1
|
Mahmoudi N, Roque M, Paiva Dos Santos B, Oliveira H, Siadous R, Rey S, Garanger E, Lecommandoux S, Catros S, Garbay B, Amédée Vilamitjana J. An Elastin-Derived Composite Matrix for Enhanced Vascularized and Innervated Bone Tissue Reconstruction: From Material Development to Preclinical Evaluation. Adv Healthc Mater 2024; 13:e2303765. [PMID: 38651610 DOI: 10.1002/adhm.202303765] [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: 10/29/2023] [Revised: 03/28/2024] [Indexed: 04/25/2024]
Abstract
Despite progress in bone tissue engineering, reconstruction of large bone defects remains an important clinical challenge. Here, a biomaterial designed to recruit bone cells, endothelial cells, and neuronal fibers within the same matrix is developed, enabling bone tissue regeneration. The bioactive matrix is based on modified elastin-like polypeptides (ELPs) grafted with laminin-derived adhesion peptides IKVAV and YIGSR, and the SNA15 peptide for retention of hydroxyapatite (HA) particles. The composite matrix shows suitable porosity, interconnectivity, biocompatibility for endothelial cells, and the ability to support neurites outgrowth by sensory neurons. Subcutaneous implantation leads to the formation of osteoid tissue, characterized by the presence of bone cells, vascular networks, and neuronal structures, while minimizing inflammation. Using a rat femoral condyle defect model, longitudinal micro-CT analysis is performed, which demonstrates a significant increase in the volume of mineralized tissue when using the ELP-based matrix compared to empty defects and a commercially available control (Collapat). Furthermore, visible blood vessel networks and nerve fibers are observed within the lesions after a period of two weeks. By incorporating multiple key components that support cell growth, mineralization, and tissue integration, this ELP-based composite matrix provides a holistic and versatile solution to enhance bone tissue regeneration.
Collapse
Affiliation(s)
- Nadia Mahmoudi
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Micaela Roque
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Bruno Paiva Dos Santos
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Hugo Oliveira
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Robin Siadous
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Sylvie Rey
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | | | | | - Sylvain Catros
- CHU Bordeaux, Dentistry and Oral Health Department, Bordeaux, 33076, France
| | - Bertrand Garbay
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR, Pessac, 5629, France
| | | |
Collapse
|
2
|
Song Y, Li H, Wang Z, Shi J, Li J, Wang L, Liao L, Ma S, Zhang Y, Liu B, Yang Y, Zhou P. Define of Optimal Addition Period of Osteogenic Peptide to Accelerate the Osteogenic Differentiation of Human Pluripotent Stem Cells. Tissue Eng Regen Med 2024; 21:291-308. [PMID: 37903982 PMCID: PMC10825087 DOI: 10.1007/s13770-023-00597-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/15/2023] [Accepted: 09/05/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The addition of growth factiors is commonly applied to improve the osteogenic differentiation of stem cells. However, for human pluripotent stem cells (hPSCs), their complex differentiation processes result in the unknown effect at different stages. In this study, we focused on the widely used bone forming peptide-1 (BFP-1) and investigated the effect and mechanisms of its addition on the osteogenic induction of hPSCs as a function of the supplementation period. METHODS Monolayer-cultured hPSCs were cultured in osteogenic induction medium for 28 days, and the effect of BFP-1 peptide addition at varying weeks was examined. After differentiation for varying days (0, 7, 14, 21 and 28), the differentiation efficiency was determined by RT-PCR, flow cytometry, immunofluorescence, and alizarin red staining assays. Moreover, the expression of marker genes related to germ layers and epithelial-mesenchymal transition (EMT) was investigated at day 7. RESULTS Peptide treatment during the first week promoted the generation of mesoderm cells and mesenchymal-like cells from hiPSCs. Then, the upregulated expression of osteogenesis marker genes/proteins was detected in both hESCs and hiPSCs during subsequent inductions with BFP-1 peptide treatment. Fortunately, further experimental design confirmed that treating the BFP-1 peptide during 7-21 days showed even better performance for hESCs but was ineffective for hiPSCs. CONCLUSION The differentiation efficiency of cells could be improved by determining the optimal treatment period. Our study has great value in maximizing the differentiation of hPSCs by adding osteogenesis peptides based on the revealed mechanisms and promoting the application of hPSCs in bone tissue regeneration.
Collapse
Affiliation(s)
- Yameng Song
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Hongjiao Li
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Zixuan Wang
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jiamin Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jing Li
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Lu Wang
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Lingzi Liao
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Shengqin Ma
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Yun Zhang
- Lanzhou Hospital of Stomatology, Lanzhou, 730000, People's Republic of China
| | - Bin Liu
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China.
| | - Yaling Yang
- Lanzhou Hospital of Stomatology, Lanzhou, 730000, People's Republic of China.
| | - Ping Zhou
- School and Hospital of Stomatology, Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, People's Republic of China.
| |
Collapse
|
3
|
Tong L, Yang Z, Dai W, Sun Z, Yang J, Xue Q, Li Y. Experimental study on determining the degree of bone healing by wall thickness ratio analysis. J Orthop Surg Res 2024; 19:79. [PMID: 38243260 PMCID: PMC10799492 DOI: 10.1186/s13018-024-04565-7] [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: 11/24/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024] Open
Abstract
To verify the reliability and accuracy of wall thickness ratio analysis to determine the degree of bone healing, fracture models were established with 6 beagles. X-ray, micro-CT, and CT scans were performed at 24 weeks. The healthy side and the affected side were used to simulate the three-dimensional geometric model after internal fixation, and the mesh was divided. The mean and median CT wall thickness values were obtained through the wall thickness analysis. X-ray, CT, micro-CT, and gross appearance were used to determine the degree of bone healing, which was compared with wall thickness analysis. There was a positive correlation between the average CT value and the median wall thickness. The correlation coefficient analysis of the median wall thickness ratio (R2) and healing index ratio (R3) showed a positive correlation. The results of the wall thickness ratio (R2) and the healing index ratio (R3) were used to determine bone healing, and the results were consistent with the results of the actual mechanical test and image analysis. The results of wall thickness ratio analysis were significantly correlated with the degree of bone healing. This method is simple, rapid, and practical to analyze and judge the degree of bone healing.
