1
|
Ureiro-Cueto G, Rodil SE, Silva-Bermúdez P, Santana-Vázquez M, Hoz-Rodríguez L, Arzate H, Montoya-Ayala G. Amorphous titanium oxide (aTiO 2) thin films biofunctionalized with CAP-p15 induce mineralized-like differentiation of human oral mucosal stem cells (hOMSCs). Biomed Mater 2024; 19:055003. [PMID: 38917837 DOI: 10.1088/1748-605x/ad5bab] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/25/2024] [Indexed: 06/27/2024]
Abstract
Insufficient osseointegration of titanium-based implants is a factor conditioning their long-term success. Therefore, different surface modifications, such as multifunctional oxide coatings, calcium phosphates, and the addition of molecules such as peptides, have been developed to improve the bioactivity of titanium-based biomaterials. In this work, we investigate the behavior of human oral mucosal stem cells (hOMSCs) cultured on amorphous titanium oxide (aTiO2), surfaces designed to simulate titanium (Ti) surfaces, biofunctionalized with a novel sequence derived from cementum attachment protein (CAP-p15), exploring its impact on guiding hOMSCs towards an osteogenic phenotype. We carried out cell attachment and viability assays. Next, hOMSCs differentiation was assessed by red alizarin stain, ALP activity, and western blot analysis by evaluating the expression of RUNX2, BSP, BMP2, and OCN at the protein level. Our results showed that functionalized surfaces with CAP-p15 (1 µg ml-1) displayed a synergistic effect increasing cell proliferation and cell attachment, ALP activity, and expression of osteogenic-related markers. These data demonstrate that CAP-p15 and its interaction with aTiO2surfaces promote osteoblastic differentiation and enhanced mineralization of hOMSCs when compared to pristine samples. Therefore, CAP-p15 shows the potential to be used as a therapeutical molecule capable of inducing mineralized tissue regeneration onto titanium-based implants.
Collapse
Affiliation(s)
- Guadalupe Ureiro-Cueto
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Sandra E Rodil
- Instituto de Investigaciones en Materiales, UNAM, Mexico
| | - Phaedra Silva-Bermúdez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico
| | - Maricela Santana-Vázquez
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Lia Hoz-Rodríguez
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Higinio Arzate
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Gonzalo Montoya-Ayala
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| |
Collapse
|
2
|
Sanyal S, Rajput S, Sadhukhan S, Rajender S, Mithal A, Chattopadhyay N. Polymorphisms in the Runx2 and osteocalcin genes affect BMD in postmenopausal women: a systematic review and meta-analysis. Endocrine 2024; 84:63-75. [PMID: 38055125 DOI: 10.1007/s12020-023-03621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023]
Abstract
PURPOSE Runx2 and osteocalcin have pivotal roles in bone homeostasis. Polymorphism of these two genes could alter the function of osteoblasts and consequently bone mineral density (BMD). Attempts to understand the relationship between these polymorphisms and BMD in postmenopausal women across a variety of populations have yielded inconsistent results. This meta-analysis seeks to define the relationship between these polymorphisms with BMD in postmenopausal women. METHODS Eligible studies were identified from three electronic databases. Data were extracted from the eligible studies (4 studies on Runx2 and 6 studies on osteocalcin), and associations of Runx2 T > C and osteocalcin HindIII polymorphisms with BMD in postmenopausal women were assessed using standard difference in means (SDM) and 95% confidence intervals (CI) as statistical measures. RESULTS A significant difference in the lumbar spine (LS) BMD in postmenopausal women was observed between the TT and CC homozygotes for the Runx2 T > C (SDM = -0.445, p-value = 0.034). The mutant genotypes (CC) showed significantly lower LS BMD in comparison to wild type genotypes under recessive model of genetic analysis (TC + TT vs. CC: SDM = -0.451, p-value = 0.032). For osteocalcin, HindIII polymorphism, the mutant genotypes (HH) was associated with significantly higher BMD for both LS and femoral neck (FN) than the wild type (hh) homozygotes (SDM = 0.152, p-value = 0.008 and SDM = 0.139, p-value = 0.016 for LS and FN, respectively). There was no association between total hip (TH) BMD and the osteocalcin HindIII polymorphism. CONCLUSIONS Runx2 T > C and osteocalcin HindIII polymorphisms influence the level of BMD in postmenopausal women and may be used as predictive markers of osteoporosis.
Collapse
Affiliation(s)
- Somali Sanyal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, Uttar Pradesh, 226018, India.
| | - Swati Rajput
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sreyanko Sadhukhan
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Singh Rajender
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
| | - Ambrish Mithal
- Institute of Endocrinology and Diabetes, Max Healthcare, Institutional Area, Press Enclave Road, Saket, New Delhi, India.
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
3
|
Xu HJ, Liu XZ, Yang L, Ning Y, Xu LL, Sun DM, Liao W, Yang Y, Li ZH. Runx2 overexpression promotes bone repair of osteonecrosis of the femoral head (ONFH). Mol Biol Rep 2023; 50:4769-4779. [PMID: 37029290 DOI: 10.1007/s11033-023-08411-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/28/2023] [Indexed: 04/09/2023]
Abstract
BACKGROUND Runt-related transcription factor-2 (Runx2) has been considered an inducer to improve bone repair ability of mesenchymal stem cells (MSCs). METHODS AND RESULTS Twenty-four rabbits were used to establish Osteonecrosis of the femoral head (ONFH) and randomly devided into four groups: Adenovirus Runx2 (Ad-Runx2) group, Runx2-siRNA group, MSCs group and Model group. At 1 week after model establishment, the Ad-Runx2 group was treated with 5 × 107 MSCs transfected through Ad-Runx2, the Runx2-siRNA group was treated with 5 × 107 MSCs transfected through Runx2-siRNA, the MSCs group was injected with 5 × 107 untreated MSCs, and the Model group was treated with saline. The injection was administered at 1 week and 3 weeks after model establishment. The expression of bone morphogenetic protein 2 (BMP-2), Runx2 and Osterix from the femoral head was detected at 3 and 6 weeks after MSCs being injected, and Masson Trichrome Staining, Gross Morphology, X-ray and CT images observation were used to evaluate the repair effect of ONFH. The data revealed that the expression of BMP-2, Runx2 and Osterix in the Runx2-siRNA group was reduced at 3 weeks compared with the MSCs group, and then the expression further reduced at 6 weeks, but was still higher than the Model group besides Osterix; The expression of these three genes in the Ad-Runx2 group was higher than in the MSCs group. Masson Trichrome Staining, Gross Morphology and X-ray and CT images observation revealed that necrotic femoral head of the MSCs group was more regular and smooth than the Runx2-siRNA group, which has a collapsed and irregular femoral head. In the Ad-Runx2 group, necrotic femoral head was basically completely repaired and covered by rich cartilage and bone tissue. CONCLUSIONS Overexpression of Runx2 can improve osteoblastic phenotype maintenance of MSCs and promote necrotic bone repair of ONFH.
Collapse
Affiliation(s)
- Hai-Jia Xu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiang-Zhong Liu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Lu Yang
- Department of Anesthesiology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Ning
- Department of Orthopedics, XiangYang Hospital of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Xiangyang, 441000, China
| | | | - Da-Ming Sun
- Wuhan Sports University, Wuhan, 430079, China
| | - Wen Liao
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Yang
- Wuhan Sports University, Wuhan, 430079, China
| | - Zhang-Hua Li
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
4
|
Seyedi Z, Amiri MS, Mohammadzadeh V, Hashemzadeh A, Haddad-Mashadrizeh A, Mashreghi M, Qayoomian M, Hashemzadeh MR, Simal-Gandara J, Taghavizadeh Yazdi ME. Icariin: A Promising Natural Product in Biomedicine and Tissue Engineering. J Funct Biomater 2023; 14:44. [PMID: 36662090 PMCID: PMC9862744 DOI: 10.3390/jfb14010044] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/14/2023] Open
Abstract
Among scaffolds used in tissue engineering, natural biomaterials such as plant-based materials show a crucial role in cellular function due to their biocompatibility and chemical indicators. Because of environmentally friendly behavior and safety, green methods are so important in designing scaffolds. A key bioactive flavonoid of the Epimedium plant, Icariin (ICRN), has a broad range of applications in improving scaffolds as a constant and non-immunogenic material, and in stimulating the cell growth, differentiation of chondrocytes as well as differentiation of embryonic stem cells towards cardiomyocytes. Moreover, fusion of ICRN into the hydrogel scaffolds or chemical crosslinking can enhance the secretion of the collagen matrix and proteoglycan in bone and cartilage tissue engineering. To scrutinize, in various types of cancer cells, ICRN plays a decisive role through increasing cytochrome c secretion, Bax/Bcl2 ratio, poly (ADP-ribose) polymerase as well as caspase stimulations. Surprisingly, ICRN can induce apoptosis, reduce viability and inhibit proliferation of cancer cells, and repress tumorigenesis as well as metastasis. Moreover, cancer cells no longer grow by halting the cell cycle at two checkpoints, G0/G1 and G2/M, through the inhibition of NF-κB by ICRN. Besides, improving nephrotoxicity occurring due to cisplatin and inhibiting multidrug resistance are the other applications of this biomaterial.
