(simvastatin) Diblock Copolymer. RSC Adv 2014; 4:58287-58298. [PMID: 25431653 PMCID: PMC4242434 DOI: 10.1039/c4ra10310f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Biodegradable polyesters are commonly used as drug delivery vehicles, but their role is typically passive, and encapsulation approaches have limited drug payload. An alternative drug delivery method is to polymerize the active agent or its precursor into a degradable polymer. The prodrug simvastatin contains a lactone ring that lends itself to ring-opening polymerization (ROP). Consequently, simvastatin polymerization was initiated with 5 kDa monomethyl ether poly(ethylene glycol) (mPEG) and catalyzed via stannous octoate. Melt condensation reactions produced a 9.5 kDa copolymer with a polydispersity index of 1.1 at 150 °C up to a 75 kDa copolymer with an index of 6.9 at 250 °C. Kinetic analysis revealed first-order propagation rates. Infrared spectroscopy of the copolymer showed carboxylic and methyl ether stretches unique to simvastatin and mPEG, respectively. Slow degradation was demonstrated in neutral and alkaline conditions. Lastly, simvastatin, simvastatin-incorporated molecules, and mPEG were identified as the degradation products released. The present results show the potential of using ROP to polymerize lactone-containing drugs such as simvastatin.
Collapse
Affiliation(s)
| | - Thomas D. Dziubla
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, USA
| | - David A. Puleo
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
173
|
Eldesoqi K, Seebach C, Nguyen Ngoc C, Meier S, Nau C, Schaible A, Marzi I, Henrich D. High calcium bioglass enhances differentiation and survival of endothelial progenitor cells, inducing early vascularization in critical size bone defects. PLoS One 2013; 8:e79058. [PMID: 24244419 PMCID: PMC3828289 DOI: 10.1371/journal.pone.0079058] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 09/21/2013] [Indexed: 01/21/2023] Open
Abstract
Early vascularization is a prerequisite for successful bone healing and endothelial progenitor cells (EPC), seeded on appropriate biomaterials, can improve vascularization. The type of biomaterial influences EPC function with bioglass evoking a vascularizing response. In this study the influence of a composite biomaterial based on polylactic acid (PLA) and either 20 or 40% bioglass, BG20 and BG40, respectively, on the differentiation and survival of EPCs in vitro was investigated. Subsequently, the effect of the composite material on early vascularization in a rat calvarial critical size defect model with or without EPCs was evaluated. Human EPCs were cultured with β-TCP, PLA, BG20 or BG40, and seeding efficacy, cell viability, cell morphology and apoptosis were analysed in vitro. BG40 released the most calcium, and improved endothelial differentiation and vitality best. This effect was mimicked by adding an equivalent amount of calcium to the medium and was diminished in the presence of the calcium chelator, EGTA. To analyze the effect of BG40 and EPCs in vivo, a 6-mm diameter critical size calvarial defect was created in rats (n = 12). Controls (n = 6) received BG40 and the treatment group (n = 6) received BG40 seeded with 5×105 rat EPCs. Vascularization after 1 week was significantly improved when EPCs were seeded onto BG40, compared to implanting BG40 alone. This indicates that Ca2+ release improves EPC differentiation and is useful for enhanced early vascularization in critical size bone defects.
Collapse
Affiliation(s)
- Karam Eldesoqi
- Department of Trauma-, Hand- and Reconstructive Surgery, Hospital of the Goethe- University, Frankfurt/Main, Germany
| | | | | | | | | | | | | | | |
Collapse
|
174
|
Mantripragada VP, Lecka-Czernik B, Ebraheim NA, Jayasuriya AC. An overview of recent advances in designing orthopedic and craniofacial implants. J Biomed Mater Res A 2013; 101:3349-64. [PMID: 23766134 PMCID: PMC4854641 DOI: 10.1002/jbm.a.34605] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/11/2013] [Accepted: 01/11/2013] [Indexed: 01/22/2023]
Abstract
Great deal of research is still going on in the field of orthopedic and craniofacial implant development to resolve various issues being faced by the industry today. Despite several disadvantages of the metallic implants, they continue to be used, primarily because of their superior mechanical properties. In order to minimize the harmful effects of the metallic implants and its by-products, several modifications are being made to these materials, for instance nickel-free stainless steel, cobalt-chromium and titanium alloys are being introduced to eliminate the toxic effects of nickel being released from the alloys, introduce metallic implants with lower modulus, reduce the cost of these alloys by replacing rare elements with less expensive elements etc. New alloys like tantalum, niobium, zirconium, and magnesium are receiving attention given their satisfying mechanical and biological properties. Non-oxide ceramics like silicon nitride and silicon carbide are being currently developed as a promising implant material possessing a combination of properties such as good wear and corrosion resistance, increased ductility, good fracture and creep resistance, and relatively high hardness in comparison to alumina. Polymer/magnesium composites are being developed to improve mechanical properties as well as retain polymer's property of degradation. Recent advances in orthobiologics are proving interesting as well. This paper thus deals with the latest improvements being made to the existing implant materials and includes new materials being introduced in the field of biomaterials.
Collapse
|
175
|
Influence of the Precipitation Temperature on Properties of Nanohydroxyapatite Powder for the Fabrication of Highly Porous Bone Scaffolds. ACTA ACUST UNITED AC 2013. [DOI: 10.4028/www.scientific.net/kem.587.27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of the present work is to study the influence of the precipitation temperature in the synthesis of nanohydroxyapatite (n-HAp) on the properties of the resulting n-HAp powder for the fabrication of highly porous scaffolds for bone tissue engineering. The n-HAp powder was obtained by a wet precipitation technique starting from calcium nitrate tetrahydrate (Ca (NO3)2*4H2O) and phosphoric acid (H3PO4) at different temperatures: 10°C, 37°C and 50°C. Highly porous scaffolds were fabricated using the three different powders by the sponge replica method and sintering at 1300°C. Combined X-ray diffraction (XRD) and transmission electron microscopy (TEM) analyses on powders indicated that on increasing the precipitation temperature the formation of pure n-HAp is accelerated, without significant changes in particles morphology and size. Scaffolds characterized by high porosity (89%) and good compressive strength (0.53 MPa for n-HAp prepared at 37°C) were obtained. XRD analyses on sintered n-HAp confirmed the thermal stability of the material. Therefore, the as-synthesized n-HAp powder can be successfully used for the fabrication of highly porous scaffolds as bone substitutes.
