1
|
Song Y, Li H, Wang Z, Shi J, Li J, Wang L, Liao L, Ma S, Zhang Y, Liu B, Yang Y, Zhou P. Define of Optimal Addition Period of Osteogenic Peptide to Accelerate the Osteogenic Differentiation of Human Pluripotent Stem Cells. Tissue Eng Regen Med 2024; 21:291-308. [PMID: 37903982 PMCID: PMC10825087 DOI: 10.1007/s13770-023-00597-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/15/2023] [Accepted: 09/05/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The addition of growth factiors is commonly applied to improve the osteogenic differentiation of stem cells. However, for human pluripotent stem cells (hPSCs), their complex differentiation processes result in the unknown effect at different stages. In this study, we focused on the widely used bone forming peptide-1 (BFP-1) and investigated the effect and mechanisms of its addition on the osteogenic induction of hPSCs as a function of the supplementation period. METHODS Monolayer-cultured hPSCs were cultured in osteogenic induction medium for 28 days, and the effect of BFP-1 peptide addition at varying weeks was examined. After differentiation for varying days (0, 7, 14, 21 and 28), the differentiation efficiency was determined by RT-PCR, flow cytometry, immunofluorescence, and alizarin red staining assays. Moreover, the expression of marker genes related to germ layers and epithelial-mesenchymal transition (EMT) was investigated at day 7. RESULTS Peptide treatment during the first week promoted the generation of mesoderm cells and mesenchymal-like cells from hiPSCs. Then, the upregulated expression of osteogenesis marker genes/proteins was detected in both hESCs and hiPSCs during subsequent inductions with BFP-1 peptide treatment. Fortunately, further experimental design confirmed that treating the BFP-1 peptide during 7-21 days showed even better performance for hESCs but was ineffective for hiPSCs. CONCLUSION The differentiation efficiency of cells could be improved by determining the optimal treatment period. Our study has great value in maximizing the differentiation of hPSCs by adding osteogenesis peptides based on the revealed mechanisms and promoting the application of hPSCs in bone tissue regeneration.
Collapse
Affiliation(s)
- Yameng Song
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Hongjiao Li
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Zixuan Wang
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jiamin Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jing Li
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Lu Wang
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Lingzi Liao
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Shengqin Ma
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Yun Zhang
- Lanzhou Hospital of Stomatology, Lanzhou, 730000, People's Republic of China
| | - Bin Liu
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China.
| | - Yaling Yang
- Lanzhou Hospital of Stomatology, Lanzhou, 730000, People's Republic of China.
| | - Ping Zhou
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, People's Republic of China.
| |
Collapse
|
2
|
Tilkin RG, Mahy JG, Monteiro AP, Belet A, Feijóo J, Laird M, Carcel C, Régibeau N, Goderis B, Grandfils C, Wong Chi Man M, Lambert SD. Protein encapsulation in mesoporous silica: Influence of the mesostructured and pore wall properties. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.128629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
3
|
Xu Y, Yang Y, Hua Z, Li S, Yang Z, Liu Q, Fu G, Ji P, Wu Q. BMP2 immune complexes promote new bone formation by facilitating the direct contact between osteoclasts and osteoblasts. Biomaterials 2021; 275:120890. [PMID: 34130144 DOI: 10.1016/j.biomaterials.2021.120890] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/22/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022]
Abstract
BMP2 antibody is proposed as a promising replacement for rhBMP2 in bone tissue engineering. Although studies have demonstrated its osteoinductive efficacy, the underlying osteogenic mechanism and adverse reactions of specific BMP2 antibody are not clarified yet, making it difficult to optimize the antibody for future application. By establishing BMP2 immune complexes (BMP2-ICs) ex vivo, we were able to introduce BMP2-ICs directly in vivo and found that BMP2-ICs promoted bone formation while suppressing osteoclastogenesis. However, ex vivo osteoclastogenic assays showed that BMP2-ICs promoted osteoclastogenesis by binding FcγR and activating PLCγ2 phosphorylation. Given that BMP2-ICs react with osteoblast and osteoclast lineage cells by the conjugated BMP2 domain and the Fc domain respectively, we introduced BMP2-ICs into coculture system of the two lineage cells and found that BMP2-ICs promoted osteogenesis while suppressing osteoclastogenesis by facilitating osteoblast-osteoclast contact and activating the EphrinB2-EphB4 signaling. This bidirectional function of BMP2-ICs was reproduced in the cranial bone resorption model, where osteoblast and osteoclast lineage cells co-localized. This study excluded the hidden problem of osteoclast overactivation that usually comes with rhBMP2 and clarified the first evidence of the mechanism of antibody-mediated bone regeneration, suggesting BMP2-ICs may present a promising therapy for bone diseases related with disrupted osteoclast-osteoblast interaction.
Collapse
Affiliation(s)
- Yamei Xu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Yao Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Ziyi Hua
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Shuang Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Zhenyu Yang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Qianzi Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Gang Fu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China; Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Qingqing Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China; Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China.
| |
Collapse
|
4
|
Wilkinson P, Bozo IY, Braxton T, Just P, Jones E, Deev RV, Giannoudis PV, Feichtinger GA. Systematic Review of the Preclinical Technology Readiness of Orthopedic Gene Therapy and Outlook for Clinical Translation. Front Bioeng Biotechnol 2021; 9:626315. [PMID: 33816447 PMCID: PMC8011540 DOI: 10.3389/fbioe.2021.626315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/12/2021] [Indexed: 12/09/2022] Open
Abstract
Bone defects and improper healing of fractures are an increasing public health burden, and there is an unmet clinical need in their successful repair. Gene therapy has been proposed as a possible approach to improve or augment bone healing with the potential to provide true functional regeneration. While large numbers of studies have been performed in vitro or in vivo in small animal models that support the use of gene therapy for bone repair, these systems do not recapitulate several key features of a critical or complex fracture environment. Larger animal models are therefore a key step on the path to clinical translation of the technology. Herein, the current state of orthopedic gene therapy research in preclinical large animal models was investigated based on performed large animal studies. A summary and an outlook regarding current clinical studies in this sector are provided. It was found that the results found in the current research literature were generally positive but highly methodologically inconsistent, rendering a comparison difficult. Additionally, factors vital for translation have not been thoroughly addressed in these model systems, and the risk of bias was high in all reviewed publications. These limitations directly impact clinical translation of gene therapeutic approaches due to lack of comparability, inability to demonstrate non-inferiority or equivalence compared with current clinical standards, and lack of safety data. This review therefore aims to provide a current overview of ongoing preclinical and clinical work, potential bottlenecks in preclinical studies and for translation, and recommendations to overcome these to enable future deployment of this promising technology to the clinical setting.
Collapse
Affiliation(s)
- Piers Wilkinson
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom.,CDT Tissue Engineering and Regenerative Medicine, Institute of Medical and Biological Engineering, University of Leeds, Leeds, United Kingdom
| | - Ilya Y Bozo
- Federal Medical Biophysical Center, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Thomas Braxton
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom.,CDT Tissue Engineering and Regenerative Medicine, Institute of Medical and Biological Engineering, University of Leeds, Leeds, United Kingdom
| | - Peter Just
- Into Numbers Data Science GmbH, Vienna, Austria
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | | | - Peter V Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds General Infirmary, Leeds, United Kingdom.,NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, United Kingdom
| | - Georg A Feichtinger
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
5
|
Cardiopulmonary and Neurologic Dysfunctions in Fibrodysplasia Ossificans Progressiva. Biomedicines 2021; 9:biomedicines9020155. [PMID: 33562570 PMCID: PMC7915901 DOI: 10.3390/biomedicines9020155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 12/28/2022] Open
Abstract
Fibrodysplasia Ossificans Progressiva (FOP) is an ultra-rare but debilitating disorder characterized by spontaneous, progressive, and irreversible heterotopic ossifications (HO) at extraskeletal sites. FOP is caused by gain-of-function mutations in the Activin receptor Ia/Activin-like kinase 2 gene (Acvr1/Alk2), with increased receptor sensitivity to bone morphogenetic proteins (BMPs) and a neoceptor response to Activin A. There is extensive literature on the skeletal phenotypes in FOP, but a much more limited understanding of non-skeletal manifestations of this disease. Emerging evidence reveals important cardiopulmonary and neurologic dysfunctions in FOP including thoracic insufficiency syndrome, pulmonary hypertension, conduction abnormalities, neuropathic pain, and demyelination of the central nervous system (CNS). Here, we review the recent research and discuss unanswered questions regarding the cardiopulmonary and neurologic phenotypes in FOP.
