1
|
Chen M, Ren M, Liu X, Wang Z, Shi Y, Wu Z, Wang X, Zhang P, Wei H. Synergistic enhancement of angiogenesis and osseointegration in 3D-printed porous polyetheretherketone scaffolds using biomimetic coatings of bone morphogenetic protein-2/fibronectin. Int J Biol Macromol 2025; 297:139876. [PMID: 39814279 DOI: 10.1016/j.ijbiomac.2025.139876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
This study explored a novel modification method for porous polyetheretherketone (PEEK) implants using a biomimetic coating to achieve synergistic enhancement of vascularization and bone regeneration. Inspired by the natural extracellular matrix (ECM) structure (consists of growth factors and matrix proteins), a biomimetic dual-factor coating capable of releasing bone morphogenetic protein-2 (BMP-2) and fibronectin (FN) was coated on the surface of 3D-printed porous PEEK scaffolds using polydopamine (PDA) as a binder. Experiments conducted with MC3T3-E1 cells or HUVECs in co-culture with scaffolds revealed that the biomimetic coating not only synergically promoted cell migration, adhesion and proliferation, but also enhanced angiogenesis and osteogenic differentiation simultaneously in vivo. The synergistic effect is attributed to the crosstalks between intracellular signaling pathways of FN and BMP-2, as well as the sustained release on account of their combination mitigates explosive release and degradation of BMP-2. Overall, this study designed a novel biomimetic coating modified PEEK scaffolds and confirmed the synergistic mechanism of the scaffolds on osteogenic differentiation and angiogenesis for the first time. These insights have significant implications for the clinical transformation of PEEK dental implants.
Collapse
Affiliation(s)
- Meiqing Chen
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Mei Ren
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Xiuyu Liu
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yingqi Shi
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhenxu Wu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xinyu Wang
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Hongtao Wei
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
2
|
Tao Z, Yang M, Shen CL. Tauroursodeoxycholic acid combined with selenium accelerates bone regeneration in ovariectomized rats. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:64. [PMID: 39404912 PMCID: PMC11480188 DOI: 10.1007/s10856-024-06803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/20/2024] [Indexed: 10/19/2024]
Abstract
More recently, increased studies have revealed that antioxidants can cure osteoporosis by inhibiting oxidative stress. Tauroursodeoxycholic acid (TUDCA) and Selenium (Se) have been confirmed to possess potent anti-oxidative effects and accelerate bone regeneration. In addition, very little is currently known about the effects of a combination with Se and TUDCA on bone defects in osteoporotic states. We, therefore, aimed to assess the protective effect of combination with Se and TUDCA on bone regeneration and investigate the effect and underlying mechanisms. When MC3T3-E1 was cultured in the presence of H2H2, Se, TUDCA and Se/TUDCA therapy could increase the matrix mineralization and promote expression of anti-oxidative stress markers in MC3T3-E1, while reducing intracellular reactive oxygen species (ROS) and mitochondrial ROS levels. Meanwhile, silent information regulator type 1 (SIRT1) was upregulated in response to Se, TUDCA and Se/TUDCA exposures in H2H2 treated-MC3T3-E1. In the OVX rat model, Se, TUDCA and Se/TUDCA showed a clear positive effect against impaired bone repair in osteoporosis. The results above demonstrate that Se/TUDCA exhibits superior efficacy in both cellular and animal experiments, as compared to Se and TUDCA. In conclusion, combination with Se and TUDCA stimulates bone regeneration and is a promising candidate for promoting bone repair in osteoporosis.
Collapse
Affiliation(s)
- ZhouShan Tao
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China
| | - Min Yang
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China.
| | - Cai-Liang Shen
- Department of Spinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| |
Collapse
|
3
|
Dick TA, Sone ED, Uludağ H. Mineralized vectors for gene therapy. Acta Biomater 2022; 147:1-33. [PMID: 35643193 DOI: 10.1016/j.actbio.2022.05.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/01/2022]
Abstract
There is an intense interest in developing materials for safe and effective delivery of polynucleotides using non-viral vectors. Mineralization of organic templates has long been used to produce complex materials with outstanding biocompatibility. However, a lack of control over mineral growth has limited the applicability of mineralized materials to a few in vitro applications. With better control over mineral growth and surface functionalization, mineralized vectors have advanced significantly in recent years. Here, we review the recent progress in chemical synthesis, physicochemical properties, and applications of mineralized materials in gene therapy, focusing on structure-function relationships. We contrast the classical understanding of the mineralization mechanism with recent ideas of mineralization. A brief introduction to gene delivery is summarized, followed by a detailed survey of current mineralized vectors. The vectors derived from calcium phosphate are articulated and compared to other minerals with unique features. Advanced mineral vectors derived from templated mineralization and specialty coatings are critically analyzed. Mineral systems beyond the co-precipitation are explored as more complex multicomponent systems. Finally, we conclude with a perspective on the future of mineralized vectors by carefully demarcating the boundaries of our knowledge and highlighting ambiguous areas in mineralized vectors. STATEMENT OF SIGNIFICANCE: Therapy by gene-based medicines is increasingly utilized to cure diseases that are not alleviated by conventional drug therapy. Gene medicines, however, rely on macromolecular nucleic acids that are too large and too hydrophilic for cellular uptake. Without tailored materials, they are not functional for therapy. One emerging class of nucleic acid delivery system is mineral-based materials. The fact that they can undergo controlled dissolution with minimal footprint in biological systems are making them attractive for clinical use, where safety is utmost importance. In this submission, we will review the emerging synthesis technology and the range of new generation minerals for use in gene medicines.