Collapse
|
176
|
Shui W, Zhang W, Yin L, Nan G, Liao Z, Zhang H, Wang N, Wu N, Chen X, Wen S, He Y, Deng F, Zhang J, Luu HH, Shi LL, Hu Z, Haydon RC, Mok JM, He TC. Characterization of scaffold carriers for BMP9-transduced osteoblastic progenitor cells in bone regeneration. J Biomed Mater Res A 2013; 102:3429-38. [PMID: 24133046 DOI: 10.1002/jbma.35006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 01/25/2023]
Abstract
Successful bone tissue engineering at least requires sufficient osteoblast progenitors, efficient osteoinductive factors, and biocompatible scaffolding materials. We have demonstrated that BMP9 is one of the most potent factors in inducing osteogenic differentiation of mesenchymal progenitors. To facilitate the potential use of cell-based BMP9 gene therapy for bone regeneration, we characterize the in vivo osteoconductive activities and bone regeneration potential of three clinically used scaffold materials, type I collagen sponge, hydroxyapatite-tricalcium phosphate (HA-TCP), and demineralized bone matrix (DBM), using BMP9-expressing C2C12 osteoblastic progenitor cells. We find that recombinant adenovirus-mediated BMP9 expression effectively induces osteogenic differentiation in C2C12 cells. Although direct subcutaneous injection of BMP9-transduced C2C12 cells forms ectopic bony masses, subcutaneous implantation of BMP9-expressing C2C12 cells with collagen sponge or HA-TCP scaffold yields the most robust and mature cancellous bone formation, whereas the DBM carrier group forms no or minimal bone masses. Our results suggest that collagen sponge and HA-TCP scaffold carriers may provide more cell-friendly environment to support the survival, propagation, and ultimately differentiation of BMP9-expressing progenitor cells. This line of investigation should provide important experimental evidence for further preclinical studies in BMP9-mediated cell-based approach to bone tissue engineering.
Collapse
|
177
|
Ansari S, Moshaverinia A, Pi SH, Han A, Abdelhamid AI, Zadeh HH. Functionalization of scaffolds with chimeric anti-BMP-2 monoclonal antibodies for osseous regeneration. Biomaterials 2013; 34:10191-8. [PMID: 24055525 DOI: 10.1016/j.biomaterials.2013.08.069] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/21/2013] [Indexed: 01/09/2023]
Abstract
Recent studies have demonstrated the ability of murine anti-BMP-2 monoclonal antibodies (mAb) immobilized on an absorbable collagen sponge (ACS) to mediate de novo bone formation, a process termed antibody-mediated osseous regeneration (AMOR). The objectives of this study were to assess the efficacy of a newly generated chimeric anti-BMP-2 mAb in mediating AMOR, as well as to evaluate the suitability of different biomaterials as scaffolds to participate in AMOR. Chimeric anti-BMP-2 mAb was immobilized on 4 biomaterials, namely, titanium microbeads (Ti), alginate hydrogel, macroporous biphasic calcium phosphate (MBCP) and ACS, followed by surgical implantation into rat critical-size calvarial defects. Animals were sacrificed after 8 weeks and the degree of bone fill was assessed using micro-CT and histomorphometry. Results demonstrated local persistence of chimeric anti-BMP-2 mAb up to 8 weeks, as well as significant de novo bone regeneration in sites implanted with chimeric anti-BMP-2 antibody immobilized on each of the 4 scaffolds. Ti and MBCP showed the highest volume of bone regeneration, presumably due to their resistance to compression. Alginate and ACS also mediated de novo bone formation, though significant volumetric shrinkage was noted. In vitro assays demonstrated cross-reactivity of chimeric anti-BMP-2 mAb with BMP-4 and BMP-7. Immune complex of anti-BMP-2 mAb with BMP-2 induced osteogenic differentiation of C2C12 cells in vitro, involving expression of RUNX2 and phosphorylation of Smad1. The present data demonstrated the ability of chimeric anti-BMP-2 mAb to functionalize different biomaterial with varying characteristics to mediate osteogenesis.
Collapse
Affiliation(s)
- Sahar Ansari
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
178
|
Recent developments of functional scaffolds for craniomaxillofacial bone tissue engineering applications. ScientificWorldJournal 2013; 2013:863157. [PMID: 24163634 PMCID: PMC3791836 DOI: 10.1155/2013/863157] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 07/14/2013] [Indexed: 12/15/2022] Open
Abstract
Autogenous bone grafting remains a gold standard for the reconstruction critical-sized bone defects in the craniomaxillofacial region. Nevertheless, this graft procedure has several disadvantages such as restricted availability, donor-site morbidity, and limitations in regard to fully restoring the complicated three-dimensional structures in the craniomaxillofacial bone. The ultimate goal of craniomaxillofacial bone reconstruction is the regeneration of the physiological bone that simultaneously fulfills both morphological and functional restorations. Developments of tissue engineering in the last two decades have brought such a goal closer to reality. In bone tissue engineering, the scaffolds are fundamental, elemental and mesenchymal stem cells/osteoprogenitor cells and bioactive factors. A variety of scaffolds have been developed and used as spacemakers, biodegradable bone substitutes for transplanting to the new bone, matrices of drug delivery system, or supporting structures enhancing adhesion, proliferation, and matrix production of seeded cells according to the circumstances of the bone defects. However, scaffolds to be clinically completely satisfied have not been developed yet. Development of more functional scaffolds is required to be applied widely to cranio-maxillofacial bone defects. This paper reviews recent trends of scaffolds for crania-maxillofacial bone tissue engineering, including our studies.