Collapse
|
6
|
Li A, Li J, Zhang Z, Li Z, Chi H, Song C, Wang X, Wang Y, Chen G, Yan J. Nanohydroxyapatite/polyamide 66 crosslinked with QK and BMP-2-derived peptide prevented femur nonunion in rats. J Mater Chem B 2021; 9:2249-2265. [PMID: 33599673 DOI: 10.1039/d0tb02554b] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A dual-peptide controlled released system based on nHA/PA66 scaffold for enhancing bone regeneration.
Collapse
|
7
|
The sialylation profile of IgG determines the efficiency of antibody directed osteogenic differentiation of iMSCs by modulating local immune responses and osteoclastogenesis. Acta Biomater 2020; 114:221-232. [PMID: 32771590 DOI: 10.1016/j.actbio.2020.07.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/03/2020] [Accepted: 07/31/2020] [Indexed: 12/25/2022]
Abstract
Antibody-mediated osseous regeneration (AMOR) has been proved as a promising strategy for osteogenic differentiation of induced pluripotent stem cells derived MSCs (iMSCs). The key characteristic of antibody that determines the AMOR potential is largely unknown. The glycosylation profile of immunoglobulin G (IgG) represents a key checkpoint that determines its effector functions. Herein, we modified the sialylation profile of BMP2 antibodies to investigate the effects of glycosylation on antibody-mediated osteogenic differentiation of iMSCs. We found that over-sialylated BMP2 antibodies stimulated the highest amount of new bone while those non- or low-sialylated led to bone porosity and collapse. The immune response aroused by BMP2 immune complexes (BMP2-ICs) was intensified by desialylation, which contributed to an environment that favored osteoclastogenesis while inhibited osteoblastogenesis. In vitro study further demonstrated that the osteogenic potential of BMP2-ICs was not significantly affected by the degree of sialylation. On the other hand, BMP2-ICs could stimulate osteoclastogenesis by binding FcγRs on preosteoclasts directly, which was significantly intensified by desialylation and attenuated by over-sialylation. Bone defects implanted with alginate microbeads loaded with iMSCs and over-sialylated antibodies showed more bone formation than those sites with non- or low sialylated antibodies. Taken together, our study demonstrated that sialylation profile is one of the traits that decide the AMOR potential of BMP2 antibodies. Enhancement of sialylation may be a promising strategy to optimize antibody for iMSCs application in bone tissue engineering. STATEMENT OF SIGNIFICANCE: Antibody-mediated osseous regeneration (AMOR) is a promising strategy for bone tissue engineering that takes advantage of the specific reactivity of antibodies to sequester endogenous BMP2 and present it to osteoprogenitor cells. We previously demonstrated that BMP2 immune complex can drive iPSCs derived MSCs to osteogenic lineage. In this study, we analyze the effects of glycosylation profile on antibody directed osteogenic differentiation of iMSCs because glycosylation profile represents a key checkpoint that determines the effector functions of antibodies, and it is susceptible to variations in different clones. The results showed that sialylation profile is one of the traits that decides the AMOR potential of BMP2 antibody, and the enhancement of sialylation maybe a promising strategy to optimize antibodies for AMOR.
Collapse
|
8
|
Karyagina AS, Grunina TM, Lyaschuk AM, Voronina EV, Marigin RA, Cherepushkin SA, Trusova IN, Grishin AV, Poponova MS, Orlova PA, Manskikh VN, Strukova NV, Generalova MS, Nikitin KE, Soboleva LA, Boksha IS, Gromov AV. Recombinant Human Erythropoietin Proteins Synthesized in Escherichia coli Cells: Effects of Additional Domains on the in vitro and in vivo Activities. BIOCHEMISTRY (MOSCOW) 2019; 84:20-32. [PMID: 30927522 DOI: 10.1134/s0006297919010036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aim of this work was to compare biological activities of three variants of bacterially expressed human recombinant erythropoietin (EPO) with additional protein domains: 6His-s-tag-EPO protein carrying the s-tag (15-a.a. oligopeptide from bovine pancreatic ribonuclease A) at the N-terminus and HBD-EPO and EPO-HBD proteins containing heparin-binding protein domains (HBD) of the bone morphogenetic protein 2 from Danio rerio at the N- and C-termini, respectively. The commercial preparation Epostim (LLC Pharmapark, Russia) produced by synthesis in Chinese hamster ovary cells was used for comparison. The EPO variant with the C-terminal HBD domain connected by a rigid linker (EPO-HBD) possesses the best properties as compared to HBD-EPO with the reverse domain arrangement. It was ~13 times more active in vitro (i.e., promoted proliferation of human erythroleukemia TF-1 cells) and demonstrated a higher rate of association with the erythropoietin receptor. EPO-HBD also exhibited the greatest binding to the demineralized bone matrix (DBM) and more prolonged release from the DBM among the four proteins studied. Subcutaneous administration of EPO-HBD immobilized on DBM resulted in significantly more pronounced vascularization of surrounding tissues in comparison with the other proteins and DBM alone. Therefore, EPO-HBD displayed better performance with regard to all the investigated parameters than other examined EPO variants, and it seems promising to study the possibility of its medical use.
Collapse
Affiliation(s)
- A S Karyagina
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia. .,All-Russia Research Institute of Agricultural Biotechnology, Moscow, 127550, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - T M Grunina
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - A M Lyaschuk
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - E V Voronina
- State Research Institute of Genetics and Selection of Industrial Microorganisms, Kurchatov Institute National Research Center, Moscow, 117545, Russia
| | - R A Marigin
- State Research Institute of Genetics and Selection of Industrial Microorganisms, Kurchatov Institute National Research Center, Moscow, 117545, Russia
| | - S A Cherepushkin
- State Research Institute of Genetics and Selection of Industrial Microorganisms, Kurchatov Institute National Research Center, Moscow, 117545, Russia
| | - I N Trusova
- State Research Institute of Genetics and Selection of Industrial Microorganisms, Kurchatov Institute National Research Center, Moscow, 117545, Russia
| | - A V Grishin
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia.,All-Russia Research Institute of Agricultural Biotechnology, Moscow, 127550, Russia
| | - M S Poponova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - P A Orlova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - V N Manskikh
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - N V Strukova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - M S Generalova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - K E Nikitin
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - L A Soboleva
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - I S Boksha
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia.,Research Center of Mental Health, Moscow, 115522, Russia
| | - A V Gromov
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia.