Collapse
Affiliation(s)
- Zahra Seyedi
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
- Department of Cancer and Oncology, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
| | | | - Vahideh Mohammadzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Alireza Hashemzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Aliakbar Haddad-Mashadrizeh
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Mohammad Mashreghi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Mohsen Qayoomian
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 91778, Iran
| | - Mohammad Reza Hashemzadeh
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
- Department of Cancer and Oncology, Royesh Stem Cell Biotechnology Institute, Mashhad 9188758156, Iran
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E32004 Ourense, Spain
| | | |
Collapse
|
5
|
Martin KE, Kalelkar PP, Coronel MM, Theriault HS, Schneider RS, García AJ. Host type 2 immune response to xenogeneic serum components impairs biomaterial-directed osteo-regenerative therapies. Biomaterials 2022; 286:121601. [PMID: 35660823 PMCID: PMC11458135 DOI: 10.1016/j.biomaterials.2022.121601] [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: 03/18/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 11/21/2022]
Abstract
The transformative potential of cells as therapeutic agents is being realized in a wide range of applications, from regenerative medicine to cancer therapy to autoimmune disorders. The majority of these therapies require ex vivo expansion of the cellular product, often utilizing fetal bovine serum (FBS) in the culture media. However, the impact of residual FBS on immune responses to cell therapies and the resulting cell therapy outcomes remains unclear. Here, we show that hydrogel-delivered FBS elicits a robust type 2 immune response characterized by infiltration of eosinophils and CD4+ T cells. Host secretion of cytokines associated with type 2 immunity, including IL-4, IL-5, and IL-13, is also increased in FBS-containing hydrogels. We demonstrate that the immune response to xenogeneic serum components dominates the local environment and masks the immunomodulatory effects of biomaterial-delivered mesenchymal stromal/stem cells. Importantly, delivery of relatively small amounts of FBS (3.2% by volume) within BMP-2-containing biomaterial constructs dramatically reduces the ability of these constructs to promote de novo bone formation in a radial defect model in immunocompetent mice. These results urge caution when interpreting the immunological and tissue repair outcomes in immunocompetent pre-clinical models from cells and biomaterial constructs that have come in contact with xenogeneic serum components.
Collapse
Affiliation(s)
- Karen E Martin
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pranav P Kalelkar
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - María M Coronel
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hannah S Theriault
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rebecca S Schneider
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
6
|
Shao H, Wu R, Cao L, Gu H, Chai F. Trelagliptin stimulates osteoblastic differentiation by increasing runt-related transcription factor 2 (RUNX2): a therapeutic implication in osteoporosis. Bioengineered 2021; 12:960-968. [PMID: 33734011 PMCID: PMC8291811 DOI: 10.1080/21655979.2021.1900633] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis, an aging-associated bone metabolic disease, is affecting millions of people worldwide. The deregulated process of osteoblastic differentiation has been linked with the progression of osteoporosis. Trelagliptin is a long-acting inhibitor of DPP-4 used for the management of type 2 diabetes mellitus. However, it is unknown whether Trelagliptin possesses a beneficial effect in osteoblastic differentiation. Interestingly, we found that treatment with Trelagliptin enhanced differentiation and promoted the mineralization of MC3T3-E1 cells. Firstly, Trelagliptin increased the activity of alkaline phosphatase (ALP) and promoted osteoblastic calcium deposition. Additionally, treatment with Trelagliptin upregulated ALP, osteocalcin (OCN), osteopontin (OPN), and bone morphogenetic protein-2 (BMP-2). Notably, Trelagliptin increased RUNX2, a major regulator of osteoblastic differentiation. Mechanistically, Trelagliptin upregulated the levels of p-AMPKα. Blockage of AMPK with compound C abolished the effects of Trelagliptin in RUNX2 and osteoblastic differentiation, suggesting the involvement of AMPK. Our findings suggest that Trelagliptin might possess a potential for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Haiyu Shao
- Department of Orthopedics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Renzheng Wu
- Department of Orthopaedics, Dongyang Garden Tianshi Hospital, Dongyang, Zhejiang, China
| | - Li Cao
- Department of Orthopedics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Haifeng Gu
- Department of Orthopedics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fang Chai
- Department of Orthopedics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Qi S, He J, Han H, Zheng H, Jiang H, Hu CY, Zhang Z, Li X. Anthocyanin-rich extract from black rice (Oryza sativa L. Japonica) ameliorates diabetic osteoporosis in rats. Food Funct 2019; 10:5350-5360. [PMID: 31393485 DOI: 10.1039/c9fo00681h] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Diabetic osteoporosis (DOP) is a systemic endocrine-metabolic osteopathy which has the characteristics of bone mineral density (BMD) reduction and bone microstructural destruction. Although anthocyanin-rich extract from black rice (AEBR) was reported to have a beneficial effect on diabetic rats, no studies have been performed on whether black rice anthocyanins are beneficial for diabetic osteoporosis. Therefore, in this study, a streptozotocin-induced diabetic rat model was established to investigate the protective effect of AEBR on diabetes-induced osteoporosis and its possible mechanism. AEBR at three doses (0.5, 1.0, and 2.0 g kg-1 d-1) were administered by oral gavage to diabetic rats for 8 weeks. The blood glucose, BMD, bone histomorphometry parameters, serum bone turnover biomarkers, bone marrow adipocyte numbers, as well as osteoprotegerin (OPG), runt-related transcription factor 2 (RUNX 2), and receptor activator of nuclear factor-κ B ligand (RANKL) protein expression in bone and serum were detected. The results indicated that AEBR dose-dependently decreased the blood glucose, increased the BMD, and decreased the serum bone turnover markers. The bone microstructure and osteoclast numbers in bone tissues returned to normal in the high AEBR dosage group; at the same time, the AEBR dose-dependently suppressed bone marrow adipogenesis. The RUNX 2 as well as the OPG/RANKL ratio in diabetic rats' bone tissues increased significantly in the AEBR treatment group. Our results indicate that AEBR administration can ameliorate bone loss caused by diabetes; this is mainly attributed to its inhibition of bone turnover, suppression of bone marrow adipogenesis, and up-regulation of RUNX 2 and the OPG/RANKL expression ratio.
Collapse
Affiliation(s)
- Shanshan Qi
- Vitamin D Research Institute, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China. and Shaanxi Black Organic Food Engineering Center, Hanzhong 723000, Shaanxi, China
| | - Jia He
- Vitamin D Research Institute, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China.
| | - Hao Han
- Shaanxi Provincial Bio-resource key Laboratory, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China.
| | - Hongxing Zheng
- Shaanxi Provincial Bio-resource key Laboratory, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China. and Shaanxi Black Organic Food Engineering Center, Hanzhong 723000, Shaanxi, China
| | - Hai Jiang
- Vitamin D Research Institute, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China.
| | - Ching Yuan Hu
- Shaanxi Provincial Bio-resource key Laboratory, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China.
| | - Zhijian Zhang
- Vitamin D Research Institute, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China. and Shaanxi Black Organic Food Engineering Center, Hanzhong 723000, Shaanxi, China
| | - Xinsheng Li
- Shaanxi Provincial Bio-resource key Laboratory, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China.
| |
Collapse
|
8
|
Ramaraju H, Kohn DH. Cell and Material-Specific Phage Display Peptides Increase iPS-MSC Mediated Bone and Vasculature Formation In Vivo. Adv Healthc Mater 2019; 8:e1801356. [PMID: 30835955 DOI: 10.1002/adhm.201801356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/04/2019] [Indexed: 01/27/2023]
Abstract
Biomimetically designed materials matching the chemical and mechanical properties of tissue support higher mesenchymal stem cell (MSC) adhesion. However, directing cell-specific attachment and ensuring uniform cell distribution within the interior of 3D biomaterials remain key challenges in healing critical sized defects. Previously, a phage display derived MSC-specific peptide (DPIYALSWSGMA, DPI) was combined with a mineral binding sequence (VTKHLNQISQSY, VTK) to increase the magnitude and specificity of MSC attachment to calcium-phosphate biomaterials in 2D. This study investigates how DPI-VTK influences quantity and uniformity of iPS-MSC mediated bone and vasculature formation in vivo. There is greater bone formation in vivo when iPS-MSCs are transplanted on bone-like mineral (BLM) constructs coated with DPI-VTK compared to VTK (p < 0.002), uncoated BLM (p < 0.037), acellular BLM/DPI-VTK (p < 0.003), and acellular BLM controls (p < 0.01). This study demonstrates, for the first time, the ability of non-native phage-display designed peptides to spatially control uniform cell distribution on 3D scaffolds and increase the magnitude and uniformity of bone and vasculature formation in vivo. Taken together, the study validates phage display as a novel technology platform to engineer non-native peptides with the ability to drive cell specific attachment on biomaterials, direct bone regeneration, and engineer uniform vasculature in vivo.