Collapse
|
179
|
Mehrotra D. TMJ Bioengineering: A review. J Oral Biol Craniofac Res 2013; 3:140-5. [PMID: 25737903 PMCID: PMC3941445 DOI: 10.1016/j.jobcr.2013.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 07/30/2013] [Indexed: 01/09/2023] Open
Abstract
Regeneration using scaffolds, growth factors, and stem cells is being investigated worldwide. Pubmed search for scaffolds for condyle resulted in 102 articles, of which 24 analyzed Temporomandibular joint (TMJ) scaffolds and only 6 evaluated hydroxyapatite scaffolds. 17 articles report studies on TMJ disc regeneration. The ideal bone construct for repair should be able to replicate the lost structure, restore function, be harmless, reliable and biodegradable. Scaffolds act as carriers for mesenchymal stem cells and/or growth factors and are useful for cell adhesion, migration, proliferation, and differentiation. Gene therapy has also led to the accelerated effective bone regeneration. The major materials used as scaffolds are natural or synthetic polymers, ceramics, composite materials, and electrospun nanofibers. Mesenchymal stem cells are responsible for the formation of virtually all dental, oral, and craniofacial structures. Tissue-engineered bone can possess the customized shape and dimensions. It has the potential for the biological replacement of craniofacial bones.
Collapse
Affiliation(s)
- Divya Mehrotra
- Professor, Department of Oral & Maxillofacial Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
180
|
Santo VE, Gomes ME, Mano JF, Reis RL. Controlled release strategies for bone, cartilage, and osteochondral engineering--Part I: recapitulation of native tissue healing and variables for the design of delivery systems. TISSUE ENGINEERING. PART B, REVIEWS 2013; 19:308-26. [PMID: 23268651 PMCID: PMC3690094 DOI: 10.1089/ten.teb.2012.0138] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 12/11/2012] [Indexed: 12/12/2022]
Abstract
The potential of growth factors to stimulate tissue healing through the enhancement of cell proliferation, migration, and differentiation is undeniable. However, critical parameters on the design of adequate carriers, such as uncontrolled spatiotemporal presence of bioactive factors, inadequate release profiles, and supraphysiological dosages of growth factors, have impaired the translation of these systems onto clinical practice. This review describes the healing cascades for bone, cartilage, and osteochondral interface, highlighting the role of specific growth factors for triggering the reactions leading to tissue regeneration. Critical criteria on the design of carriers for controlled release of bioactive factors are also reported, focusing on the need to provide a spatiotemporal control over the delivery and presentation of these molecules.
Collapse
Affiliation(s)
- Vítor E. Santo
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuela E. Gomes
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João F. Mano
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
181
|
Félix Lanao RP, Jonker AM, Wolke JG, Jansen JA, van Hest JC, Leeuwenburgh SC. Physicochemical properties and applications of poly(lactic-co-glycolic acid) for use in bone regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2013; 19:380-90. [PMID: 23350707 PMCID: PMC3690090 DOI: 10.1089/ten.teb.2012.0443] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 01/11/2013] [Indexed: 11/12/2022]
Abstract
Poly(lactic-co-glycolic acid) (PLGA) is the most often used synthetic polymer within the field of bone regeneration owing to its biocompatibility and biodegradability. As a consequence, a large number of medical devices comprising PLGA have been approved for clinical use in humans by the American Food and Drug Administration. As compared with the homopolymers of lactic acid poly(lactic acid) and poly(glycolic acid), the co-polymer PLGA is much more versatile with regard to the control over degradation rate. As a material for bone regeneration, the use of PLGA has been extensively studied for application and is included as either scaffolds, coatings, fibers, or micro- and nanospheres to meet various clinical requirements.
Collapse
Affiliation(s)
- Rosa P. Félix Lanao
- Department of Biomaterials, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Anika M. Jonker
- Institute for Molecules and Materials, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Joop G.C. Wolke
- Department of Biomaterials, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - John A. Jansen
- Department of Biomaterials, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Jan C.M. van Hest
- Institute for Molecules and Materials, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Sander C.G. Leeuwenburgh
- Department of Biomaterials, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
182
|
Moshaverinia A, Ansari S, Chen C, Xu X, Akiyama K, Snead ML, Zadeh HH, Shi S. Co-encapsulation of anti-BMP2 monoclonal antibody and mesenchymal stem cells in alginate microspheres for bone tissue engineering. Biomaterials 2013; 34:6572-9. [PMID: 23773817 DOI: 10.1016/j.biomaterials.2013.05.048] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/23/2013] [Indexed: 12/19/2022]
Abstract
Recently, it has been shown that tethered anti-BMP2 monoclonal antibodies (mAbs) can trap BMP ligands and thus provide BMP inductive signals for osteo-differentiation of progenitor cells. The objectives of this study were to: (1) develop a co-delivery system based on murine anti-BMP2 mAb-loaded alginate microspheres encapsulating human bone marrow mesenchymal stem cells (hBMMSCs); and (2) investigate osteogenic differentiation of encapsulated stem cells in alginate microspheres in vitro and in vivo. Alginate microspheres of 1 ± 0.1 mm diameter were fabricated with 2 × 10(6) hBMMSCs per mL of alginate. Critical-size calvarial defects (5 mm diameter) were created in immune-compromised mice and alginate microspheres preloaded with anti-BMP mAb encapsulating hBMMSCs were transplanted into defect sites. Alginate microspheres pre-loaded with isotype-matched non-specific antibody were used as the negative control. After 8 weeks, micro CT and histologic analyses were used to analyze bone formation. In vitro analysis demonstrated that anti-BMP2 mAbs tethered BMP2 ligands that can activate the BMP receptors on hBMMSCs. The co-delivery system described herein, significantly enhanced hBMMSC-mediated osteogenesis, as confirmed by the presence of BMP signal pathway-activated osteoblast determinants Runx2 and ALP. Our results highlight the importance of engineering the microenvironment for stem cells, and particularly the value of presenting inductive signals for osteo-differentiation of hBMMSCs by tethering BMP ligands using mAbs. This strategy of engineering the microenvironment with captured BMP signals is a promising modality for repair and regeneration of craniofacial, axial and appendicular bone defects.