| |
Collapse
|
9
|
Li D, Zhang K, Shi C, Liu L, Yan G, Liu C, Zhou Y, Hu Y, Sun H, Yang B. Small molecules modified biomimetic gelatin/hydroxyapatite nanofibers constructing an ideal osteogenic microenvironment with significantly enhanced cranial bone formation. Int J Nanomedicine 2018; 13:7167-7181. [PMID: 30464466 PMCID: PMC6228053 DOI: 10.2147/ijn.s174553] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Repair of nonunion critical-sized bone defects is a significant clinical challenge all over the world. Construction of osteogenic microenvironment that provides osteoconductive and osteoinductive signals is a leading strategy. Materials and methods In the present study, ascorbic acid (AA) and β-glycerophosphate disodium salt hydrate (β-GP) modified biomimetic gelatin/hydroxyapatite (GH) nanofibrous scaffolds were developed by electrospinning. Then the scaffolds were crosslinked by N-hydroxysulfo-succinimide sodium salt (NHS) and 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC). The morphology of the non-crosslinked and crosslinked scaffolds was evaluated by scanning electron microscope (SEM). Fourier transform infrared spectroscopy (FT-IR) was used to assess the interacting model between the small molecules and GH scaffold. Then MTT, Alamar Blue, and CCK8 assays were used to investigate the biocompatibility of the various crosslinked scaffolds. Subsequently, the osteogenic genes expression of bone marrow stromal cells (BMSCs) cultured on the scaffolds were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Finally, the crosslinked scaffolds were implanted in a rat calvarial defect model to assess the osteogenic effects in vivo. Results SEM results showed that the various scaffolds presented extracellular matrix (ECM)-like fibrous porous structure. (FT-IR) spectrum indicated that AA and β-GP were covalently bonded with GH scaffolds. The MTT, Alamar Blue, and CCK8 assays demonstrated that all the scaffolds can support BMSCs' growth well. The qRT-PCR results showed that the expression level of Alp and Runx2 in BMSCs on GH/A/B scaffold was about 3.5- and 1.5-fold, respectively, compared with that of GH group on day 7. The results also showed that AA- and β-GP-modified GH scaffolds can significantly induce the higher levels of osteogenic gene expression in a temporal specific manner. Importantly, AA and β-GP synergistically promoted osteoblast differentiation in vitro and dramatically induced bone regeneration in vivo. Impressively, AA and β-GP dual modified GH nanofibrous scaffold could serve as a template for guiding bone regeneration and the bone defects were almost repaired completely (94.28%±5.00%) at 6 weeks. Besides, single AA or β-GP-modified GH nanofibrous scaffolds could repair 62.95%±9.39% and 66.56%±18.45% bone defects, respectively, at 12 weeks in vivo. In addition, AA and β-GP exhibit an anti-inflammatory effect in vivo. Conclusion Our data highlighted that, AA, β-GP, and GH nanofibers created a fine osteoconductive and osteoinductive microenvironments for bone regeneration. We demonstrated that AA and β-GP dual modified GH nanofiber is a versatile bone tissue engineering scaffold.
Collapse
Affiliation(s)
- Daowei Li
- Department of Oral Biology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, People's Republic of China.,Department of Oral Pathology, Liaoning Province Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, People's Republic of China
| | - Kai Zhang
- Department of Oral Pathology, Liaoning Province Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, People's Republic of China
| | - Ce Shi
- Department of Oral Biology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, People's Republic of China.,Department of Oral Pathology, Liaoning Province Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, People's Republic of China
| | - Lijun Liu
- Department of Oral Biology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Guangxing Yan
- Department of Oral Biology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Cangwei Liu
- Department of Oral Biology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Yijun Zhou
- Department of Oral Biology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Yue Hu
- Department of Oral Biology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Hongchen Sun
- Department of Multiscale Diagnosis and Treatment Chemistry, State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, People's Republic of China,
| | - Bai Yang
- Department of Oral Pathology, Liaoning Province Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
10
|
Manavitehrani I, Fathi A, Schindeler A, Dehghani F. Sustained Protein Release from a Core-Shell Drug Carrier System Comprised of Mesoporous Nanoparticles and an Injectable Hydrogel. Macromol Biosci 2018; 18:e1800201. [PMID: 30395416 DOI: 10.1002/mabi.201800201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/23/2018] [Indexed: 11/11/2022]
Abstract
The manufacture of a biocompatible carrier for controlled delivery of bioactive compounds is described. This carrier is composed of a mesoporous silica nanoparticle as core that is homogenously distributed in an injectable hydrogel. For the synthesis of nanoparticles, a one step sol-gel method is developed to produce pores with the range of 100 nm. BMP2 and Fluorescein-conjugated bovine serum albumin is used as proteinaceous agents for measuring release, and is loaded into mesoporous silica nanoparticles at the optimum conditions of 48 h incubation period using 1:10 ratio of protein to nanoparticles. The release of proteins from either mesoporous nanoparticles or hydrogel individually involves a burst release stage, however the release from the core/shell carrier designed in this study follows a zero order kinetic. In summary, this biomaterial may be favorable for delivery of bioactive compounds such as BMP2 for a range of applications including bone tissue regeneration.
Collapse
Affiliation(s)
- Iman Manavitehrani
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, 2006, Australia
| | - Ali Fathi
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, 2006, Australia
| | - Aaron Schindeler
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, 2006, Australia.,Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, 2145, Australia
| | - Fariba Dehghani
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, 2006, Australia
| |
Collapse
|
11
|
Grgurevic L, Oppermann H, Pecin M, Erjavec I, Capak H, Pauk M, Karlovic S, Kufner V, Lipar M, Bubic Spoljar J, Bordukalo-Niksic T, Maticic D, Peric M, Windhager R, Sampath TK, Vukicevic S. Recombinant Human Bone Morphogenetic Protein 6 Delivered Within Autologous Blood Coagulum Restores Critical Size Segmental Defects of Ulna in Rabbits. JBMR Plus 2018; 3:e10085. [PMID: 31131338 DOI: 10.1002/jbm4.10085] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/29/2018] [Accepted: 12/16/2018] [Indexed: 12/21/2022] Open
Abstract
BMP2 and BMP7, which use bovine Achilles tendon-derived absorbable collagen sponge and bovine bone collagen as scaffold, respectively, have been approved as bone graft substitutes for orthopedic and dental indications. Here, we describe an osteoinductive autologous bone graft substitute (ABGS) that contains recombinant human BMP6 (rhBMP6) dispersed within autologous blood coagulum (ABC) scaffold. The ABGS is created as an injectable or implantable coagulum gel with rhBMP6 binding tightly to plasma proteins within fibrin meshwork, as examined by dot-blot assays, and is released slowly as an intact protein over 6 to 8 days, as assessed by ELISA. The biological activity of ABGS was examined in vivo in rats (Rattus norvegicus) and rabbits (Oryctolagus cuniculus). In a rat subcutaneous implant assay, ABGS induced endochondral bone formation, as observed by histology and micro-CT analyses. In the rabbit ulna segmental defect model, a reproducible and robust bone formation with complete bridging and restoration of the defect was observed, which is dose dependent, as determined by radiographs, micro-CT, and histological analyses. In ABGS, ABC scaffold provides a permissive environment for bone induction and contributes to the use of lower doses of rhBMP6 compared with BMP7 in bovine bone collagen as scaffold. The newly formed bone undergoes remodeling and establishes cortices uniformly that is restricted to implant site by bridging with host bone. In summary, ABC carrier containing rhBMP6 may serve as an osteoinductive autologous bone graft substitute for several orthopedic applications that include delayed and nonunion fractures, anterior and posterior lumbar interbody fusion, trauma, and nonunions associated with neurofibromatosis type I.