Collapse
Affiliation(s)
- Liangcheng Tong
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Zhiwei Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Wei Dai
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Zhongyang Sun
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Junsheng Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Qing Xue
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Ying Li
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
4
|
Azadi S, Yazdanpanah MA, Afshari A, Alahdad N, Chegeni S, Angaji A, Rezayat SM, Tavakol S. Bioinspired synthetic peptide-based biomaterials regenerate bone through biomimicking of extracellular matrix. J Tissue Eng 2024; 15:20417314241303818. [PMID: 39670180 PMCID: PMC11635874 DOI: 10.1177/20417314241303818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
There have been remarkable advancements in regenerative medicine for bone regeneration, tackling the worldwide health concern of tissue loss. Tissue engineering uses the body's natural capabilities and applies biomaterials and bioactive molecules to replace damaged or lost tissues and restore their functionality. While synthetic ceramics have overcome some challenges associated with allografts and xenografts, they still need essential growth factors and biomolecules. Combining ceramics and bioactive molecules, such as peptides derived from biological motifs of vital proteins, is the most effective approach to achieve optimal bone regeneration. These bioactive peptides induce various cellular processes and modify scaffold properties by mimicking the function of natural osteogenic, angiogenic and antibacterial biomolecules. The present review aims to consolidate the latest and most pertinent information on the advancements in bioactive peptides, including angiogenic, osteogenic, antimicrobial, and self-assembling peptide nanofibers for bone tissue regeneration, elucidating their biological effects and potential clinical implications.
Collapse
Affiliation(s)
- Sareh Azadi
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Yazdanpanah
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Ali Afshari
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Niloofar Alahdad
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Solmaz Chegeni
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolhamid Angaji
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Research and Development, Tavakol Biomimetic Technologies Company, Tehran, Iran
| |
Collapse
|
5
|
Xu Y, Zhuo J, Wang Q, Xu X, He M, Zhang L, Liu Y, Wu X, Luo K, Chen Y. Site-specific periosteal cells with distinct osteogenic and angiogenic characteristics. Clin Oral Investig 2023; 27:7437-7450. [PMID: 37848582 DOI: 10.1007/s00784-023-05333-3] [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: 06/28/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVES This study aimed to investigate the site-specific characteristics of rat mandible periosteal cells (MPCs) and tibia periosteal cells (TPCs) to assess the potential application of periosteal cells (PCs) in bone tissue engineering (BTE). MATERIALS AND METHODS MPCs and TPCs were isolated and characterized. The potential of proliferation, migration, osteogenesis and adipogenesis of MPCs and TPCs were evaluated by CCK-8, scratch assay, Transwell assay, alkaline phosphatase staining and activity, Alizarin Red S staining, RT‒qPCR, and Western blot (WB) assays, respectively. Then, these cells were cocultured with human umbilical vein endothelial cells (HUVECs) to investigate their angiogenic capacity, which was assessed by scratch assay, Transwell assay, Matrigel tube formation assay, RT‒qPCR, and WB assays. RESULTS MPCs exhibited higher osteogenic potential, higher alkaline phosphatase activity, and more mineralized nodule formation, while TPCs showed a greater capability for proliferation, migration, and adipogenesis. MPCs showed higher expression of angiogenic factors, and the conditioned medium of MPCs accelerated the migration of HUVECs, while MPC- conditioned medium induced the formation of more tubular structure in HUVECs in vitro. These data suggest that compared to TPCs, MPCs exert more consequential proangiogenic effects on HUVECs. CONCLUSIONS PCs possess skeletal site-specific differences in biological characteristics. MPCs exhibit more eminent osteogenic and angiogenic potentials, which highlights the potential application of MPCs for BTE. CLINICAL RELEVANCE Autologous bone grafting as the main modality for maxillofacial bone defect repair has many limitations. Constituting an important cell type in bone repair and regeneration, MPCs show greater potential for application in BTE, which provides a promising treatment option for maxillofacial bone defect repair.
Collapse
Affiliation(s)
- Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Jin Zhuo
- Xuzhou Stomatological Hospital, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Qisong Wang
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 354000, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Xiaohong Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| | - Yuling Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| |
Collapse
|
6
|
Syahruddin MH, Anggraeni R, Ana ID. A microfluidic organ-on-a-chip: into the next decade of bone tissue engineering applied in dentistry. Future Sci OA 2023; 9:FSO902. [PMID: 37753360 PMCID: PMC10518836 DOI: 10.2144/fsoa-2023-0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
A comprehensive understanding of the complex physiological and pathological processes associated with alveolar bones, their responses to different therapeutics strategies, and cell interactions with biomaterial becomes necessary in precisely treating patients with severe progressive periodontitis, as a bone-related issue in dentistry. However, existing monolayer cell culture or pre-clinical models have been unable to mimic the complex physiological, pathological and regeneration processes in the bone microenvironment in response to different therapeutic strategies. In this point, 'organ-on-a-chip' (OOAC) technology, specifically 'alveolar-bone-on-a-chip', is expected to resolve the problems by better imitating infection site microenvironment and microphysiology within the oral tissues. The OOAC technology is assessed in this study toward better approaches in disease modeling and better therapeutics strategy for bone tissue engineering applied in dentistry.
Collapse
Affiliation(s)
- Muhammad Hidayat Syahruddin
- Postgraduate Student, Dental Science Doctoral Study Program, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Rahmi Anggraeni
- Research Center for Preclinical & Clinical Medicine, National Research & Innovation Agency of the Republic of Indonesia, Cibinong Science Center, Bogor, 16915, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| |
Collapse
|
7
|
Li Y, Yang Z, Tong L, Yang J, Wang J, Wen Y. Wall thickness analysis method for judging the degree of lower extremity long bone healing. Sci Rep 2023; 13:20650. [PMID: 38001361 PMCID: PMC10673992 DOI: 10.1038/s41598-023-48212-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 11/23/2023] [Indexed: 11/26/2023] Open
Abstract
To evaluate the possibility of judging the degree of bone healing by wall thickness analysis provide reference for quantitative analysis of bone healing. Patients with lower limb fracture from April 2014 to October 2019 were recruited and divided into bone healing (group A), poor bone healing (group B), and nonunion (group C). Models were built in Mimics 20.0 with DICOM 3.0 data obtained from patient's CT. Three-dimensional geometric models of unaffected limb and affected limb after simulated removal of internal fixation were established, corresponding to basic phase and simulated phase, respectively. Wall thickness analysis was performed to obtain median wall thickness after meshing. R2 (median wall thickness ratio), R4 (CT value ratio), and R5 (healing index ratio) were obtained by calculating the ratio of each value in simulated phase to that in basic phase. Receiver operating characteristic curve analysis was used to evaluate the ability of Wall Thickness Analysis to indicate fracture healing. 112 CT scans of 79 patients were included in the study. The frequency of categorization in groups A, B, and C was 49, 37 and 26, respectively. The median R2 in groups A, B, and C was 0.91, 0.80, and 0.67, respectively (group A > group B > group C, all P < 0.05). The best cutoff point for R2 in predicting bone healing was 0.84, and predicting bone nonunion was 0.74. The Wall Thickness Analysis can be used to quantitatively evaluate fracture healing state, with median wall thickness ratio as a more intuitive and reliable judgment index.