Collapse
Affiliation(s)
- Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Amini AR, Laurencin CT, Nukavarapu SP. Bone tissue engineering: recent advances and challenges. Crit Rev Biomed Eng 2013; 40:363-408. [PMID: 23339648 DOI: 10.1615/critrevbiomedeng.v40.i5.10] [Citation(s) in RCA: 1350] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The worldwide incidence of bone disorders and conditions has trended steeply upward and is expected to double by 2020, especially in populations where aging is coupled with increased obesity and poor physical activity. Engineered bone tissue has been viewed as a potential alternative to the conventional use of bone grafts, due to their limitless supply and no disease transmission. However, bone tissue engineering practices have not proceeded to clinical practice due to several limitations or challenges. Bone tissue engineering aims to induce new functional bone regeneration via the synergistic combination of biomaterials, cells, and factor therapy. In this review, we discuss the fundamentals of bone tissue engineering, highlighting the current state of this field. Further, we review the recent advances of biomaterial and cell-based research, as well as approaches used to enhance bone regeneration. Specifically, we discuss widely investigated biomaterial scaffolds, micro- and nano-structural properties of these scaffolds, and the incorporation of biomimetic properties and/or growth factors. In addition, we examine various cellular approaches, including the use of mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), adult stem cells, induced pluripotent stem cells (iPSCs), and platelet-rich plasma (PRP), and their clinical application strengths and limitations. We conclude by overviewing the challenges that face the bone tissue engineering field, such as the lack of sufficient vascularization at the defect site, and the research aimed at functional bone tissue engineering. These challenges will drive future research in the field.
Collapse
Affiliation(s)
- Ami R Amini
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|
184
|
Wang X, Schröder HC, Feng Q, Draenert F, Müller WEG. The deep-sea natural products, biogenic polyphosphate (Bio-PolyP) and biogenic silica (Bio-Silica), as biomimetic scaffolds for bone tissue engineering: fabrication of a morphogenetically-active polymer. Mar Drugs 2013; 11:718-46. [PMID: 23528950 PMCID: PMC3705367 DOI: 10.3390/md11030718] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/04/2013] [Accepted: 02/06/2013] [Indexed: 12/12/2022] Open
Abstract
Bone defects in human, caused by fractures/nonunions or trauma, gain increasing impact and have become a medical challenge in the present-day aging population. Frequently, those fractures require surgical intervention which ideally relies on autografts or suboptimally on allografts. Therefore, it is pressing and likewise challenging to develop bone substitution materials to heal bone defects. During the differentiation of osteoblasts from their mesenchymal progenitor/stem cells and of osteoclasts from their hemopoietic precursor cells, a lineage-specific release of growth factors and a trans-lineage homeostatic cross-talk via signaling molecules take place. Hence, the major hurdle is to fabricate a template that is functioning in a way mimicking the morphogenetic, inductive role(s) of the native extracellular matrix. In the last few years, two naturally occurring polymers that are produced by deep-sea sponges, the biogenic polyphosphate (bio-polyP) and biogenic silica (bio-silica) have also been identified as promoting morphogenetic on both osteoblasts and osteoclasts. These polymers elicit cytokines that affect bone mineralization (hydroxyapatite formation). In this manner, bio-silica and bio-polyP cause an increased release of BMP-2, the key mediator activating the anabolic arm of the hydroxyapatite forming cells, and of RANKL. In addition, bio-polyP inhibits the progression of the pre-osteoclasts to functionally active osteoclasts. Based on these findings, new bioinspired strategies for the fabrication of bone biomimetic templates have been developed applying 3D-printing techniques. Finally, a strategy is outlined by which these two morphogenetically active polymers might be used to develop a novel functionally active polymer.
Collapse
Affiliation(s)
- Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany; E-Mail:
- National Research Center for Geoanalysis, Chinese Academy of Geological Sciences, 26 Baiwanzhuang Dajie, 100037 Beijing, China
| | - Heinz C. Schröder
- ERC Advanced Investigator Grant Research Group at Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany; E-Mail:
| | - Qingling Feng
- Department of Materials Science and Engineering, Tsinghua University, 100084 Beijing, China; E-Mail:
| | - Florian Draenert
- Department and Clinic for Oral and Maxillofacial Surgery, Baldingerstraße, D-35033 Marburg, Germany; E-Mail:
| | - Werner E. G. Müller
- ERC Advanced Investigator Grant Research Group at Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany; E-Mail:
| |
Collapse
|
185
|
Smolen D, Chudoba T, Malka I, Kedzierska A, Lojkowski W, Swieszkowski W, Kurzydlowski KJ, Kolodziejczyk-Mierzynska M, Lewandowska-Szumiel M. Highly biocompatible, nanocrystalline hydroxyapatite synthesized in a solvothermal process driven by high energy density microwave radiation. Int J Nanomedicine 2013; 8:653-68. [PMID: 23431124 PMCID: PMC3575162 DOI: 10.2147/ijn.s39299] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A microwave, solvothermal synthesis of highly biocompatible hydroxyapatite (HAp) nanopowder was developed. The process was conducted in a microwave radiation field having a high energy density of 5 W/mL and over a time less than 2 minutes. The sample measurements included: powder X-ray diffraction, density, specific surface area, and chemical composition. The morphology and structure were investigated by scanning electron microscopy as well as transmission electron microscopy (TEM). The thermal behavior analysis was conducted using a simultaneous thermal analysis technique coupled with quadruple mass spectrometry. Additionally, Fourier transform infrared spectroscopy tests of heated samples were performed. A degradation test and a biocompatibility study in vitro using human osteoblast cells were also conducted. The developed method enables the synthesis of pure, fully crystalline hexagonal HAp nanopowder with a specific surface area close to 240 m(2)/g and a Ca/P molar ratio equal to 1.57. TEM measurements showed that this method results in particles with an average grain size below 6 nm. A 28-day degradation test conducted according to the ISO standard indicated a 22% loss of initial weight and a calcium ion concentration at 200 μmol/dm(3) in the tris(hydroxymethyl)aminomethane hydrochloride test solution. The cytocompatibility of the obtained material was confirmed in a culture of human bone derived cells, both in an indirect test using the material extract, and in direct contact. A quantitative analysis was based on the 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide. Viability assay as well as on DNA content measurements in the PicoGreen test. Indirect observations were performed at one point in time according to the ISO standard for in vitro cytotoxicity (ie, after 24 hours of cell exposure to the extracts). The direct contact tests were completed at three time points: after 24 hours, on day 7, and on day 14 of a culture in an osteogenic medium. All of the tests revealed good tolerance of cells toward the material; this was also shown by means of live/dead fluorescent staining. Both quantitative results and morphological observations revealed much better cell tolerance toward the obtained HAp compared to commercially available HAp NanoXIM, which was used as a reference material.