Collapse
Affiliation(s)
- Lovorka Grgurevic
- Laboratory for Mineralized Tissues School of Medicine University of Zagreb Zagreb Croatia
| | | | - Marko Pecin
- Clinics for Surgery, Orthopedics, and Ophthalmology School of Veterinary Medicine University of Zagreb Zagreb Croatia
| | - Igor Erjavec
- Laboratory for Mineralized Tissues School of Medicine University of Zagreb Zagreb Croatia
| | - Hrvoje Capak
- Department of Radiology School of Veterinary Medicine University of Zagreb Zagreb Croatia
| | - Martina Pauk
- Laboratory for Mineralized Tissues School of Medicine University of Zagreb Zagreb Croatia
| | - Sven Karlovic
- Faculty of Food Technology and Biotechnology University of Zagreb Zagreb Croatia
| | - Vera Kufner
- Laboratory for Mineralized Tissues School of Medicine University of Zagreb Zagreb Croatia
| | - Marija Lipar
- Clinics for Surgery, Orthopedics, and Ophthalmology School of Veterinary Medicine University of Zagreb Zagreb Croatia
| | - Jadranka Bubic Spoljar
- Laboratory for Mineralized Tissues School of Medicine University of Zagreb Zagreb Croatia
| | | | - Drazen Maticic
- Clinics for Surgery, Orthopedics, and Ophthalmology School of Veterinary Medicine University of Zagreb Zagreb Croatia
| | - Mihaela Peric
- Laboratory for Mineralized Tissues School of Medicine University of Zagreb Zagreb Croatia
| | - Reinhard Windhager
- Department of Orthopedics and Trauma Surgery Medical University of Vienna Vienna Austria
| | | | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues School of Medicine University of Zagreb Zagreb Croatia
| |
Collapse
|
12
|
Wu Q, Yang B, Cao C, Hu K, Wang P, Man Y. Therapeutic antibody directed osteogenic differentiation of induced pluripotent stem cell derived MSCs. Acta Biomater 2018; 74:222-235. [PMID: 29778895 DOI: 10.1016/j.actbio.2018.05.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 12/31/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are regarded as a new cell source for regenerative medicine. Recent advances in tissue engineering have brought to light the therapeutic application of induced pluripotent stem cells (iPSCs) in bone defect repair. However, a safe and efficient way to differentiate iPSCs into osteogenic lineage remains to be a major challenge. Here we describe an approach using anti-BMP2 antibodies (Abs) to mediate osteogenic differentiation of iPSC-derived mesenchymal stromal cells (iMSCs). We first proved that 3G7 (an anti-BMP2 Ab) not only bound to BMP2, but also allowed the bound BMP2 to engage the BMP2 receptors on iMSCs. Subcutaneous implantation sites loaded with iMSCs + 3G7 group showed significant bone formation and vascularization in mice while those sites with exogenous BMP2 exhibited dystrophic calcification and significantly lower vascularization. Our in vitro study demonstrated that the anti-BMP2 Ab/BMP2 immune complex were capable of dictating the acquisition of osteogenic phenotype of iMSCs and subsequent mineralization. The study provided the first evidence of antibody-mediated differentiation of iMSCs and osseous regeneration in vivo. This novel strategy takes full advantage of the endogenous bioactive molecules for osseous regeneration and its potential therapeutic application is promising. STATEMENT OF SIGNIFICANCE Induced pluripotent stem cells (iPSCs) and its derived cells hold significant promise for the treatment of bone defects. In present study, we carried out the concept of antibody-mediated bone regeneration into the iPSC research for the first time. We demonstrated that anti-BMP2 Ab/BMP2 immune complex was capable of promoting osteogenic differentiation of iPSC-derived MSCs (iMSCs), likely through the classical BMP2/Smad1/Runx2 pathway. Subcutaneous co-delivery of iMSCs and anti-BMP2 Abs resulted in significant bone formation and vascularization. These findings suggested antibody mediated osteogenic differentiation may be a favorable approach for iPSC-based bone tissue engineering.
Collapse
|
13
|
Abstract
BACKGROUND Nonhealing bone defects represent an immense biomedical burden. Despite recent advances in protein-based bone regeneration, safety concerns over bone morphogenetic protein-2 have prompted the search for alternative factors. Previously, the authors examined the additive/synergistic effects of hedgehog and Nel-like protein-1 (NELL-1) on the osteogenic differentiation of mesenchymal stem cells in vitro. In this study, the authors sought to leverage their previous findings by applying the combination of Smoothened agonist (SAG), hedgehog signal activator, and NELL-1 to an in vivo critical-size bone defect model. METHODS A 4-mm parietal bone defect was created in mixed-gender CD-1 mice. Treatment groups included control (n = 6), SAG (n = 7), NELL-1 (n = 7), and SAG plus NELL-1 (n = 7). A custom fabricated poly(lactic-co-glycolic acid) disk with hydroxyapatite coating was used as an osteoinductive scaffold. RESULTS Results at 4 and 8 weeks showed increased bone formation by micro-computed tomographic analyses with either stimulus alone (SAG or NELL-1), but significantly greater bone formation with both components combined (SAG plus NELL-1). This included greater bone healing scores and increased bone volume and bone thickness. Histologic analyses confirmed a significant increase in new bone formation with the combination therapy SAG plus NELL-1, accompanied by increased defect vascularization. CONCLUSIONS In summary, the authors' results suggest that combining the hedgehog signaling agonist SAG and NELL-1 has potential as a novel therapeutic strategy for the healing of critical-size bone defects. Future directions will include optimization of dosage and delivery strategy for an SAG and NELL-1 combination product.
Collapse
|
14
|
|
15
|
Barcak EA, Beebe MJ. Bone Morphogenetic Protein: Is There Still a Role in Orthopedic Trauma in 2017? Orthop Clin North Am 2017; 48:301-309. [PMID: 28577779 DOI: 10.1016/j.ocl.2017.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Approximately 10 years ago bone morphogenic protein (BMP) was seen as a miraculous adjuvant to assist with bone growth. However, in the face of an increasing number of complications and a lack of understanding its long-term effects, it is unclear what role BMP has in the current treatment of orthopedic trauma patients. This article reviews the current recommendations, trends, and associated complications of BMP use in fracture care.
Collapse
Affiliation(s)
- Eric A Barcak
- Department of Orthopedic Surgery, University of Tennessee-Campbell Clinic, 1211 Union Avenue #500, Memphis, TN 38104, USA.
| | - Michael J Beebe
- Department of Orthopedic Surgery, University of Tennessee-Campbell Clinic, 1211 Union Avenue #500, Memphis, TN 38104, USA
| |
Collapse
|
16
|
Karyagina AS, Boksha IS, Grunina TM, Demidenko AV, Poponova MS, Sergienko OV, Lyashchuk AM, Galushkina ZM, Soboleva LA, Osidak EO, Bartov MS, Gromov AV, Lunin VG. Two Variants of Recombinant Human Bone Morphogenetic Protein-2 (rhBMP-2) with Additional Protein Domains: Synthesis in an Escherichia coli Heterologous Expression System. BIOCHEMISTRY (MOSCOW) 2017; 82:613-624. [PMID: 28601071 DOI: 10.1134/s0006297917050091] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Two variants of recombinant human bone morphogenetic protein-2 (rhBMP-2) with additional N-terminal protein domains were obtained by expression in E. coli. The N-terminal domains were s-tag (15-a.a. oligopeptide from bovine pancreatic ribonuclease A) and lz (leucine zipper dimerization domain from yeast transcription factor GCN4). The s-tag-BMP-2 and lz-BMP-2 were purified by a procedure that excluded a long refolding stage. The resulting dimeric proteins displayed higher solubility compared to rhBMP-2 without additional protein domains. Biological activity of both proteins was demonstrated in vitro by induction of alkaline phosphatase in C2C12 cells, and the activity of s-tag-BMP-2 in vivo was shown in various experimental animal models.
Collapse
Affiliation(s)
- A S Karyagina
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Formulation, Delivery and Stability of Bone Morphogenetic Proteins for Effective Bone Regeneration. Pharm Res 2017; 34:1152-1170. [PMID: 28342056 PMCID: PMC5418324 DOI: 10.1007/s11095-017-2147-x] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/17/2017] [Indexed: 12/22/2022]
Abstract
Bone morphogenetic proteins (BMPs) are responsible for bone formation during embryogenesis and bone regeneration and remodeling. The osteoinductive action of BMPs, especially BMP-2 and BMP-7, has led to their use in a range of insurmountable treatments where intervention is required for effective bone regeneration. Introduction of BMP products to the market, however, was not without reports of multiple complications and side effects. Aiming for optimization of the therapeutic efficacy and safety, efforts have been focused on improving the delivery of BMPs to lower the administered dose, localize the protein, and prolong its retention time at the site of action. A major challenge with these efforts is that the protein stability should be maintained. With this review we attempt to shed light on how the stability of BMPs can be affected in the formulation and delivery processes. We first provide a short overview of the current standing of the complications experienced with BMP products. We then discuss the different delivery parameters studied in association with BMPs, and their influence on the efficacy and safety of BMP treatments. In particular, the literature addressing the stability of BMPs and their possible interactions with components of the delivery system as well as their sensitivity to conditions of the formulation process is reviewed. In summary, recent developments in the fields of bioengineering and biopharmaceuticals suggest that a good understanding of the relationship between the formulation/delivery conditions and the stability of growth factors such as BMPs is a prerequisite for a safe and effective treatment.