Collapse
Affiliation(s)
- Ying Li
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
- Department of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Zhiwei Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Liangcheng Tong
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Junsheng Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Jianling Wang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Yaoke Wen
- School of Mechanical Engineering, Nanjing University of Science and Technology, No. 200 Xiaolingwei, Nanjing, 210094, Jiangsu, China.
| |
Collapse
|
8
|
Zhang H, Wang M, Wu R, Guo J, Sun A, Li Z, Ye R, Xu G, Cheng Y. From materials to clinical use: advances in 3D-printed scaffolds for cartilage tissue engineering. Phys Chem Chem Phys 2023; 25:24244-24263. [PMID: 37698006 DOI: 10.1039/d3cp00921a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Osteoarthritis caused by articular cartilage defects is a particularly common orthopedic disease that can involve the entire joint, causing great pain to its sufferers. A global patient population of approximately 250 million people has an increasing demand for new therapies with excellent results, and tissue engineering scaffolds have been proposed as a potential strategy for the repair and reconstruction of cartilage defects. The precise control and high flexibility of 3D printing provide a platform for subversive innovation. In this perspective, cartilage tissue engineering (CTE) scaffolds manufactured using different biomaterials are summarized from the perspective of 3D printing strategies, the bionic structure strategies and special functional designs are classified and discussed, and the advantages and limitations of these CTE scaffold preparation strategies are analyzed in detail. Finally, the application prospect and challenges of 3D printed CTE scaffolds are discussed, providing enlightening insights for their current research.
Collapse
Affiliation(s)
- Hewen Zhang
- School of the Faculty of Mechanical Engineering and Mechanic, Ningbo University, Ningbo, Zhejiang Province, 315211, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Meng Wang
- Department of Joint Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China.
| | - Rui Wu
- Department of Orthopedics, Ningbo First Hospital Longshan Hospital Medical and Health Group, Ningbo 315201, P. R. China
| | - Jianjun Guo
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Aihua Sun
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Zhixiang Li
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Ruqing Ye
- Department of Joint Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315020, China.
| | - Gaojie Xu
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| | - Yuchuan Cheng
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Zhejiang Key Laboratory of Additive Manufacturing Materials, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China.
| |
Collapse
|
9
|
Song Y, Wang N, Shi H, Zhang D, Wang Q, Guo S, Yang S, Ma J. Biomaterials combined with ADSCs for bone tissue engineering: current advances and applications. Regen Biomater 2023; 10:rbad083. [PMID: 37808955 PMCID: PMC10551240 DOI: 10.1093/rb/rbad083] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
In recent decades, bone tissue engineering, which is supported by scaffold, seed cells and bioactive molecules (BMs), has provided new hope and direction for treating bone defects. In terms of seed cells, compared to bone marrow mesenchymal stem cells, which were widely utilized in previous years, adipose-derived stem cells (ADSCs) are becoming increasingly favored by researchers due to their abundant sources, easy availability and multi-differentiation potentials. However, there is no systematic theoretical basis for selecting appropriate biomaterials loaded with ADSCs. In this review, the regulatory effects of various biomaterials on the behavior of ADSCs are summarized from four perspectives, including biocompatibility, inflammation regulation, angiogenesis and osteogenesis, to illustrate the potential of combining various materials with ADSCs for the treatment of bone defects. In addition, we conclude the influence of additional application of various BMs on the bone repair effect of ADSCs, in order to provide more evidences and support for the selection or preparation of suitable biomaterials and BMs to work with ADSCs. More importantly, the associated clinical case reports and experiments are generalized to provide additional ideas for the clinical transformation and application of bone tissue engineering loaded with ADSCs.
Collapse
Affiliation(s)
- Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ning Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Dan Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Jia Ma
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| |
Collapse
|
10
|
Agnes CJ, Karoichan A, Tabrizian M. The Diamond Concept Enigma: Recent Trends of Its Implementation in Cross-linked Chitosan-Based Scaffolds for Bone Tissue Engineering. ACS APPLIED BIO MATERIALS 2023. [PMID: 37310896 PMCID: PMC10354806 DOI: 10.1021/acsabm.3c00108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
An increasing number of publications over the past ten years have focused on the development of chitosan-based cross-linked scaffolds to regenerate bone tissue. The design of biomaterials for bone tissue engineering applications relies heavily on the ideals set forth by a polytherapy approach called the "Diamond Concept". This methodology takes into consideration the mechanical environment, scaffold properties, osteogenic and angiogenic potential of cells, and benefits of osteoinductive mediator encapsulation. The following review presents a comprehensive summarization of recent trends in chitosan-based cross-linked scaffold development within the scope of the Diamond Concept, particularly for nonload-bearing bone repair. A standardized methodology for material characterization, along with assessment of in vitro and in vivo potential for bone regeneration, is presented based on approaches in the literature, and future directions of the field are discussed.
Collapse
Affiliation(s)
- Celine J Agnes
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
- Shriner's Hospital for Children, Montreal, Quebec H4A 0A9 Canada
| | - Antoine Karoichan
- Shriner's Hospital for Children, Montreal, Quebec H4A 0A9 Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec H3A 1G1 Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec H3A 1G1 Canada
| |
Collapse
|
11
|
Ni X, Xing X, Deng Y, Li Z. Applications of Stimuli-Responsive Hydrogels in Bone and Cartilage Regeneration. Pharmaceutics 2023; 15:pharmaceutics15030982. [PMID: 36986842 PMCID: PMC10056098 DOI: 10.3390/pharmaceutics15030982] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Bone and cartilage regeneration is an area of tremendous interest and need in health care. Tissue engineering is a potential strategy for repairing and regenerating bone and cartilage defects. Hydrogels are among the most attractive biomaterials in bone and cartilage tissue engineering, mainly due to their moderate biocompatibility, hydrophilicity, and 3D network structure. Stimuli-responsive hydrogels have been a hot topic in recent decades. They can respond to external or internal stimulation and are used in the controlled delivery of drugs and tissue engineering. This review summarizes current progress in the use of stimuli-responsive hydrogels in bone and cartilage regeneration. The challenges, disadvantages, and future applications of stimuli-responsive hydrogels are briefly described.