Collapse
Affiliation(s)
- Dariusz Smolen
- Polish Academy of Science, Institute of High Pressure Physics, Warsaw, Poland.
| | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Ozbolat IT, Yu Y. Bioprinting toward organ fabrication: challenges and future trends. IEEE Trans Biomed Eng 2013; 60:691-9. [PMID: 23372076 DOI: 10.1109/tbme.2013.2243912] [Citation(s) in RCA: 316] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tissue engineering has been a promising field of research, offering hope for bridging the gap between organ shortage and transplantation needs. However, building three-dimensional (3-D) vascularized organs remains the main technological barrier to be overcome. Organ printing, which is defined as computer-aided additive biofabrication of 3-D cellular tissue constructs, has shed light on advancing this field into a new era. Organ printing takes advantage of rapid prototyping (RP) technology to print cells, biomaterials, and cell-laden biomaterials individually or in tandem, layer by layer, directly creating 3-D tissue-like structures. Here, we overview RP-based bioprinting approaches and discuss the current challenges and trends toward fabricating living organs for transplant in the near future.
Collapse
Affiliation(s)
- Ibrahim T Ozbolat
- Mechanical and Industrial Engineering Department, The University of Iowa, Iowa City, IA 52242, USA.
| | | |
Collapse
|
187
|
Biomimetic Assemblies by Matrix-Assisted Pulsed Laser Evaporation. LASER TECHNOLOGY IN BIOMIMETICS 2013. [DOI: 10.1007/978-3-642-41341-4_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
188
|
Gyawali D, Nair P, Kim HKW, Yang J. Citrate-based Biodegradable Injectable hydrogel Composites for Orthopedic Applications. Biomater Sci 2013; 1:52-64. [PMID: 23977427 DOI: 10.1039/c2bm00026a] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Previous studies have confirmed that natural bone apatite crystals are bound with citrate-rich molecules. Citrates on apatite crystals impact bone development and its load-bearing function. However, such understanding has never been translated into bone biomaterials design. Herein, a first citrate-based injectable composite material for orthopedic applications is developed based on our recently developed biodegradable poly(ethylene glycol) maleate citrate (PEGMC) and hydroxyapatite (HA). PEGMC contains rich carboxylic groups that could chelate with calcium-containing HA thus facilitating polymer/HA interactions, similar to natural citrate-bound apatite crystal. The crosslinking of poly(ethylene glycol) diacrylate (PEGDA) with PEGMC/HA composites allows an addition control over degradation and mechanical properties of the crosslinked PEGMC/HA (CPEGMC/HA) composites. CPEGMC/HA composite can serve as an ideal injectable cell carrier as confirmed by the enhanced DNA content, ALP activity, and calcium production through a human fetal osteoblast encapsulation study. Ex vivo study on porcine femoral head demonstrated that PEGMC/HA is a potentially promising injectable biodegradable bone material for the treatment of osteonecrosis of the femoral head. Development of biodegradable citrate-based injectable PEGMC/HA composite is an initial step for the development of the next generation of bone tissue engineering and orthopedic biomaterials.
Collapse
Affiliation(s)
- Dipendra Gyawali
- Department of Bioengineering, The University of Texas at Arlington, Arlington, TX ; Joint Biomedical Engineering Program, The University of Texas Southwestern Medical Center and The University of Texas at Arlington, Dallas, TX 75390
| | | | | | | |
Collapse
|
189
|
Schwab EH, Halbig M, Glenske K, Wagner AS, Wenisch S, Cavalcanti-Adam EA. Distinct effects of RGD-glycoproteins on Integrin-mediated adhesion and osteogenic differentiation of human mesenchymal stem cells. Int J Med Sci 2013; 10:1846-59. [PMID: 24324361 PMCID: PMC3856375 DOI: 10.7150/ijms.6908] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 10/05/2013] [Indexed: 12/27/2022] Open
Abstract
The detailed interactions of mesenchymal stem cells (MSCs) with their extracellular matrix (ECM) and the resulting effects on MSC differentiation are still largely unknown. Integrins are the main mediators of cell-ECM interaction. In this study, we investigated the adhesion of human MSCs to fibronectin, vitronectin and osteopontin, three ECM glycoproteins which contain an integrin-binding sequence, the RGD motif. We then assayed MSCs for their osteogenic commitment in the presence of the different ECM proteins. As early as 2 hours after seeding, human MSCs displayed increased adhesion when plated on fibronectin, whereas no significant difference was observed when adhering either to vitronectin or osteopontin. Over a 10-day observation period, cell proliferation was increased when cells were cultured on fibronectin and osteopontin, albeit after 5 days in culture. The adhesive role of fibronectin was further confirmed by measurements of cell area, which was significantly increased on this type of substrate. However, integrin-mediated clusters, namely focal adhesions, were larger and more mature in MSCs adhering to vitronectin and osteopontin. Adhesion to fibronectin induced elevated expression of α₅-integrin, which was further upregulated under osteogenic conditions also for vitronectin and osteopontin. In contrast, during osteogenic differentiation the expression level of β₃-integrin was decreased in MSCs adhering to the different ECM proteins. When MSCs were cultured under osteogenic conditions, their commitment to the osteoblast lineage and their ability to form a mineralized matrix in vitro was increased in presence of fibronectin and osteopontin. Taken together these results indicate a distinct role of ECM proteins in regulating cell adhesion, lineage commitment and phenotype of MSCs, which is due to the modulation of the expression of specific integrin subunits during growth or osteogenic differentiation.