Collapse
|
18
|
Visser R, Rico-Llanos GA, Pulkkinen H, Becerra J. Peptides for bone tissue engineering. J Control Release 2016; 244:122-135. [PMID: 27794492 DOI: 10.1016/j.jconrel.2016.10.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/21/2016] [Accepted: 10/23/2016] [Indexed: 01/07/2023]
Abstract
Molecular signals in the form of growth factors are the main modulators of cell behavior. However, the use of growth factors in tissue engineering has several drawbacks, including their costs, difficult production, immunogenicity and short half-life. Furthermore, many of them are pleiotropic and, since a single growth factor can have different active domains, their effect is not always fully controllable. A very interesting alternative that has recently emerged is the use of biomimetic peptides. Sequences derived from the active domains of soluble or extracellular matrix proteins can be used to functionalize the biomaterials used as scaffolds for new tissue growth to either direct the attachment of cells or to be released as soluble ligands. Since these short peptides can be easily designed and cost-effectively synthesized in vitro, their use has opened up a world of new opportunities to obtain cheaper and more effective implants for regenerative medicine strategies. In this extensive review we will go through many of the most important peptides with potential interest for bone tissue engineering, not limiting to those that only mediate cell adhesion or induce the osteogenic differentiation of progenitor cells, but also focusing on those that direct angiogenesis because of its close relation with bone formation.
Collapse
Affiliation(s)
- Rick Visser
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain; Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, Spain; BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Junta de Andalucia, University of Malaga, Spain.
| | - Gustavo A Rico-Llanos
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain; Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, Spain; BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Junta de Andalucia, University of Malaga, Spain
| | - Hertta Pulkkinen
- BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Junta de Andalucia, University of Malaga, Spain; Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Jose Becerra
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain; Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, Spain; BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Junta de Andalucia, University of Malaga, Spain
| |
Collapse
|
19
|
Lee S, Shen J, Pan HC, Shrestha S, Asatrian G, Nguyen A, Meyers C, Nguyen V, Lee M, Soo C, Ting K, James AW. Calvarial Defect Healing Induced by Small Molecule Smoothened Agonist. Tissue Eng Part A 2016; 22:1357-1366. [PMID: 27702396 DOI: 10.1089/ten.tea.2016.0167] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hedgehog (Hh) signaling positively regulates both endochondral and intramembranous ossification. Use of small molecules for tissue engineering applications poses several advantages. In this study, we examined whether use of an acellular scaffold treated with the small molecule Smoothened agonist (SAG) could aid in critical-size mouse calvarial defect repair. First, we verified the pro-osteogenic effect of SAG in vitro, using primary neonatal mouse calvarial cells (NMCCs). Next, a 4 mm nonhealing defect was created in the mid-parietal bone of 10-week-old CD-1 mice. The scaffold consisted of a custom-fabricated poly(lactic-co-glycolic acid) disc with hydroxyapatite coating (measuring 4 mm diameter × 0.5 mm thickness). Treatment groups included dimethylsulfoxide control (n = 6), 0.5 mM SAG (n = 7) or 1.0 mM SAG (n = 7). Evaluation was performed at 4 and 8 weeks postoperative, by a combination of high-resolution microcomputed tomography, histology (H & E, Masson's Trichrome), histomorphometry, and immunohistochemistry (BSP, OCN, VEGF). In vivo results showed that SAG treatment induced a significant and dose-dependent increase in calvarial bone healing by all radiographic parameters. Histomorphometric analysis showed an increase in all parameters of bone formation with SAG treatment, but also an increase in blood vessel number and density. In summary, SAG is a pro-osteogenic, provasculogenic stimulus when applied locally in a bone defect environment.
Collapse
Affiliation(s)
- Soonchul Lee
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California.,2 Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University , Republic of Korea.,3 Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California , Los Angeles, Los Angeles, California
| | - Jia Shen
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Hsin Chuan Pan
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Swati Shrestha
- 3 Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California , Los Angeles, Los Angeles, California
| | - Greg Asatrian
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Alan Nguyen
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Carolyn Meyers
- 4 Department of Pathology, Johns Hopkins University , Baltimore, Maryland
| | - Vi Nguyen
- 1 Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Min Lee
- 5 Section of Biomaterials, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Chia Soo
- 3 Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California , Los Angeles, Los Angeles, California.,6 Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California , Los Angeles, Los Angeles, California
| | - Kang Ting
- 2 Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University , Republic of Korea.,3 Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California , Los Angeles, Los Angeles, California
| | - Aaron W James
- 3 Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California , Los Angeles, Los Angeles, California.,4 Department of Pathology, Johns Hopkins University , Baltimore, Maryland
| |
Collapse
|
20
|
Glowacki J. Demineralized Bone and BMPs: Basic Science and Clinical Utility. J Oral Maxillofac Surg 2015; 73:S126-31. [DOI: 10.1016/j.joms.2015.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/08/2015] [Indexed: 10/22/2022]
|
21
|
Balmayor ER. Targeted delivery as key for the success of small osteoinductive molecules. Adv Drug Deliv Rev 2015; 94:13-27. [PMID: 25959428 DOI: 10.1016/j.addr.2015.04.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/20/2015] [Accepted: 04/29/2015] [Indexed: 02/08/2023]
Abstract
Molecules such as growth factors, peptides and small molecules can guide cellular behavior and are thus important for tissue engineering. They are rapidly emerging as promising compounds for the regeneration of tissues of the musculoskeletal system. Growth factors have disadvantages such as high cost, short half-life, supraphysiological amounts needed, etc. Therefore, small molecules may be an alternative. These molecules have been discovered using high throughput screening. Small osteoinductive molecules exhibit several advantages over growth factors owing to their small sizes, such as high stability and non-immunogenicity. These molecules may stimulate directly signaling pathways that are important for osteogenesis. However, systemic application doesn't induce osteogenesis in most cases. Therefore, local administration is needed. This may be achieved by using a bone graft material providing additional osteoconductive properties. These graft materials can also act by themselves as a delivery matrix for targeted and local delivery. Furthermore, vascularization is necessary in the process of osteogenesis. Many of the small molecules are also capable of promoting vascularization of the tissue to be regenerated. Thus, in this review, special attention is given to molecules that are capable of inducing both angiogenesis and osteogenesis simultaneously. Finally, more recent preclinical and clinical uses in bone regeneration of those molecules are described, highlighting the needs for the clinical translation of these promising compounds.
Collapse
|
22
|
Kim YH, Tabata Y. Dual-controlled release system of drugs for bone regeneration. Adv Drug Deliv Rev 2015; 94:28-40. [PMID: 26079284 DOI: 10.1016/j.addr.2015.06.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/23/2015] [Accepted: 06/08/2015] [Indexed: 02/08/2023]
Abstract
Controlled release systems have been noted to allow drugs to enhance their ability for bone regeneration. To this end, various biomaterials have been used as the release carriers of drugs, such as low-molecular-weight drugs, growth factors, and others. The drugs are released from the release carriers in a controlled fashion to maintain their actions for a long time period. Most research has been focused on the controlled release of single drugs to demonstrate the therapeutic feasibility. Controlled release of two combined drugs, so-called dual release systems, are promising and important for tissue regeneration. This is because the tissue regeneration process of bone formation is generally achieved by multiple bioactive molecules, which are produced from cells by other molecules. If two types of bioactive molecules, (i.e., drugs), are supplied in an appropriate fashion, the regeneration process of living bodies will be efficiently promoted. This review focuses on the bone regeneration induced by dual-controlled release of drugs. In this paper, various dual-controlled release systems of drugs aiming at bone regeneration are overviewed explaining the type of drugs and their release materials.