Collapse
Affiliation(s)
- Xiaoqi Ni
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Xin Xing
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yunfan Deng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhi Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
12
|
Nikody M, Li J, Balmayor ER, Moroni L, Habibovic P. The addition of zinc ions to polymer-ceramic composites accelerated osteogenic differentiation of human mesenchymal stromal cells. BIOMATERIALS ADVANCES 2023; 149:213391. [PMID: 36990024 DOI: 10.1016/j.bioadv.2023.213391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023]
Abstract
Critical-sized bone defects, caused by congenital disorders or trauma, are defects that will not heal spontaneously and require surgical intervention. Recent advances in biomaterial design for the treatment of such defects focus on improving their osteoinductive properties. Here, we propose a bioactive composite with high ceramic content composed of poly(ethyleneoxide terephthalate)/poly(butylene terephthalate) (1000PEOT70PBT30, PolyActive, PA) and 50 % beta-tricalcium phosphate (β-TCP) with the addition of zinc in a form of a coating on the TCP particles. Due to its essential role in bone homeostasis, we hypothesised that the addition of zinc to the polymer-ceramic composite will further enhance its osteogenic properties. β-TCP particles were immersed in a zinc solution with a concentration of 15 or 45 mM. The addition of zinc did not alter the β-TCP composition or the release of calcium or phosphate ions. 3D porous 1000PEOT70PBT30 - β-TCP scaffolds were additively manufactured by "3D fibre deposition" and their ability to support the osteogenic differentiation was assessed by culturing clinically relevant human mesenchymal stromal cells (hMSCs) on the scaffolds for 3, 7, 14 and 28 days. The expression of osteogenic gene markers was increased in the presence of both zinc concentrations. Remarkably, upregulation of osteocalcin (OCN), a late osteogenic marker, was observed after three days of culture. Furthermore, enhanced extracellular matrix (ECM) production and mineralization was observed. These findings support the existing evidence on the osteogenic properties of zinc and further demonstrate that the incorporation of zinc into a polymer-ceramic composite could be a promising strategy in the field of regeneration of critical-sized bone defects.
Collapse
Affiliation(s)
- Martyna Nikody
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands; Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands.
| | - Jiaping Li
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands; Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Elizabeth Rosado Balmayor
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands; Experimental Orthopaedics and Trauma Surgery, Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands.
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
13
|
Li A, Sasaki JI, Abe GL, Katata C, Sakai H, Imazato S. Vascularization of a Bone Organoid Using Dental Pulp Stem Cells. Stem Cells Int 2023; 2023:5367887. [PMID: 37200632 PMCID: PMC10188257 DOI: 10.1155/2023/5367887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/06/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023] Open
Abstract
Bone organoids offer a novel path for the reconstruction and repair of bone defects. We previously fabricated scaffold-free bone organoids using cell constructs comprising only bone marrow-derived mesenchymal stem cells (BMSCs). However, the cells in the millimetre-scale constructs were likely to undergo necrosis because of difficult oxygen diffusion and nutrient delivery. Dental pulp stem cells (DPSCs) are capable of differentiating into vascular endothelial lineages and have great vasculogenic potential under endothelial induction. Therefore, we hypothesized that DPSCs can serve as a vascular source to improve the survival of the BMSCs within the bone organoid. In this study, the DPSCs had greater sprouting ability, and the proangiogenic marker expressions were significantly greater than those of BMSCs. DPSCs were incorporated into the BMSC constructs at various ratios (5%-20%), and their internal structures and vasculogenic and osteogenic characteristics were investigated after endothelial differentiation. As a result, the DPSCs are differentiated into the CD31-positive endothelial lineage in the cell constructs. The incorporation of DPSCs significantly suppressed cell necrosis and improved the viability of the cell constructs. In addition, lumen-like structures were visualized by fluorescently labelled nanoparticles in the DPSC-incorporated cell constructs. The vascularized BMSC constructs were successfully fabricated using the vasculogenic ability of the DPSCs. Next, osteogenic induction was initiated in the vascularized BMSC/DPSC constructs. Compared with only BMSCs, constructs with DPSCs had increased mineralized deposition and a hollow structure. Overall, this study demonstrated that vascularized scaffold-free bone organoids were successfully fabricated by incorporating DPSCs into BMSC constructs, and the biomimetic biomaterial is promising for bone regenerative medicine and drug development.
Collapse
Affiliation(s)
- Aonan Li
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Jun-Ichi Sasaki
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Gabriela L. Abe
- Department of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Chihiro Katata
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hirohiko Sakai
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Satoshi Imazato
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
14
|
Paladini F, Pollini M. Novel Approaches and Biomaterials for Bone Tissue Engineering: A Focus on Silk Fibroin. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6952. [PMID: 36234293 PMCID: PMC9572978 DOI: 10.3390/ma15196952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 05/16/2023]
Abstract
Bone tissue engineering (BTE) represents a multidisciplinary research field involving many aspects of biology, engineering, material science, clinical medicine and genetics to create biological substitutes to promote bone regeneration. The definition of the most appropriate biomaterials and structures for BTE is still a challenge for researchers, aiming at simultaneously combining different features such as tissue generation properties, biocompatibility, porosity and mechanical strength. In this scenario, among the biomaterials for BTE, silk fibroin represents a valuable option for the development of functional devices because of its unique biological properties and the multiple chances of processing. This review article aims at providing the reader with a general overview of the most recent progresses in bone tissue engineering in terms of approaches and materials with a special focus on silk fibroin and the related mechanisms involved in bone regeneration, and presenting interesting results obtained by different research groups, which assessed the great potential of this protein for bone tissue engineering.
Collapse
Affiliation(s)
- Federica Paladini
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy
- Caresilk S.r.l.s., Via Monteroni c/o Technological District DHITECH, 73100 Lecce, Italy
| | - Mauro Pollini
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy
- Caresilk S.r.l.s., Via Monteroni c/o Technological District DHITECH, 73100 Lecce, Italy
| |
Collapse
|
15
|
Bai B, Hou M, Hao J, Liu Y, Ji G, Zhou G. Research progress in seed cells for cartilage tissue engineering. Regen Med 2022; 17:659-675. [PMID: 35703020 DOI: 10.2217/rme-2022-0023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cartilage defects trouble millions of patients worldwide and their repair via conventional treatment is difficult. Excitingly, tissue engineering technology provides a promising strategy for efficient cartilage regeneration with structural regeneration and functional reconstruction. Seed cells, as biological prerequisites for cartilage regeneration, determine the quality of regenerated cartilage. The proliferation, differentiation and chondrogenesis of seed cells are greatly affected by their type, origin and generation. Thus, a systematic description of the characteristics of seed cells is necessary. This article reviews in detail the cellular characteristics, research progress, clinical translation challenges and future research directions of seed cells while providing guidelines for selecting appropriate seed cells for cartilage regeneration.