Collapse
|
4
|
Pang K, Seo YK, Lee JH. Effects of the combination of bone morphogenetic protein-2 and nano-hydroxyapatite on the osseointegration of dental implants. J Korean Assoc Oral Maxillofac Surg 2021; 47:454-464. [PMID: 34969019 PMCID: PMC8721409 DOI: 10.5125/jkaoms.2021.47.6.454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES This study aimed to investigate the in vitro osteoinductivity of the combination of bone morphogenetic protein-2 (BMP-2) and nanohydroxyapatite (nHAp) and the in vivo effects of implants coated with nHAp/BMP-2. MATERIALS AND METHODS To evaluate the in vitro efficacy of nHAp/BMP-2 on bone formation, bone marrow-derived mesenchymal stem cells (BMMSCs) were seeded onto titanium disks coated with collagen (Col), Col/nHAp, or Col/nHAp/BMP-2. Protein levels were determined by a biochemical assay and reverse transcriptase-polymerase chain reaction. Stem cell differentiation was analyzed by flow cytometry. For in vivo studies with mice, Col, Col/nHAp, and Col/nHAp/BMP-2 were injected in subcutaneous pockets. Titanium implants or implants coated with Col/nHAp/BMP-2 were placed bilaterally on rabbit tibias and evaluated for 4 weeks. RESULTS In the in vitro study, BM-MSCs on Col/nHAp/BMP-2 showed reduced levels of CD73, CD90, and CD105 and increased levels of glycosaminoglycan, osteopontin, and alkaline phosphatase activity. After 4 weeks, the Col/nHAp/BMP-2 implant showed greater bone formation than the control (P=0.07), while no differences were observed in bone implant contact and removal torque. CONCLUSION These results suggest that a combination of BMP-2 and an nHAp carrier would activate osseointegration on dental implant surfaces.
Collapse
Affiliation(s)
- KangMi Pang
- Department of Dentistry, Oral and Maxillofacial Surgery, Seoul National University Dental Hospital, Seoul, Korea
| | - Young-Kwon Seo
- Department of Medical Biotechnology, College of Life Science and Biotechnology, Dongguk University, Seoul, Korea
| | - Jong-Ho Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, , Seoul, Korea
- Dental Life Science Research Institute and Clinical Translational Research Center for Dental Science, Seoul National University Dental Hospital, Seoul, Korea
| |
Collapse
|
5
|
Nedorubova IA, Bukharova TB, Vasilyev AV, Goldshtein DV, Kulakov AA. Non-viral delivery of the BMP2 gene for bone regeneration. GENES & CELLS 2020; 15:33-39. [DOI: 10.23868/202012005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Gene-activated bone grafts and substitutes are promising tools for the bone defect healing, which are capable to induce prolonged production of growth factors with a therapeutic effect at physiological concentrations. Non-viral methods of delivering plasmid constructs with target genes are the safest for clinical use, but their efficiency is lower in comparison with viral vectors. To solve the problem of plasmid delivery into cells, some systems with a high transfection capacity and ensure sufficient cell viability are being developed. Moreover, there are different approaches to improve the level of expression of target genes and targeted delivery to the bone defect in order to achieve local therapeutic concentrations. This review considers approaches which are aimed to increase the efficiency of bone tissue regeneration methods based on non-viral delivery systems for osteoinduction genes using the example of the bone morphogenetic protein-2 gene.
Collapse
|
6
|
BMP-2 Gene Delivery-Based Bone Regeneration in Dentistry. Pharmaceutics 2019; 11:pharmaceutics11080393. [PMID: 31387267 PMCID: PMC6723260 DOI: 10.3390/pharmaceutics11080393] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/22/2019] [Accepted: 08/02/2019] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic protein-2 (BMP-2) is a potent growth factor affecting bone formation. While recombinant human BMP-2 (rhBMP-2) has been commercially available in cases of non-union fracture and spinal fusion in orthopaedics, it has also been applied to improve bone regeneration in challenging cases requiring dental implant treatment. However, complications related to an initially high dosage for maintaining an effective physiological concentration at the defect site have been reported, although an effective and safe rhBMP-2 dosage for bone regeneration has not yet been determined. In contrast to protein delivery, BMP-2 gene transfer into the defect site induces BMP-2 synthesis in vivo and leads to secretion for weeks to months, depending on the vector, at a concentration of nanograms per milliliter. BMP-2 gene delivery is advantageous for bone wound healing process in terms of dosage and duration. However, safety concerns related to viral vectors are one of the hurdles that need to be overcome for gene delivery to be used in clinical practice. Recently, commercially available gene therapy has been introduced in orthopedics, and clinical trials in dentistry have been ongoing. This review examines the application of BMP-2 gene therapy for bone regeneration in the oral and maxillofacial regions and discusses future perspectives of BMP-2 gene therapy in dentistry.