Collapse
Affiliation(s)
- Harsha Ramaraju
- Department of Biologic and Material SciencesDepartment of Biomedical EngineeringUniversity of Michigan 1011 N. University Ave, Room 2213 Ann Arbor MI 48109‐1078 USA
| | - David H. Kohn
- Department of Biologic and Material SciencesDepartment of Biomedical EngineeringUniversity of Michigan 1011 N. University Ave, Room 2213 Ann Arbor MI 48109‐1078 USA
| |
Collapse
|
9
|
Huynh NPT, Brunger JM, Gloss CC, Moutos FT, Gersbach CA, Guilak F. Genetic Engineering of Mesenchymal Stem Cells for Differential Matrix Deposition on 3D Woven Scaffolds. Tissue Eng Part A 2018; 24:1531-1544. [PMID: 29756533 DOI: 10.1089/ten.tea.2017.0510] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Tissue engineering approaches for the repair of osteochondral defects using biomaterial scaffolds and stem cells have remained challenging due to the inherent complexities of inducing cartilage-like matrix and bone-like matrix within the same local environment. Members of the transforming growth factor β (TGFβ) family have been extensively utilized in the engineering of skeletal tissues, but have distinct effects on chondrogenic and osteogenic differentiation of progenitor cells. The goal of this study was to develop a method to direct human bone marrow-derived mesenchymal stem cells (MSCs) to deposit either mineralized matrix or a cartilaginous matrix rich in glycosaminoglycan and type II collagen within the same biochemical environment. This differential induction was performed by culturing cells on engineered three-dimensionally woven poly(ɛ-caprolactone) (PCL) scaffolds in a chondrogenic environment for cartilage-like matrix production while inhibiting TGFβ3 signaling through Mothers against DPP homolog 3 (SMAD3) knockdown, in combination with overexpressing RUNX2, to achieve mineralization. The highest levels of mineral deposition and alkaline phosphatase activity were observed on scaffolds with genetically engineered MSCs and exhibited a synergistic effect in response to SMAD3 knockdown and RUNX2 expression. Meanwhile, unmodified MSCs on PCL scaffolds exhibited accumulation of an extracellular matrix rich in glycosaminoglycan and type II collagen in the same biochemical environment. This ability to derive differential matrix deposition in a single culture condition opens new avenues for developing complex tissue replacements for chondral or osteochondral defects.
Collapse
Affiliation(s)
- Nguyen P T Huynh
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri.,3 Department of Cell Biology, Duke University , Durham, North Carolina
| | | | - Catherine C Gloss
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri
| | | | - Charles A Gersbach
- 6 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Farshid Guilak
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri.,5 Cytex Therapeutics, Inc. , Durham, North Carolina
| |
Collapse
|
10
|
Ishihara A, Weisbrode SE, Bertone AL. Autologous implantation of BMP2-expressing dermal fibroblasts to improve bone mineral density and architecture in rabbit long bones. J Orthop Res 2015; 33:1455-65. [PMID: 25418909 PMCID: PMC4441610 DOI: 10.1002/jor.22791] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/17/2014] [Indexed: 02/04/2023]
Abstract
Cell-mediated gene therapy may treat bone fragility disorders. Dermal fibroblasts (DFb) may be an alternative cell source to stem cells for orthopedic gene therapy because of their rapid cell yield and excellent plasticity with bone morphogenetic protein-2 (BMP2) gene transduction. Autologous DFb or BMP2-expressing autologous DFb were administered in twelve rabbits by two delivery routes; a transcortical intra-medullar infusion into tibiae and delayed intra-osseous injection into femoral drill defects. Both delivery methods of DFb-BMP2 resulted in a successful cell engraftment, increased bone volume, bone mineral density, improved trabecular bone microarchitecture, greater bone defect filling, external callus formation, and trabecular surface area, compared to non-transduced DFb or no cells. Cell engraftment within trabecular bone and bone marrow tissue was most efficiently achieved by intra-osseous injection of DFb-BMP2. Our results suggested that BMP2-expressing autologous DFb have enhanced efficiency of engraftment in target bones resulting in a measurable biologic response by the bone of improved bone mineral density and bone microarchitecture. These results support that autologous implantation of DFb-BMP2 warrants further study on animal models of bone fragility disorders, such as osteogenesis imperfecta and osteoporosis to potentially enhance bone quality, particularly along with other gene modification of these diseases.
Collapse
Affiliation(s)
- Akikazu Ishihara
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, Ohio
| | - Steve E Weisbrode
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - Alicia L Bertone
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, Ohio
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| |
Collapse
|
11
|
Needham CJ, Shah SR, Dahlin RL, Kinard LA, Lam J, Watson BM, Lu S, Kasper FK, Mikos AG. Osteochondral tissue regeneration through polymeric delivery of DNA encoding for the SOX trio and RUNX2. Acta Biomater 2014; 10:4103-12. [PMID: 24854956 DOI: 10.1016/j.actbio.2014.05.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 04/22/2014] [Accepted: 05/13/2014] [Indexed: 01/08/2023]
Abstract
Native osteochondral repair is often inadequate owing to the inherent properties of the tissue, and current clinical repair strategies can result in healing with a limited lifespan and donor site morbidity. This work investigates the use of polymeric gene therapy to address this problem by delivering DNA encoding for transcription factors complexed with the branched poly(ethylenimine)-hyaluronic acid (bPEI-HA) delivery vector via a porous oligo[poly(ethylene glycol) fumarate] hydrogel scaffold. To evaluate the potential of this approach, a bilayered scaffold mimicking native osteochondral tissue organization was loaded with DNA/bPEI-HA complexes. Next, bilayered implants either unloaded or loaded in a spatial fashion with bPEI-HA and DNA encoding for either Runt-related transcription factor 2 (RUNX2) or SRY (sex determining region Y)-box 5, 6, and 9 (the SOX trio), to generate bone and cartilage tissues respectively, were fabricated and implanted in a rat osteochondral defect. At 6weeks post-implantation, micro-computed tomography analysis and histological scoring were performed on the explants to evaluate the quality and quantity of tissue repair in each group. The incorporation of DNA encoding for RUNX2 in the bone layer of these scaffolds significantly increased bone growth. Additionally, a spatially loaded combination of RUNX2 and SOX trio DNA loading significantly improved healing relative to empty hydrogels or either factor alone. Finally, the results of this study suggest that subchondral bone formation is necessary for correct cartilage healing.
Collapse
|
12
|
Zhang X, Xu M, Song L, Wei Y, Lin Y, Liu W, Heng BC, Peng H, Wang Y, Deng X. Effects of compatibility of deproteinized antler cancellous bone with various bioactive factors on their osteogenic potential. Biomaterials 2013; 34:9103-14. [DOI: 10.1016/j.biomaterials.2013.08.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 08/12/2013] [Indexed: 11/26/2022]
|
13
|
Zhang X, Liu T, Huang Y, Wismeijer D, Liu Y. Icariin: Does It Have An Osteoinductive Potential for Bone Tissue Engineering? Phytother Res 2013; 28:498-509. [DOI: 10.1002/ptr.5027] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 04/07/2013] [Accepted: 05/21/2013] [Indexed: 01/23/2023]
Affiliation(s)
- Xin Zhang
- School of Stomatology; Tongji University; Shanghai China
| | - Tie Liu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Research Institute MOVE; University of Amsterdam and VU University Amsterdam; Gustav Mahlerlaan 3004 1081 LA Amsterdam the Netherlands
| | - Yuanliang Huang
- Department of Dentistry; Shanghai East Hospital Affiliated to Tongji University; Shanghai China
| | - Daniel Wismeijer
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Research Institute MOVE; University of Amsterdam and VU University Amsterdam; Gustav Mahlerlaan 3004 1081 LA Amsterdam the Netherlands
| | - Yuelian Liu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Research Institute MOVE; University of Amsterdam and VU University Amsterdam; Gustav Mahlerlaan 3004 1081 LA Amsterdam the Netherlands
| |
Collapse
|
14
|
Liu TM, Lee EH. Transcriptional regulatory cascades in Runx2-dependent bone development. TISSUE ENGINEERING PART B-REVIEWS 2012; 19:254-63. [PMID: 23150948 DOI: 10.1089/ten.teb.2012.0527] [Citation(s) in RCA: 253] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of the musculoskeletal system is a complex process that involves very precise control of bone formation and growth as well as remodeling during postnatal life. Although the understanding of the transcriptional mechanisms of osteogenesis has increased considerably, the molecular regulatory basis, especially the gene regulatory network of osteogenic differentiation, is still poorly understood. This review provides the reader with an overview of the key transcription factors that govern bone formation, highlighting their function and regulation linked to Runt-related transcription factor 2 (Runx2). Runx2 as the master transcription factor of osteoblast differentiation, Twist, Msh homeobox 2 (Msx2), and promyelocytic leukemia zinc-finger protein (PLZF) acting upstream of Runx2, Osterix (Osx) acting downstream of Runx2, and activating transcription factor 4 (ATF4) and zinc-finger protein 521 (ZFP521) acting as cofactors of Runx2 are discussed, and their relevance for tissue engineering is presented. References are provided for more in-depth personal study.