Collapse
Affiliation(s)
- Baoshuai Bai
- Research Institute of Plastic Surgery, Wei Fang Medical University, Wei Fang, Shandong, 261053, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.,National Tissue Engineering Center of China, Shanghai, 200240, China
| | - Mengjie Hou
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.,National Tissue Engineering Center of China, Shanghai, 200240, China
| | - Junxiang Hao
- Research Institute of Plastic Surgery, Wei Fang Medical University, Wei Fang, Shandong, 261053, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.,National Tissue Engineering Center of China, Shanghai, 200240, China
| | - Yanhan Liu
- Shanghai JiaoTong University School of Medicine, Shanghai, 200240, China
| | - Guangyu Ji
- Department of Thoracic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200240, China
| | - Guangdong Zhou
- Research Institute of Plastic Surgery, Wei Fang Medical University, Wei Fang, Shandong, 261053, China.,Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.,National Tissue Engineering Center of China, Shanghai, 200240, China
| |
Collapse
|
16
|
Meng M, Xia Q, Li Y, Chen X, Wang Q, Chen J, Xu X, Wang H, Shu J, Lu J, Cheng L, Ye Z, Song B, Dong Q. Enamel matrix derivative expedites osteogenic differentiation of BMSCs via Wnt/β-catenin pathway in high glucose microenvironment. J Bone Miner Metab 2022; 40:448-459. [PMID: 35347430 DOI: 10.1007/s00774-022-01318-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/29/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The influence of enamel matrix derivative (EMD) on proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) was explored in high glucose (HG) microenvironment with interaction of Wnt/β-catenin pathway. MATERIALS AND METHODS Extraction of BMSCs from Sprague-Dawley rats, culture, and identification were manifested. The cells were treated with different concentration of EMD in HG to figure out the most available concentration for proliferation and osteogenic differentiation. Then, observation of cell growth curve and cell cycle changes, and detection of Osterix, runt-related transcription factor 2 (Runx2), COL-I, early osteogenic indexes, Calcium salt deposition, and β-catenin protein in Wnt/β-catenin pathway were assured. After adding Wnt/β-catenin pathway inhibitor (XAV-939) in the cells with osteogenesis induction, detection of binding of β-catenin to Osterix was clarified. RESULTS Via identification BMSCs cultured in vitro was qualified. Different concentrations of EMD could accelerate cell proliferation in HG and osteogenesis induction, and 75 μg/mL EMD had the best effect. The HG augmented BMSCs proliferation and the propidium iodide index of flow cytometry cycle was elevated in HG, which were strengthened via the EMD. After BMSCs' osteogenesis induction, Osterix, Runx2, CoL-1, early osteogenic indexes, and calcium salt deposition were reduced, but elevated via EMD. β-Catenin was the lowest in the HG, but elevated after EMD. After addition of XAV-939, reduction of β-catenin and the downstream (Osterix and Runx2) were manifested. Detection of binding protein bands was in β-catenin and Osterix of the HG after EMD treatment. CONCLUSION EMD may facilitate the osteogenic differentiation of BMSCs via activating the Wnt/β-catenin pathway in HG.
Collapse
Affiliation(s)
- MaoHua Meng
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - Qian Xia
- Department of Preventive and Pediatric Dentistry, Stomatological Hospital of Guizhou Medical University, Guiyang City, 550004, Guizhou Province, China
| | - Ying Li
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - Xin Chen
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - QinYing Wang
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - JingQiao Chen
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - XingXing Xu
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - Huan Wang
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - JiaYu Shu
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - Jing Lu
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China
| | - Lu Cheng
- Department of Stomatology, Guiyang Hospital of Stomatology, Guiyang City, 550005, Guizhou Province, China
| | - ZhaoYang Ye
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China.
| | - Bin Song
- Department of Prosthodontics, Guizhou Provincial People's Hospital, 83 Zhongshan East Road, Guiyang, Guiyang City, 550003, Guizhou Province, China.
| | - Qiang Dong
- School of Stomatology, Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China.
- Department of Prosthodontics, Stomatological Hospital of Guizhou Medical University, 9 Beijing Road, Yunyan District, Guiyang City, 550004, Guizhou Province, China.
| |
Collapse
|
17
|
Chen YQ, Hu WH, Dong ZC, Dong SW. Multi-functional osteoclasts in matrix-based tissue engineering bone. Chin J Traumatol 2022; 25:132-137. [PMID: 34969539 PMCID: PMC9125721 DOI: 10.1016/j.cjtee.2021.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 09/30/2021] [Accepted: 11/05/2021] [Indexed: 02/04/2023] Open
Abstract
The repair of bone defects, especially for the large segment of bone defects, has always been an urgent problem in orthopedic clinic and attracted researchers' attention. Nowadays, the application of tissue engineering bone in the repair of bone defects has become the research hotspot. With the rapid development of tissue engineering, the novel and functional scaffold materials for bone repair have emerged. In this review, we have summarized the multi-functional roles of osteoclasts in bone remodeling. The development of matrix-based tissue engineering bone has laid a theoretical foundation for further investigation about the novel bone regeneration materials which could perform high bioactivity. From the point of view on preserving pre-osteoclasts and targeting mature osteoclasts, this review introduced the novel matrix-based tissue engineering bone based on osteoclasts in the field of bone tissue engineering, which provides a potential direction for the development of novel scaffold materials for the treatment of bone defects.
Collapse
Affiliation(s)
- Yue-Qi Chen
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wen-Hui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zi-Cai Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shi-Wu Dong
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China,Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China,Corresponding author. Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
18
|
Nambiar J, Jana S, Nandi SK. Strategies for Enhancing Vascularization of Biomaterial-Based Scaffold in Bone Regeneration. CHEM REC 2022; 22:e202200008. [PMID: 35352873 DOI: 10.1002/tcr.202200008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/12/2022] [Indexed: 12/29/2022]
Abstract
Despite the recent advances in reconstructive orthopedics; fracture union is a challenge to bone regeneration. Concurrent angiogenesis is a complex process governed by events, delicately entwined with osteogenesis. However, poorly perfused scaffolds have lower success rates; necessitating the need for a better vascular component, which is important for the delivery of nutrients, oxygen, waste elimination, recruitment of cells for optimal bone repair. This review highlights the latest strategies to promote biomaterial-based scaffold vascularization by incorporation of cells, growth factors, inorganic ions, etc. into natural or synthetic polymers, ceramic materials, or composites of organic and inorganic compounds. Furthermore, it emphasizes structural modifications, biophysical stimuli, and natural molecules to fabricate scaffolds aiding the genesis of dense vascularization following their implantation to manifest a compatible regenerative microenvironment without graft rejection.