Collapse
|
7
|
Yang S, Guo Y, Zhang W, Zhang J, Zhang Y, Xu P. Effect of FGF-21 on implant bone defects through hepatocyte growth factor (HGF)-mediated PI3K/AKT signaling pathway. Biomed Pharmacother 2018; 109:1259-1267. [PMID: 30551376 DOI: 10.1016/j.biopha.2018.10.150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 11/28/2022] Open
Abstract
Implant bone defects are the most common phenomenon in the processes of bone transplantation. Evidences have identified that fibroblast growth factor-21 (FGF-21) encourages osteogenesis for patients with implant bone defects. The purpose of this study was to investigate the role of FGF-21 and its potential mechanism in bone mesenchymal stem cells (BMSCs). RT-PCR, Western blotting, flow cytometry, immunofluorescence and immunohistochemistry assays were performed to analyze the role of FGF-21 and intracellular signaling pathways involved in BMSCs. It was shown that FGF-21 increased viability of BMSCs. Treatment with FGF-21 decreased the apoptosis of BMSCs by decreasing pro-apoptosis protein Caspase-3. Results indicated that FGF-21 (2 mg/kg) treatment up-regulated HGF, PI3K and AKT expression in BMSCs. In addition, the protective effects of FGF-21 on BMSCs were canceled by PI3K/AKT inhibitor in BMSCs. Results found that knockdown of HGF abolished FGF-21-decreased PI3K/AKT signal pathway. Furthermore, results demonstrated that FGF-21 presented beneficial effects for implant bone defects in rat model. In conclusion, these results indicate that FGF-21 can improve implant bone defects through HGF-mediated PI3K/AKT signaling pathway in BMSCs.
Collapse
Affiliation(s)
- Shimao Yang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Yanwei Guo
- Department of Oral and Maxillofacial Surgery, Jining Stomatology Hospital, Jining City, Shandong Province, China
| | - Wenmei Zhang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Jin Zhang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Yujie Zhang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Peng Xu
- Department of Dental Implant, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China.
| |
Collapse
|
8
|
Controllable and durable release of BMP-2-loaded 3D porous sulfonated polyetheretherketone (PEEK) for osteogenic activity enhancement. Colloids Surf B Biointerfaces 2018; 171:668-674. [DOI: 10.1016/j.colsurfb.2018.08.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 01/07/2023]
|
9
|
Duruel T, Çakmak AS, Akman A, Nohutcu RM, Gümüşderelioğlu M. Sequential IGF-1 and BMP-6 releasing chitosan/alginate/PLGA hybrid scaffolds for periodontal regeneration. Int J Biol Macromol 2017; 104:232-241. [DOI: 10.1016/j.ijbiomac.2017.06.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 05/20/2017] [Accepted: 06/06/2017] [Indexed: 10/19/2022]
|
10
|
Oyane A, Araki H, Nakamura M, Shimizu Y, Shubhra QT, Ito A, Tsurushima H. Controlled superficial assembly of DNA–amorphous calcium phosphate nanocomposite spheres for surface-mediated gene delivery. Colloids Surf B Biointerfaces 2016; 141:519-527. [DOI: 10.1016/j.colsurfb.2016.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/01/2016] [Accepted: 02/05/2016] [Indexed: 10/22/2022]
|
11
|
Uskoković V. When 1+1>2: Nanostructured composites for hard tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 57:434-51. [PMID: 26354283 PMCID: PMC4567690 DOI: 10.1016/j.msec.2015.07.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/15/2015] [Accepted: 07/23/2015] [Indexed: 12/20/2022]
Abstract
Multicomponent, synergistic and multifunctional nanostructures have taken over the spotlight in the realm of biomedical nanotechnologies. The most prospective materials for bone regeneration today are almost exclusively composites comprising two or more components that compensate for the shortcomings of each one of them alone. This is quite natural in view of the fact that all hard tissues in the human body, except perhaps the tooth enamel, are composite nanostructures. This review article highlights some of the most prospective breakthroughs made in this research direction, with the hard tissues in main focus being those comprising bone, tooth cementum, dentin and enamel. The major obstacles to creating collagen/apatite composites modeled after the structure of bone are mentioned, including the immunogenicity of xenogeneic collagen and continuously failing attempts to replicate the biomineralization process in vitro. Composites comprising a polymeric component and calcium phosphate are discussed in light of their ability to emulate the soft/hard composite structure of bone. Hard tissue engineering composites created using hard material components other than calcium phosphates, including silica, metals and several types of nanotubes, are also discoursed on, alongside additional components deliverable using these materials, such as cells, growth factors, peptides, antibiotics, antiresorptive and anabolic agents, pharmacokinetic conjugates and various cell-specific targeting moieties. It is concluded that a variety of hard tissue structures in the body necessitates a similar variety of biomaterials for their regeneration. The ongoing development of nanocomposites for bone restoration will result in smart, theranostic materials, capable of acting therapeutically in direct feedback with the outcome of in situ disease monitoring at the cellular and subcellular scales. Progress in this research direction is expected to take us to the next generation of biomaterials, designed with the purpose of fulfilling Daedalus' dream - not restoring the tissues, but rather augmenting them.