Collapse
Affiliation(s)
- Elisabeth H Schwab
- 1. Department of Biophysical Chemistry, Institute for Physical Chemistry, University of Heidelberg, INF 253, 69120 Heidelberg, Germany & Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569 Stuttgart, Germany
| | | | | | | | | | | |
Collapse
|
190
|
Trends in tissue engineering for blood vessels. J Biomed Biotechnol 2012; 2012:956345. [PMID: 23251085 PMCID: PMC3518873 DOI: 10.1155/2012/956345] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 09/25/2012] [Indexed: 11/18/2022] Open
Abstract
Over the years, cardiovascular diseases continue to increase and affect not only human health but also the economic stability worldwide. The advancement in tissue engineering is contributing a lot in dealing with this immediate need of alleviating human health. Blood vessel diseases are considered as major cardiovascular health problems. Although blood vessel transplantation is the most convenient treatment, it has been delimited due to scarcity of donors and the patient's conditions. However, tissue-engineered blood vessels are promising alternatives as mode of treatment for blood vessel defects. The purpose of this paper is to show the importance of the advancement on biofabrication technology for treatment of soft tissue defects particularly for vascular tissues. This will also provide an overview and update on the current status of tissue reconstruction especially from autologous stem cells, scaffolds, and scaffold-free cellular transplantable constructs. The discussion of this paper will be focused on the historical view of cardiovascular tissue engineering and stem cell biology. The representative studies featured in this paper are limited within the last decade in order to trace the trend and evolution of techniques for blood vessel tissue engineering.
Collapse
|
191
|
Renth AN, Detamore MS. Leveraging "raw materials" as building blocks and bioactive signals in regenerative medicine. TISSUE ENGINEERING. PART B, REVIEWS 2012; 18:341-62. [PMID: 22462759 PMCID: PMC3458620 DOI: 10.1089/ten.teb.2012.0080] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 03/28/2012] [Indexed: 01/15/2023]
Abstract
Components found within the extracellular matrix (ECM) have emerged as an essential subset of biomaterials for tissue engineering scaffolds. Collagen, glycosaminoglycans, bioceramics, and ECM-based matrices are the main categories of "raw materials" used in a wide variety of tissue engineering strategies. The advantages of raw materials include their inherent ability to create a microenvironment that contains physical, chemical, and mechanical cues similar to native tissue, which prove unmatched by synthetic biomaterials alone. Moreover, these raw materials provide a head start in the regeneration of tissues by providing building blocks to be bioresorbed and incorporated into the tissue as opposed to being biodegraded into waste products and removed. This article reviews the strategies and applications of employing raw materials as components of tissue engineering constructs. Utilizing raw materials holds the potential to provide both a scaffold and a signal, perhaps even without the addition of exogenous growth factors or cytokines. Raw materials contain endogenous proteins that may also help to improve the translational success of tissue engineering solutions to progress from laboratory bench to clinical therapies. Traditionally, the tissue engineering triad has included cells, signals, and materials. Whether raw materials represent their own new paradigm or are categorized as a bridge between signals and materials, it is clear that they have emerged as a leading strategy in regenerative medicine. The common use of raw materials in commercial products as well as their growing presence in the research community speak to their potential. However, there has heretofore not been a coordinated or organized effort to classify these approaches, and as such we recommend that the use of raw materials be introduced into the collective consciousness of our field as a recognized classification of regenerative medicine strategies.
Collapse
Affiliation(s)
- Amanda N. Renth
- Bioengineering Program, University of Kansas, Lawrence, Kansas
| | - Michael S. Detamore
- Bioengineering Program, University of Kansas, Lawrence, Kansas
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas
| |
Collapse
|
192
|
Aw MS, Khalid KA, Gulati K, Atkins GJ, Pivonka P, Findlay DM, Losic D. Characterization of drug-release kinetics in trabecular bone from titania nanotube implants. Int J Nanomedicine 2012; 7:4883-92. [PMID: 23028217 PMCID: PMC3446838 DOI: 10.2147/ijn.s33655] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
PURPOSE The aim of this study was to investigate the application of the three-dimensional bone bioreactor for studying drug-release kinetics and distribution of drugs in the ex vivo cancellous bone environment, and to demonstrate the application of nanoengineered titanium (Ti) wires generated with titania nanotube (TNT) arrays as drug-releasing implants for local drug delivery METHODS Nanoengineered Ti wires covered with a layer of TNT arrays implanted in bone were used as a drug-releasing implant. Viable bovine trabecular bone was used as the ex vivo bone substrate embedded with the implants and placed in the bone reactor. A hydrophilic fluorescent dye (rhodamine B) was used as the model drug, loaded inside the TNT-Ti implants, to monitor drug release and transport in trabecular bone. The distribution of released model drug in the bone was monitored throughout the bone structure, and concentration profiles at different vertical (0-5 mm) and horizontal (0-10 mm) distances from the implant surface were obtained at a range of release times from 1 hour to 5 days. RESULTS Scanning electron microscopy confirmed that well-ordered, vertically aligned nanotube arrays were formed on the surface of prepared TNT-Ti wires. Thermogravimetric analysis proved loading of the model drug and fluorescence spectroscopy was used to show drug-release characteristics in-vitro. The drug release from implants inserted into bone ex vivo showed a consistent gradual release of model drug from the TNT-Ti implants, with a characteristic three-dimensional distribution into the surrounding bone, over a period of 5 days. The parameters including the flow rate of bone culture medium, differences in trabecular microarchitecture between bone samples, and mechanical loading were found to have the most significant influence on drug distribution in the bone. CONCLUSION These results demonstrate the utility of the Zetos™ system for ex vivo drug-release studies in bone, which can be applied to optimize the delivery of specific therapies and to assist in the design of new drug delivery systems. This method has the potential to provide new knowledge to understand drug distribution in the bone environment and to considerably improve existing technologies for local administration in bone, including solving some critical problems in bone therapy and orthopedic implants.