Collapse
Affiliation(s)
- Jasna Nambiar
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata, 700037, India
| | - Sonali Jana
- Department of Veterinary Physiology, West Bengal University of Animal & Fishery Sciences, Kolkata, 700037, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata, 700037, India
| |
Collapse
|
19
|
Qin D, Wang N, You XG, Zhang AD, Chen XG, Liu Y. Collagen-based biocomposites inspired by bone hierarchical structures for advanced bone regeneration: ongoing research and perspectives. Biomater Sci 2021; 10:318-353. [PMID: 34783809 DOI: 10.1039/d1bm01294k] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone is a hard-connective tissue composed of matrix, cells and bioactive factors with a hierarchical structure, where the matrix is mainly composed of type I collagen and hydroxyapatite. Collagen fibers assembled by collagen are the template for mineralization and make an important contribution to bone formation and the bone remodeling process. Therefore, collagen has been widely clinically used for bone/cartilage defect regeneration. However, pure collagen implants, such as collagen scaffolds or sponges, have limitations in the bone/cartilage regeneration process due to their poor mechanical properties and osteoinductivity. Different forms of collagen-based composites prepared by incorporating natural/artificial polymers or bioactive inorganic substances are characterized by their interconnected porous structure and promoting cell adhesion, while they improve the mechanical strength, structural stability and osteogenic activities of the collagen matrix. In this review, various forms of collagen-based biocomposites, such as scaffolds, sponges, microspheres/nanoparticles, films and microfibers/nanofibers prepared by natural/synthetic polymers, bioactive ceramics and carbon-based materials compounded with collagen are reviewed. In addition, the application of collagen-based biocomposites as cytokine, cell or drug (genes, proteins, peptides and chemosynthetic) delivery platforms for proangiogenesis and bone/cartilage tissue regeneration is also discussed. Finally, the potential application, research and development direction of collagen-based biocomposites in future bone/cartilage tissue regeneration are discussed.
Collapse
Affiliation(s)
- Di Qin
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Na Wang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Xin-Guo You
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - An-Di Zhang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Xi-Guang Chen
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Ya Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| |
Collapse
|
20
|
Liu G, Sun J, Gong M, Xing F, Wu S, Xiang Z. Urine-derived stem cells loaded onto a chitosan-optimized biphasic calcium-phosphate scaffold for repairing large segmental bone defects in rabbits. J Biomed Mater Res B Appl Biomater 2021; 109:2014-2029. [PMID: 33979024 DOI: 10.1002/jbm.b.34850] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 02/05/2023]
Abstract
The treatment of large segmental bone defects can be challenging for orthopedic surgeons. The development of bone tissue engineering technology, including the selection of seeding cells and the construction of scaffolds, provides a promising solution. In this study, we investigated osteogenic differentiation of human urine-derived stem cells (hUSCs, a newly identified class of stem cells), and developed a novel porous hybrid scaffold using biphasic calcium phosphate (BCP) bioceramic ornamented with chitosan sponges (CS). We combined hUSCs with a CS/BCP hybrid scaffold to construct tissue-engineered bone and evaluated whether the combination promotes bone regeneration in large segmental bone defects in rabbits. The study showed that hUSCs can differentiate into osteoblasts, and the hUSCs adhered, proliferated, and differentiated on CS/BCP hybrid scaffolds. Micro-computed tomography measurements, biomechanical detection, and histological analyses revealed that the combination of hUSCs and the CS/BCP hybrid scaffold enhanced bone regeneration more effectively compared with conventional pure BCP scaffolds, indicating that hUSCs can be used as a cell source for bone tissue engineering and that cell-scaffold-based biomimetic bone may be a promising approach to the repair of bone defects.
Collapse
Affiliation(s)
- Guoming Liu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, PR China.,Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Jiachen Sun
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Min Gong
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Fei Xing
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Shuang Wu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Zhou Xiang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, PR China.,Division of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
21
|
Xing Z, Jiang X, Si Q, Finne-Wistrand A, Liu B, Xue Y, Mustafa K. Endochondral Ossification Induced by Cell Transplantation of Endothelial Cells and Bone Marrow Stromal Cells with Copolymer Scaffold Using a Rat Calvarial Defect Model. Polymers (Basel) 2021; 13:polym13091521. [PMID: 34065081 PMCID: PMC8125936 DOI: 10.3390/polym13091521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
It has been recently reported that, in a rat calvarial defect model, adding endothelial cells (ECs) to a culture of bone marrow stromal cells (BMSCs) significantly enhanced bone formation. The aim of this study is to further investigate the ossification process of newly formed osteoid and host response to the poly(L-lactide-co-1,5-dioxepan-2-one) [poly(LLA-co-DXO)] scaffolds based on previous research. Several different histological methods and a PCR Array were applied to evaluate newly formed osteoid after 8 weeks after implantation. Histological results showed osteoid formed in rat calvarial defects and endochondral ossification-related genes, such as dentin matrix acidic phosphoprotein 1 (Dmp1) and collagen type II, and alpha 1 (Col2a1) exhibited greater expression in the CO (implantation with BMSC/EC/Scaffold constructs) than the BMSC group (implantation with BMSC/Scaffold constructs) as demonstrated by PCR Array. It was important to notice that cartilage-like tissue formed in the pores of the copolymer scaffolds. In addition, multinucleated giant cells (MNGCs) were observed surrounding the scaffold fragments. It was concluded that the mechanism of ossification might be an endochondral ossification process when the copolymer scaffolds loaded with co-cultured ECs/BMSCs were implanted into rat calvarial defects. MNGCs were induced by the poly(LLA-co-DXO) scaffolds after implantation, and more specific in vivo studies are needed to gain a better understanding of host response to copolymer scaffolds.
Collapse
Affiliation(s)
- Zhe Xing
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway;
| | - Xiaofeng Jiang
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
| | - Qingzong Si
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Teknikringen 56-58, SE 100-44 Stockholm, Sweden;
| | - Bin Liu
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
- Correspondence: (B.L.); (Y.X.); Tel.: +86-9318915051 (B.L.); +47-55586519 (Y.X.)
| | - Ying Xue
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway;
- Correspondence: (B.L.); (Y.X.); Tel.: +86-9318915051 (B.L.); +47-55586519 (Y.X.)