Collapse
Affiliation(s)
- Tong Ming Liu
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore.
| | | |
Collapse
|
15
|
Dalle Carbonare L, Innamorati G, Valenti MT. Transcription factor Runx2 and its application to bone tissue engineering. Stem Cell Rev Rep 2012; 8:891-7. [PMID: 22139789 DOI: 10.1007/s12015-011-9337-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cbfa1/Runx2 is a bone transcription factor homologous to the Drosophila protein, Runt. Runx2 is a master gene that encodes for a protein involved in the osteogenic differentiation process from mesenchymal precursors. It is known that in Cbfa1 deficient mice (Cbfa1(-/-)) the lack of mature osteoblasts is associated to incomplete bone mineralization. An important aim of modern biology is the development of new molecular tools for identification of therapeutic approaches. Recent discoveries in cell and molecular biology enabled researchers in the bone tissue-engineering field to develop new strategies for gene and cell-based therapies. This review summarizes the process of osteogenic differentiation from mesenchymal stem cells and the importance of bone regeneration is discussed. In particular, given the increasing interest in the study of the transcription factor Runx2, this review highlights the role of this target gene and addresses recent strategies using Runx2 for bone regeneration.
Collapse
Affiliation(s)
- Luca Dalle Carbonare
- Department of Medicine, Clinic of Internal Medicine, section D, University of Verona, Piazzale Scuro, 10, 37134 Verona, Italy
| | | | | |
Collapse
|
16
|
Cai M, Li G, Tao K, Yang Y, Lou L, Cai Z, Yu Y. Maohuoside A Acts in a BMP-dependent Manner during Osteogenesis. Phytother Res 2012; 27:1179-84. [PMID: 23007945 DOI: 10.1002/ptr.4840] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 08/09/2012] [Accepted: 08/15/2012] [Indexed: 11/06/2022]
Affiliation(s)
| | - Guodong Li
- Shanghai Tenth People's Hospital of Tongji University; Shanghai 200027 PR China
| | - Kun Tao
- Shanghai Tenth People's Hospital of Tongji University; Shanghai 200027 PR China
| | - Yunji Yang
- Shanghai Tenth People's Hospital of Tongji University; Shanghai 200027 PR China
| | - Lieming Lou
- Shanghai Tenth People's Hospital of Tongji University; Shanghai 200027 PR China
| | - Zhengdong Cai
- Shanghai Tenth People's Hospital of Tongji University; Shanghai 200027 PR China
| | - Yongchun Yu
- Shanghai Tenth People's Hospital of Tongji University; Shanghai 200027 PR China
| |
Collapse
|
17
|
Gaharwar AK, Kishore V, Rivera C, Bullock W, Wu CJ, Akkus O, Schmidt G. Physically crosslinked nanocomposites from silicate-crosslinked PEO: mechanical properties and osteogenic differentiation of human mesenchymal stem cells. Macromol Biosci 2012; 12:779-93. [PMID: 22517665 DOI: 10.1002/mabi.201100508] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 01/30/2012] [Indexed: 01/21/2023]
Abstract
The mechanical and biological properties of silicate-crosslinked PEO nanocomposites are studied. A strong correlation is observed between silicate concentration and mechanical properties. In vitro cell culture studies reveal that an increase in silicate concentration enhances the attachment and proliferation of human mesenchymal stem cells significantly. An upregulation in the expression of osteocalcin on nanocomposites compared to the tissue culture polystyrene control is observed. Together, these results suggest that silicate-based nanocomposites are bioactive and have the potential to be used in a range of biotechnological and biomedical applications such as injectable matrices, biomedical coatings, drug delivery, and regenerative medicine.
Collapse
Affiliation(s)
- Akhilesh K Gaharwar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907-2032, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Lai QG, Yuan KF, Xu X, Li DR, Li GJ, Wei FL, Yang ZJ, Luo SL, Tang XP, Li S. Transcription factor osterix modified bone marrow mesenchymal stem cells enhance callus formation during distraction osteogenesis. ORAL SURGERY, ORAL MEDICINE, ORAL PATHOLOGY, ORAL RADIOLOGY, AND ENDODONTICS 2011; 111:412-9. [PMID: 20813560 DOI: 10.1016/j.tripleo.2010.05.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 05/08/2010] [Accepted: 05/13/2010] [Indexed: 01/30/2023]
Abstract
This study was designed to investigate the effects of local delivery of bone marrow mesenchymal stem cells (BMMSCs) with or without osterix (OSX) gene transfected on bone regeneration in the distracted zone using a rabbit model of mandibular lengthening. Fifty-four New Zealand white rabbits underwent osteodistraction of the left mandible and were then randomly divided into group A, group B, and group C (n = 18 for each group). At the end of distraction BMMSCs transfected with OSX, autologous BMMSCs and physiological saline were injected into the distraction gaps in groups A, B, and C, respectively. Nine animals from each group were humanely killed at 2 and 6 weeks after completion of distraction. The distracted mandibles were harvested and processed for radiographic, histological, and immunohistochemical examination. Excellent bone formation in the distracted callus was observed in group A and group B; the former showed better bone formation and highest bone mineral density (BMD), thickness of new trabeculae (TNT, mm) and volumes of the newly formed bone area (NBV) in the distraction zones. Group C animals showed poor bone formation in the distracted callus when compared with groups A and B. Positive immunostaining of bone sialoprotein (BSP) was observed in the distracted callus in all groups; however, BSP expression was much stronger in group A than in groups B and C. The results of this study suggest transplantation of BMMSCs can promote bone formation in DO; OSX-mediated ex vivo gene therapy was more effective during bone deposition and callus formation in distraction osteogenesis.
Collapse
Affiliation(s)
- Qing-Guo Lai
- Department of Oral and Maxillofacial Surgery, Second Hospital of Shandong University, Jinan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kolambkar YM, Peister A, Ekaputra AK, Hutmacher DW, Guldberg RE. Colonization and osteogenic differentiation of different stem cell sources on electrospun nanofiber meshes. Tissue Eng Part A 2011; 16:3219-30. [PMID: 20504075 DOI: 10.1089/ten.tea.2010.0004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Numerous challenges remain in the successful clinical translation of cell-based therapies for musculoskeletal tissue repair, including the identification of an appropriate cell source and a viable cell delivery system. The aim of this study was to investigate the attachment, colonization, and osteogenic differentiation of two stem cell types, human mesenchymal stem cells (hMSCs) and human amniotic fluid stem (hAFS) cells, on electrospun nanofiber meshes. We demonstrate that nanofiber meshes are able to support these cell functions robustly, with both cell types demonstrating strong osteogenic potential. Differences in the kinetics of osteogenic differentiation were observed between hMSCs and hAFS cells, with the hAFS cells displaying a delayed alkaline phosphatase peak, but elevated mineral deposition, compared to hMSCs. We also compared the cell behavior on nanofiber meshes to that on tissue culture plastic, and observed that there is delayed initial attachment and proliferation on meshes, but enhanced mineralization at a later time point. Finally, cell-seeded nanofiber meshes were found to be effective in colonizing three-dimensional scaffolds in an in vitro system. This study provides support for the use of the nanofiber mesh as a model surface for cell culture in vitro, and a cell delivery vehicle for the repair of bone defects in vivo.
Collapse
Affiliation(s)
- Yash M Kolambkar
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | | | | | |
Collapse
|
21
|
Wojtowicz AM, Templeman KL, Hutmacher DW, Guldberg RE, García AJ. Runx2 overexpression in bone marrow stromal cells accelerates bone formation in critical-sized femoral defects. Tissue Eng Part A 2010; 16:2795-808. [PMID: 20412027 DOI: 10.1089/ten.tea.2010.0025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The repair of large nonunions in long bones remains a significant clinical problem due to high failure rates and limited tissue availability for auto- and allografts. Many cell-based strategies for healing bone defects deliver bone marrow stromal cells (BMSCs) to the defect site to take advantage of the inherent osteogenic capacity of this cell type. However, many factors, including donor age and ex vivo expansion of the cells, cause BMSCs to lose their differentiation ability. To overcome these limitations, we have genetically engineered BMSCs to constitutively overexpress the osteoblast-specific transcription factor Runx2. In the present study, we examined Runx2-modified BMSCs, delivered via polycaprolactone scaffolds loaded with type I collagen meshes, in critical-sized segmental defects in rats compared to unmodified cells, cell-free scaffolds, and empty defects. Runx2 expression in BMSCs accelerated healing of critical-sized defects compared to unmodified BMSCs and defects receiving cell-free treatments. These findings provide an accelerated method for healing large bone defects, which may reduce recovery time and the need for external fixation of critical-sized defects.