Collapse
Affiliation(s)
- Vuk Uskoković
- Advanced Materials and Nanobiotechnology Laboratory, Department of Bioengineering, University of Illinois, Chicago, IL, USA.
| |
Collapse
|
12
|
Yazaki Y, Oyane A, Sogo Y, Ito A, Yamazaki A, Tsurushima H. Area-specific cell stimulation via surface-mediated gene transfer using apatite-based composite layers. Int J Mol Sci 2015; 16:8294-309. [PMID: 25874757 PMCID: PMC4425081 DOI: 10.3390/ijms16048294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 11/16/2022] Open
Abstract
Surface-mediated gene transfer systems using biocompatible calcium phosphate (CaP)-based composite layers have attracted attention as a tool for controlling cell behaviors. In the present study we aimed to demonstrate the potential of CaP-based composite layers to mediate area-specific dual gene transfer and to stimulate cells on an area-by-area basis in the same well. For this purpose we prepared two pairs of DNA–fibronectin–apatite composite (DF-Ap) layers using a pair of reporter genes and pair of differentiation factor genes. The results of the area-specific dual gene transfer successfully demonstrated that the cells cultured on a pair of DF-Ap layers that were adjacently placed in the same well showed specific gene expression patterns depending on the gene that was immobilized in theunderlying layer. Moreover, preliminary real-time PCR results indicated that multipotential C3H10T1/2 cells may have a potential to change into different types of cells depending on the differentiation factor gene that was immobilized in the underlying layer, even in the same well. Because DF-Ap layers have a potential to mediate area-specific cell stimulation on their surfaces, they could be useful in tissue engineering applications.
Collapse
Affiliation(s)
- Yushin Yazaki
- Department of Resources and Environmental Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan.
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology, Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8562, Japan.
| | - Ayako Oyane
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology, Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8562, Japan.
| | - Yu Sogo
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.
| | - Atsuo Ito
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.
| | - Atsushi Yamazaki
- Department of Resources and Environmental Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan.
| | - Hideo Tsurushima
- Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology, Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8562, Japan.
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
13
|
Uskoković V. Nanostructured platforms for the sustained and local delivery of antibiotics in the treatment of osteomyelitis. Crit Rev Ther Drug Carrier Syst 2015; 32:1-59. [PMID: 25746204 PMCID: PMC4406243 DOI: 10.1615/critrevtherdrugcarriersyst.2014010920] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This article provides a critical view of the current state of the development of nanoparticulate and other solid-state carriers for the local delivery of antibiotics in the treatment of osteomyelitis. Mentioned are the downsides of traditional means for treating bone infection, which involve systemic administration of antibiotics and surgical debridement, along with the rather imperfect local delivery options currently available in the clinic. Envisaged are more sophisticated carriers for the local and sustained delivery of antimicrobials, including bioresorbable polymeric, collagenous, liquid crystalline, and bioglass- and nanotube-based carriers, as well as those composed of calcium phosphate, the mineral component of bone and teeth. A special emphasis is placed on composite multifunctional antibiotic carriers of a nanoparticulate nature and on their ability to induce osteogenesis of hard tissues demineralized due to disease. An ideal carrier of this type would prevent the long-term, repetitive, and systemic administration of antibiotics and either minimize or completely eliminate the need for surgical debridement of necrotic tissue. Potential problems faced by even hypothetically "perfect" antibiotic delivery vehicles are mentioned too, including (i) intracellular bacterial colonies involved in recurrent, chronic osteomyelitis; (ii) the need for mechanical and release properties to be adjusted to the area of surgical placement; (iii) different environments in which in vitro and in vivo testings are carried out; (iv) unpredictable synergies between drug delivery system components; and (v) experimental sensitivity issues entailing the increasing subtlety of the design of nanoplatforms for the controlled delivery of therapeutics.