Collapse
|
23
|
Tian H, Li CS, Scott TP, Montgomery SR, Phan K, Lao L, Zhang W, Li Y, Hayashi T, Takahashi S, Alobaidaan R, Ruangchainikom M, Zhao KW, Brochmann EJ, Murray SS, Wang JC, Daubs MD. Secreted phosphoprotein 24 kD inhibits nerve root inflammation induced by bone morphogenetic protein-2. Spine J 2015; 15:314-21. [PMID: 25264179 DOI: 10.1016/j.spinee.2014.09.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 08/10/2014] [Accepted: 09/17/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Bone morphogenetic protein-2 (BMP-2) has been used to successfully promote spine fusion, but side-effects including nerve inflammation have been observed. PURPOSE To investigate the direct neurotoxic effects of BMP-2 and test the hypotheses that the use of BMP binding proteins, such as secreted phosphoprotein 24 kD (Spp24), can reduce or eliminate these effects. STUDY DESIGN In vitro experiments and in vivo analysis in a rodent model. METHODS In vitro, dorsal root ganglion cells were cultured in the presence of BMP-2 with and without Spp24 and calcitonin gene-related peptide and Substance P, markers of neuroinflammation, were measured by immunohistochemistry. In vivo, rats underwent a left-sided laminotomy at L5 to expose the S1 nerve root and were randomized into four different groups according to the intervention at the laminotomy site: collagen sponge only (no BMP-2 or Spp24), BMP-2 in a collagen sponge only, BMP-2 in a collagen sponge+an empty collagen sponge to act as a barrier, and BMP-2 in a collagen sponge+Spp24 in a collagen sponge to act as a barrier. Functional evaluation was done using the Basso, Beattie, and Bresnahan scale and immunohistochemical analyses were performed using calcitonin gene-related peptide and Substance P staining. RESULTS The neuroinflammatory effects of BMP-2 in vitro were ameliorated by the addition of Spp24. Similarly, in vivo, Spp24 reduced the expression of markers on neuroinflammation in animals treated with BMP-2 and also improved the function after BMP-2 administration. CONCLUSIONS These results confirm that BMP binding proteins have great potential as adjuvant therapies to limit BMP-2 related side-effects in spine surgery.
Collapse
Affiliation(s)
- Haijun Tian
- Department of Orthopaedic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Rd., Shanghai, 200003, P.R. China; Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA.
| | - Chen-Shuang Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, 22 South Zhongguancun St., Beijing 100081, P.R. China
| | - Trevor P Scott
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Scott R Montgomery
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Kevin Phan
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Lifeng Lao
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Wei Zhang
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Yawei Li
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Tetsuo Hayashi
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Shinji Takahashi
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Raed Alobaidaan
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Monchai Ruangchainikom
- Department of Orthopaedic Surgery, University of California, 675 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Ke-Wei Zhao
- Research Service, VA Greater Los Angeles Healthcare System, 16111 Plummer St., North Hills, CA 91343, USA
| | - Elsa J Brochmann
- Research Service, VA Greater Los Angeles Healthcare System, 16111 Plummer St., North Hills, CA 91343, USA; Geriatric Research, Education and Clinical Center, VA Greater Los Angeles Healthcare System, 16111 Plummer St., North Hills, CA 91343, USA; Department of Medicine, University of California, 650 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Samuel S Murray
- Research Service, VA Greater Los Angeles Healthcare System, 16111 Plummer St., North Hills, CA 91343, USA; Geriatric Research, Education and Clinical Center, VA Greater Los Angeles Healthcare System, 16111 Plummer St., North Hills, CA 91343, USA; Department of Medicine, University of California, 650 Charles E. Young Dr. S., Los Angeles, CA 90024, USA
| | - Jeffrey C Wang
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, 1520 San Pablo, HCT Suite 2000, Los Angeles, CA 90033, USA
| | - Michael D Daubs
- Division of Orthopaedic Surgery, Department of Surgery, University of Nevada School of Medicine, 2040 W Charleston Blvd, Suite 601, Las Vegas, NV 89102, USA
| |
Collapse
|
24
|
Cushnie EK, Ulery BD, Nelson SJ, Deng M, Sethuraman S, Doty SB, Lo KWH, Khan YM, Laurencin CT. Simple signaling molecules for inductive bone regenerative engineering. PLoS One 2014; 9:e101627. [PMID: 25019622 PMCID: PMC4096515 DOI: 10.1371/journal.pone.0101627] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 06/09/2014] [Indexed: 11/18/2022] Open
Abstract
With greater than 500,000 orthopaedic procedures performed in the United States each year requiring a bone graft, the development of novel graft materials is necessary. We report that some porous polymer/ceramic composite scaffolds possess intrinsic osteoinductivity as shown through their capacity to induce in vivo host osteoid mineralization and in vitro stem cell osteogenesis making them attractive synthetic bone graft substitutes. It was discovered that certain low crystallinity ceramics partially dissociate into simple signaling molecules (i.e., calcium and phosphate ions) that induce stem cells to endogenously produce their own osteoinductive proteins. Review of the literature has uncovered a variety of simple signaling molecules (i.e., gases, ions, and redox reagents) capable of inducing other desirable stem cell differentiation through endogenous growth factor production. Inductive simple signaling molecules, which we have termed inducerons, represent a paradigm shift in the field of regenerative engineering where they can be utilized in place of recombinant protein growth factors.
Collapse
Affiliation(s)
- Emily K. Cushnie
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Bret D. Ulery
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Stephen J. Nelson
- School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Meng Deng
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Swaminathan Sethuraman
- Center for Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Stephen B. Doty
- Hospital for Special Surgery, New York, New York, United States of America
| | - Kevin W. H. Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Medicine, Division of Endocrinology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Yusuf M. Khan
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut, United States of America
| | - Cato T. Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
25
|
Delivery of small molecules for bone regenerative engineering: preclinical studies and potential clinical applications. Drug Discov Today 2014; 19:794-800. [PMID: 24508820 DOI: 10.1016/j.drudis.2014.01.012] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 01/01/2014] [Accepted: 01/31/2014] [Indexed: 12/20/2022]
Abstract
Stimulation of bone regeneration using growth factors is a promising approach for musculoskeletal regenerative engineering. However, common limitations with protein growth factors, such as high manufacturing costs, protein instability, contamination issues, and unwanted immunogenic responses of the host reduce potential clinical applications. New strategies for bone regeneration that involve inexpensive and stable small molecules can obviate these problems and have a significant impact on the treatment of skeletal injury and diseases. Over the past decade, a large number of small molecules with the potential of regenerating skeletal tissue have been reported in the literature. Here, we review this literature, paying specific attention to the prospects for small molecule-based bone-regenerative engineering. We also review the preclinical study of small molecules associated with bone regeneration.
Collapse
|
26
|
Spinal fusion in the next generation: gene and cell therapy approaches. ScientificWorldJournal 2014; 2014:406159. [PMID: 24672316 PMCID: PMC3927763 DOI: 10.1155/2014/406159] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/28/2013] [Indexed: 12/24/2022] Open
Abstract
Bone fusion represents a challenge in the orthopedics practice, being especially indicated for spine disorders. Spinal fusion can be defined as the bony union between two vertebral bodies obtained through the surgical introduction of an osteoconductive, osteoinductive, and osteogenic compound. Autogenous bone graft provides all these three qualities and is considered the gold standard. However, a high morbidity is associated with the harvest procedure. Intensive research efforts have been spent during the last decades to develop new approaches and technologies for successful spine fusion. In recent years, cell and gene therapies have attracted great interest from the scientific community. The improved knowledge of both mesenchymal stem cell biology and osteogenic molecules allowed their use in regenerative medicine, representing attractive approaches to achieve bone regeneration also in spinal surgery applications. In this review we aim to describe the developing gene- and cell-based bone regenerative approaches as promising future trends in spine fusion.