Collapse
Affiliation(s)
- Abigail M Wojtowicz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | | | | | | | | |
Collapse
|
22
|
Elliott NT, Yuan F. A review of three-dimensional in vitro tissue models for drug discovery and transport studies. J Pharm Sci 2010; 100:59-74. [PMID: 20533556 DOI: 10.1002/jps.22257] [Citation(s) in RCA: 326] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 05/04/2010] [Indexed: 12/12/2022]
Abstract
The use of animal models in drug discovery studies presents issues with feasibility and ethical concerns. To address these limitations, in vitro tissue models have been developed to provide a means for systematic, repetitive, and quantitative investigation of drugs. By eliminating or reducing the need for animal subjects, these models can serve as platforms for more tightly controlled, high-throughput screening of drugs and for pharmacokinetic and pharmacodynamic analyses of drugs. The focus of this review is three-dimensional (3D) tissue models that can capture cell-cell and cell-matrix interactions. Compared to the 2D culture of cell monolayers, 3D models more closely mimic native tissues since the cellular microenvironment established in the 3D models often plays a significant role in disease progression and cellular responses to drugs. A growing body of research has been published in the literature, which highlights the benefits of the 3D in vitro models of various tissues. This review provides an overview of some successful 3D in vitro models that have been developed to mimic liver, breast, cardiac, muscle, bone, and corneal tissues as well as malignant tissues in solid tumors.
Collapse
Affiliation(s)
- Nelita T Elliott
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, PO Box 90281, Durham, North Carolina 27708, USA
| | | |
Collapse
|
23
|
Lee SJ, Kang SW, Do HJ, Han I, Shin DA, Kim JH, Lee SH. Enhancement of bone regeneration by gene delivery of BMP2/Runx2 bicistronic vector into adipose-derived stromal cells. Biomaterials 2010; 31:5652-9. [PMID: 20413153 DOI: 10.1016/j.biomaterials.2010.03.019] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 03/09/2010] [Indexed: 02/07/2023]
Abstract
Adipose tissue contains multipotent mesenchymal stem cells (MSCs) that are able to differentiate into various tissues. Bone morphogenetic protein 2 (BMP2) is known as one of the key osteogenesis induction factors in MSCs. Recently, several new transcription factors that contribute to osteogenic differentiation have been reported, among them Runx2, Osterix, and Dlx5. We hypothesized that adipose-derived stromal cells (ASCs) could be induced to efficiently differentiate into osteocytes by the co-expression of the BMP2 and Runx2 genes. To prove this hypothesis, we constructed a bicistronic vector encoding the BMP2 and Runx2 genes linked to the 'self-cleaving' 2A peptide sequence. BMP2/Runx2-ASCs showed a gradual increase in alkaline phosphatase activity for two weeks. RT-PCR analysis and alizarin red staining revealed a high expression of osteogenesis-related markers (osteopontin, osteocalcin and collagen type I) and increased mineralization in BMP2/Runx2-ASCs compared to BMP2-ASCs. Six weeks after in vivo transplantation, BMP2/Runx2-ASCs also showed a significant increase in bone formation compared to ASCs and BMP2-ASCs. These findings demonstrate that the co-transfection of two osteogenic lineage-determining genes can enhance osteogenic differentiation of ASCs.
Collapse
Affiliation(s)
- Suk-Jun Lee
- Department of Medical Bioscience, CHA University, 606-16 Yeoksam 1-dong, Gangnam-gu, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
24
|
Nakajima K, Komiyama Y, Hojo H, Ohba S, Yano F, Nishikawa N, Ihara S, Aburatani H, Takato T, Chung UI. Enhancement of bone formation ex vivo and in vivo by a helioxanthin-derivative. Biochem Biophys Res Commun 2010; 395:502-8. [PMID: 20382113 DOI: 10.1016/j.bbrc.2010.04.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 04/06/2010] [Indexed: 11/27/2022]
Abstract
To effectively treat serious bone defects using bone-regenerative medicine, a small chemical compound that potently induces bone formation must be developed. We previously reported on the osteogenic effect of 4-(4-methoxyphenyl)pyrido[40,30:4,5]thieno[2,3-b]pyridine-2-carboxamide (TH), a helioxanthin-derivative, in vitro. Here, we report on TH's osteogenic effects ex vivo and in vivo. TH-induced new bone formation in both calvarial and metatarsal organ cultures. A novel monitoring system of osteoblastic differentiation using MC3T3-E1 cells revealed that TH was released from alpha-TCP bone cement and this release continued for more than one month. Lastly, the implantation of the alpha-TCP carrier containing TH into defects in mouse skull resulted in increased new bone areas within the defects after 4 weeks. A TH-containing scaffold may help establish a more efficient bone regeneration system.
Collapse
Affiliation(s)
- Keiji Nakajima
- Division of Sensory and Motor System Medicine, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhao J, Ohba S, Komiyama Y, Shinkai M, Chung UI, Nagamune T. Icariin: a potential osteoinductive compound for bone tissue engineering. Tissue Eng Part A 2010; 16:233-43. [PMID: 19698057 DOI: 10.1089/ten.tea.2009.0165] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To effectively treat bone diseases using bone regenerative medicine, there is an urgent need to develop safe and cheap drugs that can potently induce bone formation. Here, we demonstrate the osteogenic effects of icariin, the main active compound of Epimedium pubescens. Icariin induced osteogenic differentiation of preosteoblastic cells. The combination of icariin and a helioxanthin-derived small compound synergistically induced osteogenic differentiation of MC3T3-E1 cells to a similar extent to bone morphogenetic protein-2. Icariin enhanced the osteogenic induction activity of bone morphogenetic protein-2 in a fibroblastic cell line. Mineralization was enhanced by treatment with a combination of icariin and calcium-enriched medium. The in vivo anabolic effect of icariin was confirmed in a mouse calvarial defect model. Eight-week-old male C57BL/6N mice were transplanted with icariin-calcium phosphate cement (CPC) tablets or CPC tablets only (n = 5 for each), and bone regeneration was evaluated after 4 and 6 weeks. Significant new bone formation was observed in the icariin-CPC group at 4 weeks, and the new bone thickness had increased by 6 weeks. Obvious blood vessel formation was observed in the icariin-induced new bone. Treatment of senescence-accelerated mouse prone 1 and senescence-accelerated mouse prone 6 models further demonstrated that icariin was able to enhance bone formation in vivo. Therefore, icariin is a strong candidate for an osteogenic compound for use in bone tissue engineering.
Collapse
Affiliation(s)
- Jiyuan Zhao
- Department of Chemistry and Biotechnology, School of Engineering, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
26
|
El Tamer MK, Reis RL. Progenitor and stem cells for bone and cartilage regeneration. J Tissue Eng Regen Med 2009; 3:327-37. [PMID: 19418440 DOI: 10.1002/term.173] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Research in regenerative medicine is developing at a significantly quick pace. Cell-based bone and cartilage replacement is an evolving therapy aiming at the treatment of patients who suffer from limb amputation, damaged tissues and various bone and cartilage-related disorders. Stem cells are undifferentiated cells with the capability to regenerate into one or more committed cell lineages. Stem cells isolated from multiple sources have been finding widespread use to advance the field of tissue repair. The present review gives a comprehensive overview of the developments in stem cells originating from different tissues and suggests future prospects for functional bone and cartilage tissue regeneration.
Collapse
Affiliation(s)
- M K El Tamer
- 3Bs Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal.
| | | |
Collapse
|
27
|
|
28
|
Spalazzi JP, Dagher E, Doty SB, Guo XE, Rodeo SA, Lu HH. In vivo evaluation of a multiphased scaffold designed for orthopaedic interface tissue engineering and soft tissue-to-bone integration. J Biomed Mater Res A 2008; 86:1-12. [PMID: 18442111 DOI: 10.1002/jbm.a.32073] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Achieving functional graft integration with subchondral bone poses a significant challenge for orthopaedic soft tissue repair and reconstruction. Soft tissues such as the anterior cruciate ligament (ACL) integrate with bone through a fibrocartilage interface, which minimizes stress concentrations and mediates load transfer between soft and hard tissues. We propose that biological fixation can be achieved by regenerating this fibrocartilage interface on biological or synthetic ACL grafts. This study focuses on the in vivo evaluation of a stratified scaffold predesigned to mimic the multitissue transition found at the ACL-to-bone interface. Specifically, the scaffold consists of three distinct yet continuous phases: Phase A for ligament formation, Phase B for the interface, and Phase C for the bone region. Interface-relevant cell types, specifically fibroblasts, chondrocytes, and osteoblasts, will be tri-cultured on this scaffold, and the formation of cell type- and phase-specific matrix heterogeneity as well as fibrocartilage formation will be evaluated over 8 weeks in a subcutaneous athymic rat model. Acellular scaffolds as well as scaffolds co-cultured with fibroblasts and osteoblasts will serve as controls. It was found that the triphasic scaffold supported multilineage cellular interactions as well as tissue infiltration and abundant matrix production in vivo. In addition, controlled phase-specific matrix heterogeneity was induced on the scaffold, with distinct mineral and fibrocartilage-like tissue regions formed in the tri-cultured group. Cell seeding had a positive effect on both host infiltration and matrix elaboration, which also translated into increased mechanical properties in the seeded groups compared to the acellular controls. In summary, the biomimetic and multiphasic design coupled with spatial control of cell distribution enables multitissue regeneration on the stratified scaffold, and demonstrates the potential for regenerating the interface between soft tissue grafts and bone.