Collapse
Affiliation(s)
- Vuk Uskoković
- Advanced Materials and Nanobiotechnology Laboratory, Richard and Loan Hill Department of Bioengineering, College of Medicine, University of Illinois at Chicago, 851 South Morgan St, #205 Chicago, Illinois, 60607-7052
| |
Collapse
|
14
|
Zou S, Chen T, Wang Y, Tian R, Zhang L, Song P, Yang S, Zhu Y, Guo X, Huang Y, Li Z, Kan L, Hu H. Mesenchymal stem cells overexpressing Ihh promote bone repair. J Orthop Surg Res 2014; 9:102. [PMID: 25346272 PMCID: PMC4213494 DOI: 10.1186/s13018-014-0102-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/10/2014] [Indexed: 12/26/2022] Open
Abstract
Background Indian hedgehog (Ihh) signaling pathway is known to play key roles in various aspects of normal endochondral bone development. This study tested the potential roles of high Ihh signaling in the context of injury-induced bone regeneration. Methods A rabbit tibia defect model was established to test the effects of the implant of Ihh/mesenchymal stem cells (MSCs)/scaffold complex. Computed tomography (CT), gross observation, and standard histological and immunohistological techniques were used to evaluate the effectiveness of the treatment. In vitro studies with MSCs and C3H10T1/2 cells were also employed to further understand the cellular and molecular mechanisms. Results We found that the implanted Ihh/MSCs/scaffold complex promoted bone repair. Consistently, in vitro study found that Ihh induced the upregulation of chondrocytic, osteogenic, and vascular cell markers, both in C3H10T1/2 cells and MSCs. Conclusions Our study has demonstrated that high Ihh signaling in a complex with MSCs enhanced bone regeneration effectively in a clinically relevant acute injury model. Even though the exact underlying mechanisms are still far from clear, our primary data suggested that enhanced chondrogenesis, osteogenesis, and angiogenesis of MSCs at least partially contribute to the process. This study not only has implications for basic research of MSCs and Ihh signaling pathway but also points to the possibility of direct application of this specific paradigm to clinical bone repair. Electronic supplementary material The online version of this article (doi:10.1186/s13018-014-0102-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shasha Zou
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Tingting Chen
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Yanan Wang
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Ruhui Tian
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Lingling Zhang
- BIO-X Center, Shanghai Jiao Tong University, 55 Guangyuan West Road, Shanghai, 200240, China.
| | - Pingping Song
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Shi Yang
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Yong Zhu
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Xizhi Guo
- BIO-X Center, Shanghai Jiao Tong University, 55 Guangyuan West Road, Shanghai, 200240, China.
| | - Yiran Huang
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Zheng Li
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| | - Lixin Kan
- Department of Pathophysiology, School of Basic Medicine, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China. .,Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL, 60611, USA.
| | - Hongliang Hu
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Lingshan Road, Shanghai, 200135, China.
| |
Collapse
|
15
|
Lucaciu OP, Soritau O, Baciut G, Lucaciu D, Baciut M, Campian R, Bran S. The Role of Bone Morphogenetic Proteins in Tissue Engineering Particulate Bone Grafts. PARTICULATE SCIENCE AND TECHNOLOGY 2014. [DOI: 10.1080/02726351.2013.879462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Ondine Patricia Lucaciu
- a Department of Oral Rehabilitation , University of Medicine and Pharmacy “Iuliu Hatieganu,” , Cluj Napoca , Romania
| | - Olga Soritau
- b Research Department , Oncological Institute “Prof. Ion Chiricuta,” , Cluj Napoca , Romania
| | - Grigore Baciut
- c Department of Cranio-Maxillofacial Surgery , University of Medicine and Pharmacy “Iuliu Hatieganu,” , Cluj Napoca , Romania
| | - Dan Lucaciu
- d Department of Orthopedic Surgery and Traumatology , University of Medicine and Pharmacy “Iuliu Hatieganu,” , Cluj Napoca , Romania
| | - Mihaela Baciut
- e Department of Surgery and Maxillofacial Implantology , University of Medicine and Pharmacy “Iuliu Hatieganu,” , Cluj Napoca , Romania
| | - Radu Campian
- a Department of Oral Rehabilitation , University of Medicine and Pharmacy “Iuliu Hatieganu,” , Cluj Napoca , Romania
| | - Simion Bran
- e Department of Surgery and Maxillofacial Implantology , University of Medicine and Pharmacy “Iuliu Hatieganu,” , Cluj Napoca , Romania
| |
Collapse
|
16
|
Lai MC, Chang KC, Hsu SC, Chou MC, Hung WI, Hsiao YR, Lee HM, Hsieh MF, Yeh JM. In situ gelation of PEG-PLGA-PEG hydrogels containing high loading of hydroxyapatite: in vitro and in vivo characteristics. Biomed Mater 2014; 9:015011. [PMID: 24457223 DOI: 10.1088/1748-6041/9/1/015011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Thermosensitive hydrogels are renowned carriers that are used to deliver a variety of drugs with the aim of combating diseases. In this study, the injectability of thermosensitive hydrogels comprised of poly(ethylene glycol)-poly(lactic acid-co-glycolic acid)-poly(ethylene glycol) (PEG-PLGA-PEG, PELGE) and hydroxyapatite (HA) were examined for their ability to deliver bone morphological protein 2 (BMP-2). The physicochemical characteristics of PELGE, HA, and PELGE/HA hydrogel composites were investigated by (1)H NMR, GPC, FTIR, XRD, SEM, and TEM. The rheological properties, injectability, in vitro degradation, and in vivo biocompatibility were investigated. The hydrogel with a weight ratio of 4:6 of polymer to HA was found to be resistant to auto-catalyzed degradation of acidic monomers (LA, GA) for a period of 70 days owing to the presence of alkaline HA. Injectability was quantitatively determined by the ejected weight of the hydrogel composite at room temperature and was a close match to the weight amount predetermined by the syringe pump. The results not only revealed that the PELGE/HA hydrogel composite presented a minor tissue response in the subcutis of ICR mice at eight weeks, but they also indicated an acceptable tolerance of the hydrogel composite in animals. Thus, PELGE/HA hydrogel composite is expected to be a promising injectable orthopedic substitute because of its desirable thermosensitivity and injectability.