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway;
| |
Collapse
|
22
|
Li W, Liu W, Wang W, Wang J, Ma T, Chen J, Wu H, Liu C. Sinusoidal electromagnetic fields accelerate bone regeneration by boosting the multifunctionality of bone marrow mesenchymal stem cells. Stem Cell Res Ther 2021; 12:234. [PMID: 33849651 PMCID: PMC8042357 DOI: 10.1186/s13287-021-02302-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 12/26/2022] Open
Abstract
Background The repair of critical-sized bone defects is always a challenging problem. Electromagnetic fields (EMFs), used as a physiotherapy for bone defects, have been suspected to cause potential hazards to human health due to the long-term exposure. To optimize the application of EMF while avoiding its adverse effects, a combination of EMF and tissue engineering techniques is critical. Furthermore, a deeper understanding of the mechanism of action of EMF will lead to better applications in the future. Methods In this research, bone marrow mesenchymal stem cells (BMSCs) seeded on 3D-printed scaffolds were treated with sinusoidal EMFs in vitro. Then, 5.5 mm critical-sized calvarial defects were created in rats, and the cell scaffolds were implanted into the defects. In addition, the molecular and cellular mechanisms by which EMFs regulate BMSCs were explored with various approaches to gain deeper insight into the effects of EMFs. Results The cell scaffolds treated with EMF successfully accelerated the repair of critical-sized calvarial defects. Further studies revealed that EMF could not directly induce the differentiation of BMSCs but improved the sensitivity of BMSCs to BMP signals by upregulating the quantity of specific BMP (bone morphogenetic protein) receptors. Once these receptors receive BMP signals from the surrounding milieu, a cascade of reactions is initiated to promote osteogenic differentiation via the BMP/Smad signalling pathway. Moreover, the cytokines secreted by BMSCs treated with EMF can better facilitate angiogenesis and osteoimmunomodulation which play fundamental roles in bone regeneration. Conclusion In summary, EMF can promote the osteogenic potential of BMSCs and enhance the paracrine function of BMSCs to facilitate bone regeneration. These findings highlight the profound impact of EMF on tissue engineering and provide a new strategy for the clinical treatment of bone defects.
Collapse
Affiliation(s)
- Weigang Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Wenbin Liu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Wei Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jiachen Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jingyuan Chen
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Chaoxu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
23
|
Vascular Endothelial Growth Factor and Mesenchymal Stem Cells Revealed Similar Bone Formation to Allograft in a Sheep Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6676609. [PMID: 33763484 PMCID: PMC7946458 DOI: 10.1155/2021/6676609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/07/2021] [Accepted: 02/25/2021] [Indexed: 01/08/2023]
Abstract
Introduction Mesenchymal stem cells (MSCs) and vascular endothelial growth factor (VEGF) are key factors in bone regeneration. Further stimulation should establish an enhanced cell environment optimal for vessel evolvement and hereby being able to attract bone-forming cells. The aim of this study was to generate new bone by using MSCs and VEGF, being able to stimulate growth equal to allograft. Methods Eight Texel/Gotland sheep had four titanium implants in a size of 10 × 12 mm inserted into bilateral distal femurs, containing a 2 mm gap. In the gap, autologous 3 × 106 MSCs seeded on hydroxyapatite (HA) granules in combination with 10 ng, 100 ng, and 500 ng VEGF release/day were added. After 12 weeks, the implant-bone blocks were harvested, embedded, and sectioned for histomorphometric analysis. Bone formation and mechanical fixation were evaluated. Blood samples were collected for the determination of bone-related biomarkers and VEGF in serum at weeks 0, 1, 4, 8, and 12. Results The combination of 3 × 106 MSCs with 10 ng, 100 ng, and 500 ng VEGF release/day exhibited similar amount of bone formation within the gap as allograft (P > 0.05). Moreover, no difference in mechanical fixation was observed between the groups (P > 0.05). Serum biomarkers showed no significant difference compared to baseline (all P > 0.05). Conclusion MSCs and VEGF exhibit significant bone regeneration, and their bone properties equal to allograft, with no systemic increase in osteogenic markers or VEGF with no visible side effects. This study indicates a possible new approach into solving the problem of insufficient allograft, in larger bone defects.
Collapse
|
24
|
Li X, Wu J, Li D, Zou Q, Man Y, Zou L, Li W. Pro-osteogenesis and in vivo tracking investigation of a dental implantation system comprising novel mTi implant and HYH-Fe particles. Bioact Mater 2021; 6:2658-2666. [PMID: 33665498 PMCID: PMC7890097 DOI: 10.1016/j.bioactmat.2021.01.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/19/2021] [Accepted: 01/30/2021] [Indexed: 02/05/2023] Open
Abstract
Insufficient early osteogenesis seriously affects the later stage osteogenic quality and osseointegration of dental implants. To promote early osteogenesis, we first designed a Ti dental implant with a built-in magnet (mTi) to produce a local static magnetic field (SMF). Then, a dental implantation system comprising the mTi implant and the superparamagnetic hydroxyapatite (HA:Yb/Ho-Fe, named HYH-Fe) particles was implanted into the alveolar bone of beagles. The results showed that the mTi + HYH-Fe group displayed better early osteogenesis and later stage osseointegration than the Ti + HA and mTi + HA groups. A combination of the local SMF (mTi) and superparamagnetic HYH-Fe particles had a positive effect on the pro-osteogenesis of Ti implants. The results also indicated that week 10 could be adopted as the key time point to evaluate the early osteogenic effect of the mTi + HYH-Fe implantation system, which would be a promising prospect for promotion of osteogenesis, in vivo tracking investigation of material-bone relationships, and clinical applications.
Collapse
Affiliation(s)
- Xiyu Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Juan Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Danxue Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qin Zou
- Analytical and Testing Center, Sichuan University, Chengdu, 610064, China
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Wei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
25
|
Bozorgi A, Sabouri L. Osteosarcoma, personalized medicine, and tissue engineering; an overview of overlapping fields of research. Cancer Treat Res Commun 2021; 27:100324. [PMID: 33517237 DOI: 10.1016/j.ctarc.2021.100324] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/23/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Osteosarcoma is a common bone malignancy in patients of all ages. Surgical and chemotherapy interventions fail to shrink tumor growth and metastasis. The development of efficient patient-specific therapeutic strategies for osteosarcoma is of great interest in tissue engineering and personalized medicine. The present manuscript aimed to review the advancements in tissue engineering and personalized medicine strategies to overcome osteosarcoma and the relevant biological aspects as well as the current tumor models in vitro and in vivo. RESULTS Tissue engineering and personalized medicine contribute to gene/cell engineering and cell-based therapies specific to genomic and proteomic profiles of individual patients to improve the current treatment options. Also, tissue engineering scaffolds provide physical support to missing bones, could trap cancer cells and deliver immune cells. Taken together, these strategies suppress tumor growth, angiogenic potential, and the subsequent metastasis as well as elicit desirable immune responses against tumor mass. DISCUSSION Advanced and high-throughput gene and protein identification technologies have facilitated the recognition of genomic and proteomic profiles of patients to design and develop patient-specific treatments. The pre-clinical studies showed promising outcomes to inhibit tumor growth and invasion but controversial results compared to clinical investigations make the importance of more clinical reports inevitable. The experimental tumor models assist the evolution of effective treatments by understanding the mechanisms of tumor progression. CONCLUSION Tissue engineering and personalized medicine strategies seem encouraging alternatives to conventional therapies against osteosarcoma. Modeling the tumor microenvironment coupled with pre-clinical results give new intelligence into the translation of strategies into the clinic.