Collapse
Affiliation(s)
- Moom Sinn Aw
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, Australia
| | | | | | | | | | | | | |
Collapse
|
193
|
Mineralization Potential of Electrospun PDO-Hydroxyapatite-Fibrinogen Blended Scaffolds. Int J Biomater 2012; 2012:159484. [PMID: 22956956 PMCID: PMC3431095 DOI: 10.1155/2012/159484] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 07/05/2012] [Indexed: 11/17/2022] Open
Abstract
The current bone autograft procedure for cleft palate repair presents several disadvantages such as limited availability, additional invasive surgery, and donor site morbidity. The present preliminary study evaluates the mineralization potential of electrospun polydioxanone:nano-hydroxyapatite : fibrinogen (PDO : nHA : Fg) blended scaffolds in different simulated body fluids (SBF). Scaffolds were fabricated by blending PDO : nHA : Fg in the following percent by weight ratios: 100 : 0 : 0, 50 : 25 : 25, 50 : 50 : 0, 50 : 0 : 50, 0 : 0 : 100, and 0 : 50 : 50. Samples were immersed in (conventional (c), revised (r), ionic (i), and modified (m)) SBF for 5 and 14 days to induce mineralization. Scaffolds were characterized before and after mineralization via scanning electron microscopy, Alizarin Red-based assay, and modified burnout test. The addition of Fg resulted in scaffolds with smaller fiber diameters. Fg containing scaffolds also induced sheet-like mineralization while individual fiber mineralization was noticed in its absence. Mineralized electrospun Fg scaffolds without PDO were not mechanically stable after 5 days in SBF, but had superior mineralization capabilities which produced a thick bone-like mineral (BLM) layer throughout the scaffolds. 50 : 50 : 0 scaffolds incubated in either r-SBF for 5 days or c-SBF for 14 days produced scaffolds with high mineral content and individual-mineralized fibers. These mineralized scaffolds were still porous and will be further optimized as an effective bone substitute in future studies.
Collapse
|
194
|
Chen M, Le DQS, Hein S, Li P, Nygaard JV, Kassem M, Kjems J, Besenbacher F, Bünger C. Fabrication and characterization of a rapid prototyped tissue engineering scaffold with embedded multicomponent matrix for controlled drug release. Int J Nanomedicine 2012; 7:4285-97. [PMID: 22904634 PMCID: PMC3418070 DOI: 10.2147/ijn.s33083] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Indexed: 11/23/2022] Open
Abstract
Bone tissue engineering implants with sustained local drug delivery provide an opportunity for better postoperative care for bone tumor patients because these implants offer sustained drug release at the tumor site and reduce systemic side effects. A rapid prototyped macroporous polycaprolactone scaffold was embedded with a porous matrix composed of chitosan, nanoclay, and β-tricalcium phosphate by freeze-drying. This composite scaffold was evaluated on its ability to deliver an anthracycline antibiotic and to promote formation of mineralized matrix in vitro. Scanning electronic microscopy, confocal imaging, and DNA quantification confirmed that immortalized human bone marrow-derived mesenchymal stem cells (hMSC-TERT) cultured in the scaffold showed high cell viability and growth, and good cell infiltration to the pores of the scaffold. Alkaline phosphatase activity and osteocalcin staining showed that the scaffold was osteoinductive. The drug-release kinetics was investigated by loading doxorubicin into the scaffold. The scaffolds comprising nanoclay released up to 45% of the drug for up to 2 months, while the scaffold without nanoclay released 95% of the drug within 4 days. Therefore, this scaffold can fulfill the requirements for both bone tissue engineering and local sustained release of an anticancer drug in vitro. These results suggest that the scaffold can be used clinically in reconstructive surgery after bone tumor resection. Moreover, by changing the composition and amount of individual components, the scaffold can find application in other tissue engineering areas that need local sustained release of drug.
Collapse
Affiliation(s)
- Muwan Chen
- Orthopaedic Research Lab, Aarhus University Hospital, Aarhus C, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Florczyk SJ, Leung M, Jana S, Li Z, Bhattarai N, Huang JI, Hopper RA, Zhang M. Enhanced bone tissue formation by alginate gel-assisted cell seeding in porous ceramic scaffolds and sustained release of growth factor. J Biomed Mater Res A 2012; 100:3408-15. [PMID: 22767533 DOI: 10.1002/jbm.a.34288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 04/09/2012] [Accepted: 05/15/2012] [Indexed: 12/26/2022]
Abstract
Increasing cell seeding efficiency in a tissue engineering construct can enhance cellular activity and tissue formation in vivo. Here, we demonstrate the use of alginate gel as a secondary phase material in 3D porous β-tricalcium phosphate scaffolds to improve cell seeding and provide controlled release of growth factors for bone tissue engineering. Cells were seeded in scaffolds in three ways: conventional seeding (CS), alginate gel-assisted seeding (GS), and alginate GS with bone morphogenetic protein-2 (BMP-2, GSB). In vitro study with MG-63 cells showed that cell seeding efficiency and cell population 1 week after seeding were significantly elevated in GS and GSB samples compared to CS samples. The GSB system demonstrated a sustained, steady release of BMP-2 over 2 weeks. In vivo, scaffolds seeded with rat mesenchymal stem cells were implanted ectopically into Sprague-Dawley rats for 8 weeks. GS and GSB samples exhibited improved osteogenic activity, with the GSB samples inducing the greatest osteocalcin and osteoid deposition. This study suggests that the alginate gel-assisted cell seeding increases seeding efficiency and allows for sustained release of growth factors. The use of the secondary phase polymer bolsters bone formation in vivo and has the potential for improving outcome in other tissue engineering applications.