Collapse
Affiliation(s)
- Mei-Chun Lai
- Department of Chemistry, Center for Nanotechnology and Center for Biomedical Technology at Chung-Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Park JK, Jang H, Hwang S, Kim EJ, Kim DE, Oh KB, Kwon DJ, Koh JT, Kimura K, Inoue H, Jang WG, Lee JW. ER stress-inducible ATF3 suppresses BMP2-induced ALP expression and activation in MC3T3-E1 cells. Biochem Biophys Res Commun 2013; 443:333-8. [PMID: 24315873 DOI: 10.1016/j.bbrc.2013.11.121] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 11/28/2013] [Indexed: 12/14/2022]
Abstract
Endoplasmic reticulum (ER) stress suppresses osteoblast differentiation. Activating transcription factor (ATF) 3, a member of the ATF/cAMP response element-binding protein family of transcription factors, is induced by various stimuli including cytokines, hormones, DNA damage, and ER stress. However, the role of ATF3 in osteoblast differentiation has not been elucidated. Treatment with tunicamycin (TM), an ER stress inducer, increased ATF3 expression in the preosteoblast cell line, MC3T3-E1. Overexpression of ATF3 inhibited bone morphogenetic protein 2-stimulated expression and activation of alkaline phosphatase (ALP), an osteogenic marker. In addition, suppression of ALP expression by TM treatment was rescued by silencing of ATF3 using shRNA. Taken together, these data indicate that ATF3 is a novel negative regulator of osteoblast differentiation by specifically suppressing ALP gene expression in preosteoblasts.
Collapse
Affiliation(s)
- Jae-kyung Park
- Research Center of Integrative Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea.
| | - Hoon Jang
- Research Center of Integrative Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Functional Genomics, School of Engineering, University of Science and Technology (UST), Daejeon 305-806, Republic of Korea.
| | - SeongSoo Hwang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, Republic of Korea.
| | - Eun-Jung Kim
- Research Center of Integrative Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea.
| | - Dong-Ern Kim
- Research Center of Integrative Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea.
| | - Keon-Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, Republic of Korea.
| | - Dae-Jin Kwon
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, Republic of Korea.
| | - Jeong-Tae Koh
- Dental Science Research Institute and BK21, School of Dentistry, Chonnam National University, Gwangju 500-757, Republic of Korea.
| | - Kumi Kimura
- Department of Physiology and Metabolism, Brain/Liver Interface Medicine Research Center, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641, Japan
| | - Hiroshi Inoue
- Department of Physiology and Metabolism, Brain/Liver Interface Medicine Research Center, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Won-Gu Jang
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk 712-714, Republic of Korea.
| | - Jeong-Woong Lee
- Research Center of Integrative Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Functional Genomics, School of Engineering, University of Science and Technology (UST), Daejeon 305-806, Republic of Korea.
| |
Collapse
|
18
|
Recent progresses in gene delivery-based bone tissue engineering. Biotechnol Adv 2013; 31:1695-706. [DOI: 10.1016/j.biotechadv.2013.08.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/24/2013] [Accepted: 08/19/2013] [Indexed: 12/18/2022]
|
19
|
Feichtinger GA, Hofmann AT, Slezak P, Schuetzenberger S, Kaipel M, Schwartz E, Neef A, Nomikou N, Nau T, van Griensven M, McHale AP, Redl H. Sonoporation increases therapeutic efficacy of inducible and constitutive BMP2/7 in vivo gene delivery. Hum Gene Ther Methods 2013; 25:57-71. [PMID: 24164605 DOI: 10.1089/hgtb.2013.113] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
An ideal novel treatment for bone defects should provide regeneration without autologous or allogenous grafting, exogenous cells, growth factors, or biomaterials while ensuring spatial and temporal control as well as safety. Therefore, a novel osteoinductive nonviral in vivo gene therapy approach using sonoporation was investigated in ectopic and orthotopic models. Constitutive or regulated, doxycycline-inducible, bone morphogenetic protein 2 and 7 coexpression plasmids were repeatedly applied for 5 days. Ectopic and orthotopic gene transfer efficacy was monitored by coapplication of a luciferase plasmid and bioluminescence imaging. Orthotopic plasmid DNA distribution was investigated using a novel plasmid-labeling method. Luciferase imaging demonstrated an increased trend (61% vs. 100%) of gene transfer efficacy, and micro-computed tomography evaluation showed significantly enhanced frequency of ectopic bone formation for sonoporation compared with passive gene delivery (46% vs. 100%) dependent on applied ultrasound power. Bone formation by the inducible system (83%) was stringently controlled by doxycycline in vivo, and no ectopic bone formation was observed without induction or with passive gene transfer without sonoporation. Orthotopic evaluation in a rat femur segmental defect model demonstrated an increased trend of gene transfer efficacy using sonoporation. Investigation of DNA distribution demonstrated extensive binding of plasmid DNA to bone tissue. Sonoporated animals displayed a potentially increased union rate (33%) without extensive callus formation or heterotopic ossification. We conclude that sonoporation of BMP2/7 coexpression plasmids is a feasible, minimally invasive method for osteoinduction and that improvement of bone regeneration by sonoporative gene delivery is superior to passive gene delivery.