Collapse
Affiliation(s)
- Jeffrey P Spalazzi
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, New York 10027, USA
| | | | | | | | | | | |
Collapse
|
29
|
Development of high-throughput screening system for osteogenic drugs using a cell-based sensor. Biochem Biophys Res Commun 2008; 376:375-9. [PMID: 18789892 DOI: 10.1016/j.bbrc.2008.08.167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Accepted: 08/29/2008] [Indexed: 02/08/2023]
Abstract
To effectively treat osteoporosis and other bone-loss disorders, small compounds that potently induce bone formation are needed. The present study initially attempted to establish a monitoring system that could detect osteogenic differentiation easily, precisely, and noninvasively. For this purpose, we established pre-osteoblastic MC3T3E1 cells stably transfected with the GFP reporter gene driven by a 2.3 kb fragment of rat type I collagen promoter (Col1a1GFP-MC3T3E1). Among these cells, we selected a clone that fluoresced upon osteogenic stimulation by BMP2. The GFP fluorescence intensity corresponded well to the intensity of alkaline phosphatase (ALP) staining and to the level of osteocalcin (Oc) mRNA. Using this system, we screened natural and synthetic compound libraries and thus identified an isoflavone derivative, glabrisoflavone (GI). GI induced ALP staining and Oc mRNA in a dose-dependent manner. The Col1a1GFP-MC3T3E1 system may be useful for identifying novel osteogenic drugs.
Collapse
|
30
|
Phillips JE, García AJ. Retroviral-mediated gene therapy for the differentiation of primary cells into a mineralizing osteoblastic phenotype. Methods Mol Biol 2008; 433:333-54. [PMID: 18679633 DOI: 10.1007/978-1-59745-237-3_20] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Bone tissue engineering has emerged as a promising strategy for the repair of critical-sized skeletal fractures. However, the clinical application of this approach has been limited by the availability of a robust mineralizing cell source. Non-osteogenic cells, such as skin fibroblasts, are an attractive cell-source alternative because they are easy to harvest from autologous donor skin biopsies and display a high capacity for in vitro expansion. We have recently demonstrated that retroviral gene delivery of the osteoblastic transcription factor Runx2/Cbfa1 promotes osteogenic differentiation in primary dermal fibroblasts cultured in monolayer. Notably, sustained expression of Runx2 was not sufficient to promote functional osteogenesis in these cells, and co-treatment with the steroid hormone dexamethasone was required to induce deposition of biologically-equivalent matrix mineralization. On the basis of these results, we then investigated the osteogenic capacity of these genetically engineered fibroblasts when seeded on polymeric scaffolds in vitro and in vivo. These experiments demonstrated that Runx2-expressing fibroblasts seeded on collagen scaffolds produce significant levels of matrix mineralization after 28 days in vivo implantation in a subcutaneous, heterotopic site. Overall, these results offer evidence that transcription factor-based gene therapy may be a powerful strategy for the conversion of a non-osteogenic cellular phenotype into a mineralizing cell source for bone repair applications. This concept may also be applied to control functional differentiation in a broad range of cell types and tissue engineering applications. The chapter below outlines detailed methods for the isolation and ex vivo genetic modification of primary dermal fibroblasts using retroviral-mediated delivery of the Runx2 transgene in both monolayer culture and three-dimensional scaffolds.
Collapse
Affiliation(s)
- Jennifer E Phillips
- Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
31
|
Zhao J, Ohba S, Shinkai M, Chung UI, Nagamune T. Icariin induces osteogenic differentiation in vitro in a BMP- and Runx2-dependent manner. Biochem Biophys Res Commun 2008; 369:444-8. [PMID: 18295595 DOI: 10.1016/j.bbrc.2008.02.054] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 02/09/2008] [Indexed: 01/13/2023]
Abstract
To effectively treat bone diseases using bone regenerative medicine, there is an urgent need to develop safe cheap drugs that can potently induce bone formation. Here, we demonstrate the osteogenic effect of icariin, the main active compound of Epimedium pubescens. Icariin induced osteogenic differentiation in pre-osteoblastic MC3T3-E1 cells and mouse primary osteoblasts. Icariin upregulated the mRNA expression levels of the osteoblast marker genes runt-related transcription factor 2 (Runx2) and inhibitor of DNA-binding 1 (Id-1). The osteogenic effect was inhibited by the introduction of Smad6 or dominant-negative Runx2, as well as Noggin treatment. Furthermore, icariin induced the mRNA expression of bone morphogenetic protein (BMP)-4. These data suggest that icariin exerts its potent osteogenic effect through induction of Runx2 expression, production of BMP-4 and activation of BMP signaling. The extremely low cost of icariin and its high abundance make it appealing for bone regenerative medicine.
Collapse
Affiliation(s)
- Jiyuan Zhao
- Department of Chemistry and Biotechnology, School of Engineering, University of Tokyo, Tokyo 113-8656, Japan
| | | | | | | | | |
Collapse
|
32
|
Perfusion–compression bioreactor as the optimum choice for growing large-sized engineered bone constructs in vitro. ACTA ACUST UNITED AC 2008. [DOI: 10.1016/j.bihy.2008.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
33
|
Bhat BM, Robinson JA, Coleburn VE, Zhao W, Kharode Y. Evidence of in vivo osteoinduction in adult rat bone by adeno-Runx2 intra-femoral delivery. J Cell Biochem 2008; 103:1912-24. [DOI: 10.1002/jcb.21581] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
34
|
Abstract
The clinical challenges of skeletal regenerative medicine have motivated significant advances in cellular and tissue engineering in recent years. In particular, advances in molecular biology have provided the tools necessary for the design of gene-based strategies for skeletal tissue repair. Consequently, genetic engineering has emerged as a promising method to address the need for sustained and robust cellular differentiation and extracellular matrix production. As a result, gene therapy has been established as a conventional approach to enhance cellular activities for skeletal tissue repair. Recent literature clearly demonstrates that genetic engineering is a principal factor in constructing effective methods for tissue engineering approaches to bone, cartilage, and connective tissue regeneration. This review highlights this literature, including advances in the development of efficacious gene carriers, novel cell sources, successful delivery strategies, and optimal target genes. The current status of the field and the challenges impeding the clinical realization of these approaches are also discussed.
Collapse
Affiliation(s)
- Charles A Gersbach
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | |
Collapse
|
35
|
Phillips JE, Guldberg RE, García AJ. Dermal fibroblasts genetically modified to express Runx2/Cbfa1 as a mineralizing cell source for bone tissue engineering. ACTA ACUST UNITED AC 2007; 13:2029-40. [PMID: 17516856 DOI: 10.1089/ten.2006.0041] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cell-based bone tissue engineering strategies have been effectively applied toward the development of grafting templates for skeletal repair and regeneration, but remain limited by inadequate availability of a robust mineralizing cell source. Dermal fibroblasts have emerged as a particularly promising cell alternative because they are harvested from autologous donors through minimally invasive skin biopsy and display a high capacity for in vitro expansion. In the present study, we investigated retroviral gene delivery of the osteogenic transcription factor Runx2 as a mineralization induction strategy in primary dermal fibroblasts. We demonstrate that constitutive overexpression of Runx2 induced osteogenic gene expression and mineralized nodule deposition in fibroblasts cultured on 3-dimensional fibrous collagen disks in vitro. Fourier transform infrared analysis revealed that Runx2 expressing fibroblasts deposit a carbonate-containing, poorly crystalline hydroxyapatite, whereas control constructs did not contain biologically-equivalent mineral. Importantly, Runx2-transduced fibroblasts formed mineralized templates in vivo after implantation in a subcutaneous, heterotopic site, whereas minimal mineralization was evident in control constructs. Furthermore, immunohistochemical analysis indicated that Runx2-engineered cells co-localized with mineral deposits in vivo, suggesting that nodule formation primarily originated from transplanted donor cells. These results establish Runx2-genetic engineering as a strategy for the conversion of a non-osteogenic cellular phenotype into a mineralizing cell source for bone repair applications. Cellular therapies based on primary dermal fibroblasts would be particularly beneficial for patients with compromised ability to recruit endogenous osteoprogenitors to the site of injury as a result of extreme trauma, age, radiation treatment, or osteolytic disease.