Collapse
Affiliation(s)
- Stephen J Florczyk
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195-2120, USA
| | | | | | | | | | | | | | | |
Collapse
|
196
|
Rao RR, Jiao A, Kohn DH, Stegemann JP. Exogenous mineralization of cell-seeded and unseeded collagen-chitosan hydrogels using modified culture medium. Acta Biomater 2012; 8:1560-5. [PMID: 22266029 DOI: 10.1016/j.actbio.2012.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 12/06/2011] [Accepted: 01/04/2012] [Indexed: 11/24/2022]
Abstract
Induced biomineralization of materials has been employed as a strategy to increase integration with host tissue, and more recently as a method to control cell function in tissue engineering. However, mineralization is typically performed in the absence of cells, since hypertonic solutions that lack the nutrients and culture components required for the maintenance of cell viability are often used. In the present study, we exposed fibroblast-seeded three-dimensional collagen-chitosan hydrogels to a defined culture medium modified to have specific concentrations of ions involved in biomineralization. The modified medium caused a significant increase in calcium deposition in collagen-chitosan gels, relative to constructs incubated in a standard medium, though serum supplementation attenuated mineral deposition. Collagen-chitosan constructs became opaque over 3 days of mineralization in modified Dulbecco's modified Eagle medium (DMEM), in contrast to translucent control gels incubated in standard DMEM. Histological staining confirmed increased levels of mineral in the treated constructs. Rheological characterization showed that both the storage and loss moduli increased significantly in mineralized materials. Mineralization of fibroblast-seeded constructs resulted in decreased cell viability and proliferation rate over 3 days of incubation in modified medium, but the cell population remained over 75% viable and regained its proliferative potential after rescue in standard culture medium. The ability to mineralize protein matrices in the presence of cells could be useful in creating mechanically stable tissue constructs, as well as to study the effects of the tissue microenvironment on cell function.
Collapse
|
197
|
Bose S, Tarafder S. Calcium phosphate ceramic systems in growth factor and drug delivery for bone tissue engineering: a review. Acta Biomater 2012; 8:1401-21. [PMID: 22127225 DOI: 10.1016/j.actbio.2011.11.017] [Citation(s) in RCA: 490] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 11/04/2011] [Accepted: 11/13/2011] [Indexed: 12/12/2022]
Abstract
Calcium phosphates (CaPs) are the most widely used bone substitutes in bone tissue engineering due to their compositional similarities to bone mineral and excellent biocompatibility. In recent years, CaPs, especially hydroxyapatite and tricalcium phosphate, have attracted significant interest in simultaneous use as bone substitute and drug delivery vehicle, adding a new dimension to their application. CaPs are more biocompatible than many other ceramic and inorganic nanoparticles. Their biocompatibility and variable stoichiometry, thus surface charge density, functionality, and dissolution properties, make them suitable for both drug and growth factor delivery. CaP matrices and scaffolds have been reported to act as delivery vehicles for growth factors and drugs in bone tissue engineering. Local drug delivery in musculoskeletal disorder treatments can address some of the critical issues more effectively and efficiently than the systemic delivery. CaPs are used as coatings on metallic implants, CaP cements, and custom designed scaffolds to treat musculoskeletal disorders. This review highlights some of the current drug and growth factor delivery approaches and critical issues using CaP particles, coatings, cements, and scaffolds towards orthopedic and dental applications.
Collapse
|
198
|
Abstract
There remains a substantial shortfall in the treatment of severe skeletal injuries. The current gold standard of autologous bone grafting from the same patient has many undesirable side effects associated such as donor site morbidity. Tissue engineering seeks to offer a solution to this problem. The primary requirements for tissue-engineered scaffolds have already been well established, and many materials, such as polyesters, present themselves as potential candidates for bone defects; they have comparable structural features, but they often lack the required osteoconductivity to promote adequate bone regeneration. By combining these materials with biological growth factors, which promote the infiltration of cells into the scaffold as well as the differentiation into the specific cell and tissue type, it is possible to increase the formation of new bone. However due to the cost and potential complications associated with growth factors, controlling the rate of release is an important design consideration when developing new bone tissue engineering strategies. This paper will cover recent research in the area of encapsulation and release of growth factors within a variety of different polymeric scaffolds.
Collapse
|
199
|
Guo X, Chen M, Feng W, Liang J, Zhao H, Tian L, Chao H, Zou X. Electrostatic self-assembly of multilayer copolymeric membranes on the surface of porous tantalum implants for sustained release of doxorubicin. Int J Nanomedicine 2011; 6:3057-64. [PMID: 22162662 PMCID: PMC3230572 DOI: 10.2147/ijn.s25918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Many studies in recent years have focused on surface engineering of implant materials in order to improve their biocompatibility and other performance. Porous tantalum implants have increasingly been used in implant surgeries, due to their biocompatibility, physical stability, and good mechanical strength. In this study we functionalized the porous tantalum implant for sustained drug delivery capability via electrostatic self-assembly of polyelectrolytes of hyaluronic acid, methylated collagen, and terpolymer on the surface of a porous tantalum implant. The anticancer drug doxorubicin was encapsulated into the multilayer copolymer membranes on the porous tantalum implants. Results showed the sustained released of doxorubicin from the functionalized porous tantalum implants for up to 1 month. The drug release solutions in 1 month all had inhibitory effects on the proliferation of chondrosarcoma cell line SW1353. These results suggest that this functionalized implant could be used in reconstructive surgery for the treatment of bone tumor as a local, sustained drug delivery system.
Collapse
Affiliation(s)
- Xinming Guo
- Orthopaedic Research institute/ Department of Orthopaedic Surgery, First Affiliated Hospital and Department of Pharmacy, Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
200
|
Ghaemmaghami AM, Hancock MJ, Harrington H, Kaji H, Khademhosseini A. Biomimetic tissues on a chip for drug discovery. Drug Discov Today 2011; 17:173-81. [PMID: 22094245 DOI: 10.1016/j.drudis.2011.10.029] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 10/04/2011] [Accepted: 10/31/2011] [Indexed: 01/09/2023]
Abstract
Developing biologically relevant models of human tissues and organs is an important enabling step for disease modeling and drug discovery. Recent advances in tissue engineering, biomaterials and microfluidics have led to the development of microscale functional units of such models also referred to as 'organs on a chip'. In this review, we provide an overview of key enabling technologies and highlight the wealth of recent work regarding on-chip tissue models. In addition, we discuss the current challenges and future directions of organ-on-chip development.
Collapse
Affiliation(s)
- Amir M Ghaemmaghami
- Division of Immunology, School of Molecular Medical Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | | | |
Collapse
|