Collapse
Affiliation(s)
- Georg A Feichtinger
- 1 Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, European Institute of Excellence on Tissue Engineering and Regenerative Medicine Research (Expertissues EEIG) , Vienna-Branch, 1200 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Su J, Xu H, Sun J, Gong X, Zhao H. Dual delivery of BMP-2 and bFGF from a new nano-composite scaffold, loaded with vascular stents for large-size mandibular defect regeneration. Int J Mol Sci 2013; 14:12714-28. [PMID: 23778088 PMCID: PMC3709809 DOI: 10.3390/ijms140612714] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/14/2013] [Accepted: 06/13/2013] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to investigate the feasibility and advantages of the dual delivery of bone morphogenetic protein-2 (BMP-2) and basic fibroblast growth factor (bFGF) from nano-composite scaffolds (PLGA/PCL/nHA) loaded with vascular stents (PLCL/Col/nHA) for large bone defect regeneration in rabbit mandibles. Thirty-six large bone defects were repaired in rabbits using engineering bone composed of allogeneic bone marrow mesenchymal stem cells (BMSCs), bFGF, BMP-2 and scaffolds composed of PLGA/PCL/nHA loaded with PLCL/Col/nHA. The experiments were divided into six groups: BMSCs/bFGF/BMP-2/scaffold, BMSCs/BMP-2/scaffold, BMSCs/bFGF/scaffold, BMSCs/scaffold, scaffold alone and no treatment. Sodium alginate hydrogel was used as the carrier for BMP-2 and bFGF and its features, including gelling, degradation and controlled release properties, was detected by the determination of gelation and degradation time coupled with a controlled release study of bovine serum albumin (BSA). AlamarBlue assay and alkaline phosphatase (ALP) activity were used to evaluate the proliferation and osteogenic differentiation of BMSCs in different groups. X-ray and histological examinations of the samples were performed after 4 and 12 weeks post-implantation to clarify new bone formation in the mandible defects. The results verified that the use of sodium alginate hydrogel as a controlled release carrier has good sustained release ability, and the combined application of bFGF and BMP-2 could significantly promote the proliferation and osteogenic differentiation of BMSCs (p < 0.05 or p < 0.01). In addition, X-ray and histological examinations of the samples exhibited that the dual release group had significantly higher bone formation than the other groups. The above results indicate that the delivery of both growth factors could enhance new bone formation and vascularization compared with delivery of BMP-2 or bFGF alone, and may supply a promising way of repairing large bone defects in bone tissue engineering.
Collapse
Affiliation(s)
- Jiansheng Su
- Institute of Prosthodontics, School of Stomatology, Tongji University, 399 Yanchang Road, Shanghai 200092, China.
| | | | | | | | | |
Collapse
|
21
|
Oyane A, Araki H, Sogo Y, Ito A, Tsurushima H. Spontaneous assembly of DNA–amorphous calcium phosphate nanocomposite spheres for surface-mediated gene transfer. CrystEngComm 2013. [DOI: 10.1039/c3ce40264a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
22
|
Yazaki Y, Oyane A, Araki H, Sogo Y, Ito A, Yamazaki A, Tsurushima H. Fabrication of DNA-antibody-apatite composite layers for cell-targeted gene transfer. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2012; 13:064204. [PMID: 27877531 PMCID: PMC5099764 DOI: 10.1088/1468-6996/13/6/064204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 09/20/2012] [Indexed: 06/04/2023]
Abstract
Surface-mediated gene transfer systems using apatite (Ap)-based composite layers have received increased attention in tissue engineering applications owing to their safety, biocompatibility and relatively high efficiency. In this study, DNA-antibody-apatite composite layers (DA-Ap layers), in which DNA and antibody molecules are immobilized within a matrix of apatite nanocrystals, were fabricated using a biomimetic coating process. They were then assayed for their gene transfer capability for application in a specific cell-targeted gene transfer. A DA-Ap layer that was fabricated with an anti-CD49f antibody showed a higher gene transfer capability to the CD49f-positive CHO-K1 cells than a DNA-apatite composite layer (D-Ap layer). The antibody facilitated the gene transfer capability of the DA-Ap layer only to the specific cells that were expressing corresponding antigens. When the DA-Ap layer was fabricated with an anti-N-cadherin antibody, a higher gene transfer capability compared with the D-Ap layer was found in the N-cadherin-positive P19CL6 cells, but not in the N-cadherin-negative UV♀2 cells or in the P19CL6 cells that were pre-blocked with anti-N-cadherin. Therefore, the antigen-antibody binding that takes place at the cell-layer interface should be responsible for the higher gene transfer capability of the DA-Ap than D-Ap layer. These results suggest that the DA-Ap layer works as a mediator in a specific cell-targeted gene transfer system.