Collapse
|
27
|
DiGiovanni CW, Lin S, Pinzur M. Recombinant human PDGF-BB in foot and ankle fusion. Expert Rev Med Devices 2014; 9:111-22. [DOI: 10.1586/erd.11.76] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
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: 2.5] [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
|
29
|
Sonnet C, Simpson CL, Olabisi RM, Sullivan K, Lazard Z, Gugala Z, Peroni JF, Weh JM, Davis AR, West JL, Olmsted-Davis EA. Rapid healing of femoral defects in rats with low dose sustained BMP2 expression from PEGDA hydrogel microspheres. J Orthop Res 2013; 31:1597-604. [PMID: 23832813 DOI: 10.1002/jor.22407] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 05/13/2013] [Indexed: 02/04/2023]
Abstract
Current strategies for bone regeneration after traumatic injury often fail to provide adequate healing and integration. Here, we combined the poly (ethylene glycol) diacrylate (PEGDA) hydrogel with allogeneic "carrier" cells transduced with an adenovirus expressing BMP2. The system is unique in that the biomaterial encapsulates the cells, shielding them and thus suppressing destructive inflammatory processes. Using this system, complete healing of a 5 mm-long femur defect in a rat model occurs in under 3 weeks, through secretion of 100-fold lower levels of protein as compared to doses of recombinant BMP2 protein used in studies which lead to healing in 2-3 months. New bone formation was evaluated radiographically, histologically, and biomechanically at 2, 3, 6, 9, and 12 weeks after surgery. Rapid bone formation bridged the defect area and reliably integrated into the adjacent skeletal bone as early as 2 weeks. At 3 weeks, biomechanical analysis showed the new bone to possess 79% of torsional strength of the intact contralateral femur. Histological evaluation showed normal bone healing, with no infiltration of inflammatory cells with the bone being stable approximately 1 year later. We propose that these osteoinductive microspheres offer a more efficacious and safer clinical option over the use of rhBMP2.
Collapse
Affiliation(s)
- Corinne Sonnet
- Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Alkek Building, Room N1010, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lo KWH, Ashe KM, Kan HM, Laurencin CT. The role of small molecules in musculoskeletal regeneration. Regen Med 2013; 7:535-49. [PMID: 22817627 DOI: 10.2217/rme.12.33] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The uses of bone morphogenetic proteins and parathyroid hormone therapeutics are fraught with several fundamental problems, such as cost, protein stability, immunogenicity, contamination and supraphysiological dosage. These downsides may effectively limit their more universal use. Therefore, there is a clear need for alternative forms of biofactors to obviate the drawbacks of protein-based inductive factors for bone repair and regeneration. Our group has studied small molecules with the capacity to regulate osteoblast differentiation and mineralization because their inherent physical properties minimize limitations observed in protein growth factors. For instance, in general, small molecule inducers are usually more stable, highly soluble, nonimmunogenic, more affordable and require lower dosages. Small molecules with the ability to induce osteoblastic differentiation may represent the next generation of bone regenerative medicine. This review describes efforts to develop small molecule-based biofactors for induction, paying specific attention to their novel roles in bone regeneration.
Collapse
Affiliation(s)
- Kevin W-H Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | | | | | | |
Collapse
|
31
|
Koo KH, Lee JM, Ahn JM, Kim BS, La WG, Kim CS, Im GI. Controlled Delivery of Low-Dose Bone Morphogenetic Protein-2 Using Heparin-Conjugated Fibrin in the Posterolateral Lumbar Fusion of Rabbits. Artif Organs 2013; 37:487-94. [DOI: 10.1111/j.1525-1594.2012.01578.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ki-Hyoung Koo
- Department of Orthopaedic Surgery; Dongguk University Ilsan Hospital; Goyang
| | - Jong-Min Lee
- Department of Orthopaedic Surgery; Dongguk University Ilsan Hospital; Goyang
| | - Jung-Min Ahn
- Department of Orthopaedic Surgery; Dongguk University Ilsan Hospital; Goyang
| | | | | | - Chang-Sung Kim
- Department of Periodontology; College of Dentistry; Yonsei University; Seoul; Korea
| | - Gun-Il Im
- Department of Orthopaedic Surgery; Dongguk University Ilsan Hospital; Goyang
| |
Collapse
|
32
|
Freire MO, Kim HK, Kook JK, Nguyen A, Zadeh HH. Antibody-mediated osseous regeneration: the early events in the healing response. Tissue Eng Part A 2013. [PMID: 23190409 DOI: 10.1089/ten.tea.2012.0282] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bone engineering strategies often exploit modulation of the extracellular environment, including delivery of cell and growth factors to repair and regenerate damaged tissues. During bone healing, the expression of endogenous bone morphogenetic proteins is an essential component of the healing response. However, in some situations, the inherent reparative capacity available in the local microenvironment is exceeded by the requirements of the defects. We have recently reported on a novel strategy, that exploits the specificity of antibodies to capture and make available endogenous osteogenic growth factors, referred to as "antibody-mediated osseous regeneration" (AMOR). The objective of the present study was to identify some of the cellular and molecular events involved in AMOR in an effort to begin to elucidate the mechanism of AMOR. The rat critical-sized calvarial defect model was used, where anti-bone morphogenetic protein (BMP)-2 monoclonal antibody (mAb), isotype-control mAb, or recombinant human (rh)BMP-2 were immobilized on absorbable collagen calvarial sponge (ACS) by adsorption, and then implanted into calvarial defects. The results demonstrated persistence of implanted mAbs for short term from 1 to 2 weeks after implantation. Increased cell infiltration was found in defects treated with anti-BMP-2 mAb. Examination of proteins on ACS scaffolds retrieved from defect sites demonstration increased levels of BMP-2, BMP-4, and BMP-7 proteins in sites implanted with anti-BMP-2 mAb. Moreover, BMP-2, BMP-4, and BMP-7 gene expression levels were increased in sites implanted with anti-BMP-2 mAb. Micro-computed tomography and histological analysis demonstrated that the bone within calvarial defects was fully regenerated in sites implanted with either anti-BMP-2 mAb or rhBMP-2. However, rhBMP-2-regenerated bone exhibited aberrant histomorphology with dystrophic calcification and invasion of subjacent areas. Altogether, the results revealed evidence for anti-BMP-2 mAbs to form an immune complex with BMP-2, BMP-4, and BMP-7, and bind to cells to mediate osteogenesis bone regeneration in vivo. This approach suggests a significant role for antibodies in regenerative orthopedic medicine.
Collapse
Affiliation(s)
- Marcelo O Freire
- Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California 90089, USA
| | | | | | | | | |
Collapse
|
33
|
Choi H, Park NJ, Jamiyandorj O, Hong MH, Oh S, Park YB, Kim S. Improvement of osteogenic potential of biphasic calcium phosphate bone substitute coated with synthetic cell binding peptide sequences. J Periodontal Implant Sci 2012. [PMID: 23185697 PMCID: PMC3498301 DOI: 10.5051/jpis.2012.42.5.166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Purpose The aim of this study was to evaluate the improvement of osteogenic potential of biphasic calcium phosphate (BCP) bone substitute coated with synthetic cell-binding peptide sequences in a standardized rabbit sinus model. Methods Standardized 6-mm diameter defects were created bilaterally on the maxillary sinus of ten male New Zealand white rabbits, receiving BCP bone substitute coated with synthetic cell binding peptide sequences on one side (experimental group) and BCP bone substitute without coating (control group) on the other side. Histologic and histomorphometric analysis of bone formation was carried out after a healing period of 4 or 8 weeks. Results Histological analysis revealed signs of new bone formation in both experimental groups (4- and 8-week healing groups) with a statistically significant increase in bone formation in the 4-week healing group compared to the control group. However, no statistically significant difference in bone formation was found between the 8-week healing group and the control group. Conclusions This study found that BCP bone substitute coated with synthetic cell-binding peptide sequences enhanced osteoinductive potential in a standardized rabbit sinus model and its effectiveness was greater in the 4-week healing group than in the 8-week healing group.