Collapse
Affiliation(s)
- Jennifer E Phillips
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
36
|
Abstract
Aiming for regeneration of severed or lost parts of the body, the combined application of gene therapy and tissue engineering has received much attention by regenerative medicine. Techniques of molecular biology can enhance the regenerative potential of a biomaterial by co-delivery of therapeutic genes, and several different strategies have been used to achieve that goal. Possibilities for application are many-fold and have been investigated to regenerate tissues such as skin, cartilage, bone, nerve, liver, pancreas and blood vessels. This review discusses advantages and problems encountered with the different gene delivery strategies as far as they relate to tissue engineering, analyses the positive aspects of polymeric gene delivery from matrices and discusses advances and future challenges of gene transfer strategies in selected tissues.
Collapse
Affiliation(s)
- Oliver Bleiziffer
- Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Erlangen, Germany
- *Correspondence to: Ulrich KNESER, M.D. Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Krankenhausstr. 12, 91054 Erlangen, Germany. Tel.: +49-9131-85-33277; Fax: +49-9131-85-39327 E-mail:
| | - Elof Eriksson
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Feng Yao
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Erlangen, Germany
| | - Ulrich Kneser
- Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Erlangen, Germany
- *Correspondence to: Ulrich KNESER, M.D. Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Krankenhausstr. 12, 91054 Erlangen, Germany. Tel.: +49-9131-85-33277; Fax: +49-9131-85-39327 E-mail:
| |
Collapse
|
37
|
Gersbach CA, Guldberg RE, García AJ. In vitro and in vivo osteoblastic differentiation of BMP-2- and Runx2-engineered skeletal myoblasts. J Cell Biochem 2007; 100:1324-36. [PMID: 17131362 DOI: 10.1002/jcb.21118] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Genetic engineering with osteogenic factors is a promising approach for cell-based therapeutics and orthopedic regeneration. However, the relative efficacy of different strategies for inducing osteoblastic differentiation remains unclear and is further complicated by varied delivery vehicles, cell types, and evaluation criteria. In order to elucidate the effects of distinct gene-based strategies, we quantitatively evaluated osteoblastic differentiation and mineralization of primary skeletal myoblasts overexpressing either the BMP-2 growth factor or Runx2 transcription factor. Retroviral delivery of BMP-2 or Runx2 stimulated differentiation into an osteoblastic phenotype, as demonstrated by the induction of osteogenic gene expression, alkaline phosphatase activity, and matrix mineralization in monolayer culture and on collagen scaffolds both in vitro and in an intramuscular site in vivo. In general, BMP-2 stimulated osteoblastic markers faster and to a greater extent than Runx2, although we also identified experimental conditions under which these two factors produced similar effects. Additionally, Runx2-engineered cells did not utilize paracrine signaling via secreted osteogenic factors, in contrast to cells overexpressing BMP-2, as demonstrated by conditioned media studies and activation of Smad signaling. These results emphasize the complexity of gene therapy-based orthopedic therapeutics as an integrated relationship of differentiation state, construct maturation, and paracrine signaling of osteogenic cells. This study is significant in evaluating proposed therapeutic systems and defining a successful strategy for integrating gene medicine and orthopedic regeneration.
Collapse
Affiliation(s)
- Charles A Gersbach
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | | | | |
Collapse
|
38
|
Ohba S, Nakajima K, Komiyama Y, Kugimiya F, Igawa K, Itaka K, Moro T, Nakamura K, Kawaguchi H, Takato T, Chung UI. A novel osteogenic helioxanthin-derivative acts in a BMP-dependent manner. Biochem Biophys Res Commun 2007; 357:854-60. [PMID: 17451649 DOI: 10.1016/j.bbrc.2007.03.173] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2007] [Accepted: 03/28/2007] [Indexed: 10/23/2022]
Abstract
To effectively treat serious bone defects using bone regenerative medicine, there is a need for the development of a small chemical compound that potently induces bone formation. We now report a novel osteogenic helioxanthin-derivative, TH. TH induced osteogenic differentiation in MC3T3-E1 cells, mouse primary osteoblasts, and mouse embryonic stem cells. The combination of TH and bone morphogenetic protein (BMP) 2 induced the mRNA expression of osteoblast marker genes and calcification in primary fibroblasts. The TH induced the mRNA of the inhibitor of DNA-binding 1 (Id-1), and its osteogenic effect was inhibited by Smad6 or Noggin. Furthermore, TH induced the mRNA expression of Bmp4 and Bmp6. These data suggest that TH exerts its potent osteogenic effect in a BMP-dependent manner by enhancing the effects of the existing BMPs and/or increasing the expression of Bmp4 and Bmp6. TH may help establish a more efficient bone regeneration system.
Collapse
Affiliation(s)
- Shinsuke Ohba
- Division of Sensory and Motor System Medicine, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Leong DT, Abraham MC, Rath SN, Lim TC, Chew FT, Hutmacher DW. Investigating the effects of preinduction on human adipose-derived precursor cells in an athymic rat model. Differentiation 2007; 74:519-29. [PMID: 17177849 DOI: 10.1111/j.1432-0436.2006.00092.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The osteogenic potential of human adipose-derived precursor cells seeded on medical-grade polycaprolactone-tricalcium phosphate scaffolds was investigated in this in vivo study. Three study groups were investigated: (1) induced--stimulated with osteogenic factors only after seeding into scaffold; (2) preinduced--induced for 2 weeks before seeding into scaffolds; and (3) uninduced--cells without any introduced induction. For all groups, scaffolds were implanted subcutaneously into the dorsum of athymic rats. The scaffold/cell constructs were harvested at the end of 6 or 12 weeks and analyzed for osteogenesis. Gross morphological examination using scanning electron microscopy indicated good integration of host tissue with scaffold/cell constructs and extensive tissue infiltration into the scaffold interior. Alizarin Red histology and immunostaining showed a heightened level of mineralization and an increase in osteonectin, osteopontin, and collagen type I protein expression in both the induced and preinduced groups compared with the uninduced groups. However, no significant differences were observed in these indicators when compared between the induced and preinduced groups.
Collapse
Affiliation(s)
- David Tai Leong
- Department of Biological Sciences, National University of Singapore, Singapore 117576, Republic of Singapore
| | | | | | | | | | | |
Collapse
|
40
|
Ohba S, Ikeda T, Kugimiya F, Yano F, Lichtler AC, Nakamura K, Takato T, Kawaguchi H, Chung UI. Identification of a potent combination of osteogenic genes for bone regeneration using embryonic stem (ES) cell‐based sensor. FASEB J 2007; 21:1777-87. [PMID: 17317722 DOI: 10.1096/fj.06-7571com] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To identify potent bioactive factors for in vivo tissue regeneration by comprehensive screening remains a challenge for regenerative medicine. Here we report the development of an ES cell-based monitoring system for osteogenic differentiation, the identification of a potent combination of osteogenic genes using such a system, and an evaluation of its therapeutic potentials. ES cells were isolated from mice carrying a transgene expressing GFP driven by the 2.3 kb fragment of rat type I collagen alpha1 promoter. Using these cells engineered to fluoresce on osteogenic differentiation, we screened cDNA libraries and combinations of major osteogenesis-related genes. Among them, the combination of constitutively active activin receptor-like kinase 6 (caALK6) and runt-related transcription factor 2 (Runx2) was the minimal unit that induced fluorescence. The combination efficiently induced osteogenic differentiation in various cell types, including terminally differentiated nonosteogenic cells. The cooperative action of the combination occurred through protein stabilization of core binding factor beta (Cbfb), induction of Runx2-Cbfb complex formation, and its DNA binding. Furthermore, transplantation of a monolayer sheet of fibroblasts transduced with the combination achieved bone regeneration within 4 wk in mouse calvarial bone defects. Thus, we successfully identified the potent combination of genes for bone regeneration, which helped broaden cell sources.
Collapse
Affiliation(s)
- Shinsuke Ohba
- Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Phillips JE, Gersbach CA, García AJ. Virus-based gene therapy strategies for bone regeneration. Biomaterials 2007; 28:211-29. [PMID: 16928397 DOI: 10.1016/j.biomaterials.2006.07.032] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Accepted: 07/18/2006] [Indexed: 12/31/2022]
Abstract
Gene therapy has emerged as a promising strategy for the repair and regeneration of damaged musculoskeletal tissues. Application of this paradigm to bone healing has shown enhanced efficacy in preclinical animal studies compared to conventional bone grafting approaches. This review discusses current and emerging virus-based genetic engineering strategies for the delivery of therapeutic molecules which promote skeletal regeneration. Viral gene delivery vectors are discussed in the context of bone repair in order to illustrate the challenges and applications of these methods with tissue-specific examples. Moreover the concepts discussed can be broadly applied to promote healing in a wide range of tissues. We also present important considerations involved in the application of these gene therapy techniques to a variety of osteogenic (e.g. bone marrow-derived cells) and non-osteogenic (e.g. fibroblasts and skeletal myoblasts) cell types. Criteria for the selection of regenerative molecules with soluble versus intracellular modes of action and emerging combinatorial approaches are also discussed. Overall, gene transfer technologies have the potential to overcome limitations associated with existing bone grafting approaches and may enable investigators to design therapies which more closely mimic the complex spatial and temporal cascade of proteins involved in endogenous bone development and repair.