Collapse
Affiliation(s)
- Yushin Yazaki
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku, Tokyo, 169-8555, Japan
- Nanosystem Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8562, Japan
| | - Ayako Oyane
- Nanosystem Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8562, Japan
| | - Hiroko Araki
- Nanosystem Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8562, Japan
| | - Yu Sogo
- Human Technology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Atsuo Ito
- Human Technology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Atsushi Yamazaki
- Department of Resources and Environmental Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku, Tokyo, 169-8555, Japan
| | - Hideo Tsurushima
- Nanosystem Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8562, Japan
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
23
|
Abstract
A surface-mediated gene transfer system using DNA-calcium phosphate (CaP) composite layers (D-CaP layers) would be useful in tissue engineeing. In previous studies, D-CaP layers were fabricated in supersaturated CaP solutions prepared using chemical reagents. In this study, a so-called RKM solution prepared using clinically approved infusion fluids was employed as a supersaturated CaP solution. A D-CaP layer consisting of submicron spherical particles was successfully fabricated on a polystyrene substrate by immersing the substrate in the RKM solution for 24 h. When the immersion period was prolonged from 24 to 72 h, amount of CaP and DNA on the substrate increased. However, the gene transfer capability of the D-CaP layer for the CHO-K1 cells was kept unchanged irrespective of the immersion period. In the RKM solution process, immersion period of 24 h was found to be long enough for gene transfer application of the D-CaP layer. More importantly, the D-CaP layer fabricated by the RKM solution process exhibited a significantly higher gene transfer capability than our previous D-CaP layer fabricated in the conventional CaP solution with the same DNA concentration. The RKM solution process for the fabrication of D-CaP layers was found to be advantageous to the previous process in terms of not only safety but the layers gene transfer capability.
Collapse
|
24
|
Calcium phosphate composite layers for surface-mediated gene transfer. Acta Biomater 2012; 8:2034-46. [PMID: 22343517 DOI: 10.1016/j.actbio.2012.02.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/27/2012] [Accepted: 02/01/2012] [Indexed: 01/17/2023]
Abstract
In this review, the surface-mediated gene transfer system using calcium phosphate composite layers is described. Calcium phosphate ceramics are osteoconductive bioceramics used typically in orthopedic and dental applications. Additionally, calcium phosphate particles precipitated by a liquid-phase process have long been used as a safe and biocompatible transfection reagent in molecular biology. Recently, calcium phosphate composite layers immobilizing DNA were fabricated on the surfaces of base materials through a biomimetic process using supersaturated solutions. These composite layers possess useful characteristics of both osteoconductive bioceramics and transfection reagents; they thus provide a biocompatible surface to support cell adhesion and growth, and can stimulate the cell effectively via surface-mediated gene transfer. By modifying the fabrication conditions, physicochemical and biological properties of the composite layers can be varied. With such an approach, these composite layers can be designed to have improved affinity for cells and to exhibit increased gene transfer efficiency over that of conventional lipid transfection reagents. The composite layers with the increased gene transfer efficiency induced specific cell differentiation and tissue regeneration in vivo. These composite layers, given their good biocompatibility and the potential to control cell behavior on their surfaces, have great potential in tissue engineering applications.
Collapse
|
25
|
Oyane A, Yazaki Y, Araki H, Sogo Y, Ito A, Yamazaki A, Tsurushima H. Fabrication of a DNA-lipid-apatite composite layer for efficient and area-specific gene transfer. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:1011-1019. [PMID: 22367107 DOI: 10.1007/s10856-012-4581-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 02/04/2012] [Indexed: 05/31/2023]
Abstract
A surface-mediated gene transfer system using biocompatible apatite-based composite layers has great potential for tissue engineering. Among the apatite-based composite layers developed to date, we focused on a DNA-lipid-apatite composite layer (DLp-Ap layer), which has the advantage of relatively high efficiency as a non-viral system. In this study, various lipid transfection reagents, including a newly developed reagent, polyamidoamine dendron-bearing lipid (PD), were employed to prepare the DLp-Ap layer, and the preparation condition was optimized in terms of efficiency of gene transfer to epithelial-like CHO-K1 cells in the presence of serum. The optimized DLp-Ap layer derived from PD had the highest gene transfer efficiency among all the apatite-based composite layers prepared in this study. In addition, the optimized DLp-Ap layer demonstrated higher gene transfer efficiency in the presence of serum than the conventional particle-mediated systems using commercially available lipid transfection reagents. It was also shown that the optimized DLp-Ap layer mediated the area-specific gene transfer on its surface, i.e., DNA was preferentially transferred to the cells adhering to the surface of the layer. The present gene transfer system using the PD-derived DLp-Ap layer, with the advantages of high efficiency in the presence of serum and area-specificity, would be useful in tissue engineering.
Collapse
Affiliation(s)
- Ayako Oyane
- Nanosystem Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan.
| | | | | | | | | | | | | |
Collapse
|