Collapse
Affiliation(s)
- Hyunmin Choi
- Department of Prosthodontics, Yonsei University College of Dentistry, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Lo KWH, Ulery BD, Kan HM, Ashe KM, Laurencin CT. Evaluating the feasibility of utilizing the small molecule phenamil as a novel biofactor for bone regenerative engineering. J Tissue Eng Regen Med 2012; 8:728-36. [PMID: 22815259 DOI: 10.1002/term.1573] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 05/17/2012] [Accepted: 06/12/2012] [Indexed: 01/30/2023]
Abstract
Osteoblast cell adhesion and differentiation on biomaterials are important achievements necessary for implants to be useful in bone regenerative engineering. Recombinant bone morphogenetic proteins (BMPs) have been shown to be important for these processes; however, there are many challenges associated with the widespread use of these proteins. A recent report demonstrated that the small molecule phenamil, a diuretic derivative, was able to induce osteoblast differentiation and mineralization in vitro via the canonical BMP signalling cascade (Park et al., 2009). In this study, the feasibility of using phenamil as a novel biofactor in conjunction with a biodegradable poly(lactide-co-glycolide acid) (PLAGA) polymeric scaffold for engineering bone tissue was evaluated. The in vitro cellular behaviour of osteoblast-like MC3T3-E1 cells cultured on PLAGA scaffolds in the presence of phenamil at 10 μM were characterized with regard to initial cell adhesion, proliferation, alkaline phosphatase (ALP) activity and matrix mineralization. The results demonstrate that phenamil supported cell proliferation, promoted ALP activity and facilitated matrix mineralization of osteoblast-like MC3T3-E1 cells. Moreover, in this study, we found that phenamil promoted integrin-mediated cell adhesion on PLAGA scaffolds. It was also shown that phenamil encapsulated within porous, microsphere PLAGA scaffolds retained its osteogenic activity upon release. Based on these findings, the small molecule phenamil has the potential to serve as a novel biofactor for the repair and regeneration of bone tissues.
Collapse
Affiliation(s)
- Kevin W-H Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT, USA; Department of Medicine, Division of Endocrinology, University of Connecticut Health Center, School of Medicine, Farmington, CT, USA
| | | | | | | | | |
Collapse
|
35
|
Koo KH, Yeo DH, Ahn JM, Kim BS, Kim CS, Im GI. Lumbar Posterolateral Fusion Using Heparin-Conjugated Fibrin for Sustained Delivery of Bone Morphogenic Protein-2 in a Rabbit Model. Artif Organs 2012; 36:629-34. [DOI: 10.1111/j.1525-1594.2012.01444.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Wadagaki R, Mizuno D, Yamawaki-Ogata A, Satake M, Kaneko H, Hagiwara S, Yamamoto N, Narita Y, Hibi H, Ueda M. Osteogenic Induction of Bone Marrow-Derived Stromal Cells on Simvastatin-Releasing, Biodegradable, Nano- to Microscale Fiber Scaffolds. Ann Biomed Eng 2011; 39:1872-81. [PMID: 21590488 DOI: 10.1007/s10439-011-0327-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 02/23/2011] [Indexed: 11/29/2022]
Affiliation(s)
- Ryu Wadagaki
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lee CK, Koo KT, Park YJ, Lee JY, Rhee SH, Ku Y, Rhyu IC, Chung CP. Biomimetic surface modification using synthetic oligopeptides for enhanced guided bone regeneration in beagles. J Periodontol 2011; 83:101-10. [PMID: 21542731 DOI: 10.1902/jop.2011.110040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND In previous studies, oligopeptide corresponding to the cell-binding domains of bone morphogenetic proteins that bind to bone morphogenetic protein receptor enhanced the bone regenerative capacity of bovine bone minerals (BBM). The aim of this study is to evaluate the ability of BBM coated with oligopeptide to promote periodontal regeneration in a 1-wall intrabony defect model in dogs. METHODS The second and third mandibular premolars and first molars of six adult beagles were extracted bilaterally, and the extraction sites were allowed to heal for 10 weeks. The 1-wall intrabony defects were prepared bilaterally on the mesial and distal side of the fourth mandibular premolars. Twenty-four intrabony defects were assigned to four treatment groups: 1) open flap debridement; 2) guided tissue regeneration (GTR); 3) GTR with a collagen membrane and BBM; and 4) GTR with a collagen membrane and BBM coated with the oligopeptide (Pep-BBM). The animals were sacrificed 10 weeks after surgery. For the histometric analysis, defect height, junctional epithelium migration, new cementum, new bone height, and new bone area were measured. New bone volume was measured using microcomputed tomography. RESULTS Wound healing was generally uneventful. For junctional epithelium migration, the BBM and Pep-BBM groups exhibited mean (± SE) values of 0.53 ± 0.41 and 0.48 ± 0.30 mm, and for new cementum height, 1.71 ± 0.46 and 2.50 ± 2.00 mm, respectively. For junctional epithelium migration and cementum regeneration, there were no significant differences between the two groups. The mean (± SE) values of new bone height and new bone volume in the Pep-BBM group (3.88 ± 0.31 mm and 32.35% ± 9.60%) were significantly greater than the mean values for the BBM group (2.60 ± 0.41 mm and 20.56% ± 1.89%). For bone regeneration, the Pep-BBM group showed superior results compared to the BBM group with statistically significant differences. CONCLUSIONS Through various parameters to evaluate periodontal regeneration, this oligopeptide coating influenced only the ability of BBM to promote bone regeneration in 1-wall intrabony defects in beagles. Junctional epithelium migration and cementum regeneration were not affected by this oligopeptide coating, and further investigations with special focus on regeneration of the periodontal ligament are necessary.
Collapse
Affiliation(s)
- Chang-Kyun Lee
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Lo KWH, Kan HM, Ashe KM, Laurencin CT. The small molecule PKA-specific cyclic AMP analogue as an inducer of osteoblast-like cells differentiation and mineralization. J Tissue Eng Regen Med 2011; 6:40-8. [PMID: 21312339 DOI: 10.1002/term.395] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Accepted: 11/11/2010] [Indexed: 12/21/2022]
Abstract
Osteoblastic differentiation is an important landmark for bone formation, bone repair and regeneration; however, it is a very complex process controlled by different signalling mechanisms. Several groups have reported that the cyclic adenosine monophosphate (cAMP) signalling system is responsible for regulating osteoblast cell differentiation. Nonetheless, to date, the principle role of the cAMP molecules related to this process remains controversial. Moreover, the underlying cAMP-dependent signalling cascade governing the osteoblastic differentiation has not been clarified. In this study we investigated the roles of the cAMP-dependent protein kinase A (PKA) signalling in proliferation, differentiation and mineralization of osteoblast-like MC3T3-E1 cells, using the PKA-specific small molecule cAMP analogue, 6-Bnz-cAMP, at 100 µM. Alkaline phosphatase (ALP) activity, runt transcription factor 2 (Runx2), osteopontin (OPN) and osteocalcin (OCN) protein expressions were used as osteoblast-specific markers to demonstrate osteoblastic differentiation. Further, calcium measurement of the extracellular matrix was employed as the hallmark of matrix mineralization or calcification. We report here that activation of PKA by the small molecule 6-Bnz-cAMP induces osteoblastic differentiation and matrix mineralization of osteoblast-like MC3T3-E1 cells. Moreover, 6-Bnz-cAMP does not induce cytotoxicity to the cells, as revealed by our cell proliferation studies. Therefore, based on these findings, we propose that the PKA-specific small molecule 6-Bnz-cAMP may serve as a novel bone-inducing growth factor for repairing and regenerating bone tissues during bone-regenerative engineering.
Collapse
Affiliation(s)
- Kevin W-H Lo
- Department of Orthopaedic Surgery, Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | |
Collapse
|
39
|
Wang L, Zou D, Zhang S, Zhao J, Pan K, Huang Y. Repair of bone defects around dental implants with bone morphogenetic protein/fibroblast growth factor-loaded porous calcium phosphate cement: a pilot study in a canine model. Clin Oral Implants Res 2011; 22:173-81. [DOI: 10.1111/j.1600-0501.2010.01976.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
The effect of growth factors for bone augmentation to enable dental implant placement: A systematic review. JAPANESE DENTAL SCIENCE REVIEW 2010. [DOI: 10.1016/j.jdsr.2009.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
41
|
Alaoui-Ismaili MH, Falb D. Design of second generation therapeutic recombinant bone morphogenetic proteins. Cytokine Growth Factor Rev 2009; 20:501-7. [DOI: 10.1016/j.cytogfr.2009.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|