Collapse
Affiliation(s)
- Jennifer E Phillips
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA.
| | | | | |
Collapse
|
42
|
Gersbach CA, Le Doux JM, Guldberg RE, García AJ. Inducible regulation of Runx2-stimulated osteogenesis. Gene Ther 2006; 13:873-82. [PMID: 16496016 DOI: 10.1038/sj.gt.3302725] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ex vivo gene therapy is a promising approach to orthopedic regenerative medicine. These strategies typically focus on the constitutive overexpression of osteogenic factors to induce osteoblastic differentiation and matrix mineralization. However, the unregulated production of osteoinductive molecules has also resulted in abnormal bone formation and tumorigenesis. To address these limitations, this work describes a retroviral system to deliver the Runx2 osteoblastic transcription factor under control of the tetracycline-inducible (tet-off) promoter in primary skeletal myoblasts. Runx2 expression was tightly regulated by anhydrotetracyline (aTc) concentration in cell culture media. Osteoblastic gene expression, alkaline phosphatase activity, and matrix mineralization were also controlled by aTc in a dose-dependent manner. Additionally, osteoblastic differentiation was temporally regulated by adding and removing aTc from the culture media. Engineered cells were seeded onto collagen scaffolds and implanted intramuscularly in the hind limbs of syngeneic mice. In vivo mineralization by these constructs was regulated by supplementing the drinking water with aTc, as demonstrated by micro-computed tomography and histological analyses. Collectively, these results present a novel system for regulating osteoblastic differentiation of a clinically relevant autologous cell source. This system is significant to developing controlled and effective orthopedic gene therapy strategies and studying the regulation of osteoblastic differentiation.
Collapse
Affiliation(s)
- C A Gersbach
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332, USA
| | | | | | | |
Collapse
|
43
|
Lian JB, Stein GS, Javed A, van Wijnen AJ, Stein JL, Montecino M, Hassan MQ, Gaur T, Lengner CJ, Young DW. Networks and hubs for the transcriptional control of osteoblastogenesis. Rev Endocr Metab Disord 2006; 7:1-16. [PMID: 17051438 DOI: 10.1007/s11154-006-9001-5] [Citation(s) in RCA: 340] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We present an overview of the concepts of tissue-specific transcriptional control mechanisms essential for development of the bone cell phenotype. BMP2 induced transcription factors constitute a network of activities and molecular switches for bone development and osteoblast differentiation. Among these regulators are Runx2 (Cbfa1/AML3), the principal osteogenic master gene for bone formation, as well as homeodomain proteins and osterix. Runx2 has multiple regulatory activities, including activation or repression of gene expression, and integration of biological signals from developmental cues, such as BMP/TGFbeta, Wnt and Src signaling pathways. Runx2 provides a new paradigm for transcriptional control by functioning as a principal scaffolding protein in nuclear microenvironments to control gene expression in response to physiologic signals (growth factors, cytokines and hormones). The protein serves as a hub for the coordination of activities essential for the expansion and differentiation of osteogenic lineage cells through the formation of co-regulatory protein complexes organized in subnuclear domains. Mechanisms by which Runx2 supports commitment to osteogenesis and determines cell fate involve its retention on mitotic chromosomes. Disruption of a unique protein module, the subnuclear targeting signal of Runx2, has profound effects on osteoblast differentiation and metastasis of cancer cells in the bone microenvironment. Runx2 target genes include regulators of cell growth control, components of the bone extracellular matrix, angiogenesis, and signaling proteins for development of the osteoblast phenotype and bone turnover. The specificity of Runx2 regulatory activities provides a basis for novel therapeutic strategies to correct bone disorders.
Collapse
Affiliation(s)
- Jane B Lian
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Byers BA, Guldberg RE, Hutmacher DW, García AJ. Effects of Runx2 genetic engineering and in vitro maturation of tissue-engineered constructs on the repair of critical size bone defects. J Biomed Mater Res A 2006; 76:646-55. [PMID: 16287095 DOI: 10.1002/jbm.a.30549] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Genetic and tissue engineering strategies are being pursued to address the clinical limitations of current bone grafting materials. Based on our previous work demonstrating that overexpression of the Runx2 osteoblastic transcription factor and in vitro construct maturation synergistically enhanced in vivo mineralization in an ectopic site (Byers et al., Tissue Eng 2004;10:1757-1766), we examined the effects of these two parameters on the repair of critical size bone defects. Primary rat bone marrow stromal cells transduced with Runx2 or control (no Runx2 insert) retroviral vector were seeded onto 3D fused deposition-modeled polycaprolactone scaffolds. Runx2-modified cells produced biologically-equivalent mineralized matrices at nearly 2-fold higher rates than control cells. Constructs cultured in vitro for 1 day (immature) or 21 days (mineralized) were subsequently implanted into critical size calvaria defects in syngeneic rats, and bone healing was analyzed by micro-CT and histomorphometry at 28 days. Runx2-modified and control constructs precultured for 1 day healed to a greater extent than defects receiving no implant. Cell-free scaffolds yielded equivalent levels of bone formation as constructs precultured for 1 day. Interestingly, defects treated with control cell-seeded constructs precultured for 21 days exhibited low bone formation compared to other construct treatments, and repair was comparable to empty defects. In contrast, Runx2-modified constructs precultured for 21 days contained twice as much bone as control constructs precultured for 21 days and equivalent levels of new bone as cell-free and 1 day precultured constructs. These results demonstrate interplay between Runx2 genetically-modified cells and in vitro construct maturation in bone healing responses.
Collapse
Affiliation(s)
- Benjamin A Byers
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Bone marrow stromal/stem cells (BMSCs) are multipotent adult stem cells and have become the important cell source for cell therapy and engineered tissue repair. Their osteogenic differentiation potential has been well characterized in many in vitro studies. In addition, small animal model-based studies also reveal their capability of bone formation in vivo when implanted with biodegradable scaffold, indicating the great potential for therapeutic application. Bone defect is a common clinical problem that deserves an optimal therapy. Unlike traditional surgical repair that needs to sacrifice donor site tissue, the tissue-engineering approach can achieve the goal of bone regeneration and repair without the necessity of donor site morbidity. To safely translate experimental study into a clinical trial of engineered bone repair, in vivo study using large animal models has become the key issue. Our in vivo study in this aspect and the published results indicate that bone regeneration and repair by BMSCs and biodegradable scaffold is a realistic goal that can be achieved.
Collapse
Affiliation(s)
- Wei Liu
- Department of Plastic Surgery, Shanghai 9th People's Hospital, Shanghai, People's Republic of China
| | | | | |
Collapse
|
46
|
|
47
|
Hutmacher DW, Garcia AJ. Scaffold-based bone engineering by using genetically modified cells. Gene 2005; 347:1-10. [PMID: 15777645 DOI: 10.1016/j.gene.2004.12.040] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2004] [Revised: 11/19/2004] [Accepted: 12/22/2004] [Indexed: 01/24/2023]
Abstract
The first generation of clinically applied tissue engineering concepts in the area of skin, cartilage and bone marrow regeneration was based on the isolation, expansion and implantation of cells from the patient's own tissue. Although successful in selective treatments, tissue engineering needs to overcome major challenges to allow widespread clinical application with predictable outcomes. One challenge is to present the cells in a matrix to the implantation site to allow the cells to survive the wound healing contraction forces, tissue remodeling in certain tissues such as bone and biomechanical loading. Hence, several tissue engineering strategies focus on the development of load-bearing scaffold/cell constructs. From a cell source point of view, bone engineers face challenges to isolate and expand cells with the highest potential to form osseous tissue along with harvesting tissue without extensive donor site morbidity. A major hurdle to tissue engineering is de-differentiation and limited ability to control cell phenotype following in vitro expansion. Due to early successes with genetic engineering, bone tissue engineers have used different strategies to genetically alter various types of mesenchymal cells to enhance the mineralization capacity of tissue-engineered scaffold/cell constructs. Although the development of multi-component scaffold/osteogenic cell constructs requires a combination of interdisciplinary research strategies, the following review is limited to describe the general aspects of bone engineering and to present overall directions of technology platforms, which include a genetic engineering component. This paper reviews the most recent work in the field and discusses the concepts developed and executed by a collaborative effort of the multi-disciplinary teams of the two authors.
Collapse
Affiliation(s)
- Dietmar W Hutmacher
- Division of Bioengineering, Faculty of Engineering, National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260, Singapore.
| | | |
Collapse
|