Collapse
Affiliation(s)
- Azam Bozorgi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Leila Sabouri
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Wu S, Lei L, Bao C, Liu J, Weir MD, Ren K, Schneider A, Oates TW, Liu J, Xu HHK. An injectable and antibacterial calcium phosphate scaffold inhibiting Staphylococcus aureus and supporting stem cells for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 120:111688. [PMID: 33545850 DOI: 10.1016/j.msec.2020.111688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Staphylococcus aureus (S. aureus) is the major pathogen for osteomyelitis, which can lead to bone necrosis and destruction. There has been no report on antibacterial calcium phosphate cement (CPC) against S. aureus. The aims of this study were to: (1) develop novel antibacterial CPC-chitosan-alginate microbead scaffold; (2) investigate mechanical and antibacterial properties of CPC-chitosan-penicillin-alginate scaffold; (3) evaluate the encapsulation and delivery of human umbilical cord mesenchymal stem cells (hUCMSCs). Flexural strength, elastic modulus and work-of-fracture of the CPC-chitosan-penicillin-alginate microbeads scaffold and CPC-chitosan scaffold were evaluated. Penicillin release profile and antibacterial effects on S. aureus were determined. The hUCMSC delivery and release from penicillin-alginate microbeads were investigated. Injectable CPC-chitosan-penicillin-alginate microbeads scaffold was developed for the first time. CPC-chitosan-penicillin-alginate microbeads scaffold had a flexural strength of 3.16 ± 0.55 MPa, matching that of cancellous bone. With sustained penicillin release, the new scaffold had strong antibacterial effects on S. aureus, with an inhibition zone diameter of 32.2 ± 2.5 mm, greater than that of penicillin disk control (15.1 ± 2.0 mm) (p < 0.05). Furthermore, this injectable and antibacterial scaffold had no toxic effects, yielding excellent hUCMSC viability, which was similar to that of CPC control without antibacterial activity (p > 0.05). CPC-chitosan-penicillin-microbeads scaffold had injectability, good strength, strong antibacterial effects, and good biocompatibility to support stem cell viability for osteogenesis. CPC-chitosan-penicillin-microbeads scaffold is promising for dental, craniofacial and orthopedic applications to combat infections and promote bone regeneration.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Lei Lei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jin Liu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Key Laboratory of Shannxi for Craniofacial Precision Medicine Research, Clinical Research Center of Shannxi for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Shannxi 710004, China
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas W Oates
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
27
|
Clainche TL, Linklater D, Wong S, Le P, Juodkazis S, Guével XL, Coll JL, Ivanova EP, Martel-Frachet V. Mechano-Bactericidal Titanium Surfaces for Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2020; 12:48272-48283. [PMID: 33054152 DOI: 10.1021/acsami.0c11502] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Despite advances in the development of bone substitutes and strict aseptic procedures, the majority of failures in bone grafting surgery are related to nosocomial infections. Development of biomaterials combining both osteogenic and antibiotic activity is, therefore, a crucial public health issue. Herein, two types of intrinsically bactericidal titanium supports were fabricated by using commercially scalable techniques: plasma etching or hydrothermal treatment, which display two separate mechanisms of mechano-bactericidal action. Hydrothermal etching produces a randomly nanostructured surface with sharp nanosheet protrusions killing bacteria via cutting of the cell membrane, whereas plasma etching of titanium produces a microscale two-tier hierarchical topography that both reduce bacterial attachment and rupture those bacteria that encounter the surface. The adhesion, growth, and proliferation of human adipose-derived stem cells (hASCs) on the two mechano-bactericidal topographies were assessed. Both types of supports allowed the growth and proliferation of the hASCs in the same manner and cells retained their stemness and osteogenic potential. Furthermore, these supports induced osteogenic differentiation of hASCs without the need of differentiation factors, demonstrating their osteoinductive properties. This study proves that these innovative mechano-bactericidal titanium surfaces with both regenerative and bactericidal properties are a promising solution to improve the success rate of reconstructive surgery.
Collapse
Affiliation(s)
- Tristan Le Clainche
- Cancer Target and Experimental Therapeutics, Institute for Advanced Biosciences, INSERM U1209, UMR CNRS 5309, Grenoble Alpes University, Site Santé, Allée des Alpes, 38700 La Tronche, France
| | - Denver Linklater
- School of Science, RMIT University, Melbourne, VIC 3000, Australia
| | - Sherman Wong
- School of Science, RMIT University, Melbourne, VIC 3000, Australia
| | - Phuc Le
- School of Science, RMIT University, Melbourne, VIC 3000, Australia
| | - Saulius Juodkazis
- Optical Sciences Centre and ARC Training Centre in Surface Engineering for Advanced Materials (SEAM), School of Science, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Xavier Le Guével
- Cancer Target and Experimental Therapeutics, Institute for Advanced Biosciences, INSERM U1209, UMR CNRS 5309, Grenoble Alpes University, Site Santé, Allée des Alpes, 38700 La Tronche, France
| | - Jean-Luc Coll
- Cancer Target and Experimental Therapeutics, Institute for Advanced Biosciences, INSERM U1209, UMR CNRS 5309, Grenoble Alpes University, Site Santé, Allée des Alpes, 38700 La Tronche, France
| | - Elena P Ivanova
- School of Science, RMIT University, Melbourne, VIC 3000, Australia
| | - Véronique Martel-Frachet
- Cancer Target and Experimental Therapeutics, Institute for Advanced Biosciences, INSERM U1209, UMR CNRS 5309, Grenoble Alpes University, Site Santé, Allée des Alpes, 38700 La Tronche, France
- EPHE, PSL Research University, 75014 Paris, France
| |
Collapse
|
28
|
Cheng G, Guo S, Wang N, Xiao S, Jiang B, Ding Y. A novel lamellar structural biomaterial and its effect on bone regeneration. RSC Adv 2020; 10:39072-39079. [PMID: 35518390 PMCID: PMC9057690 DOI: 10.1039/d0ra05760f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/09/2020] [Indexed: 01/14/2023] Open
Abstract
To evaluate a novel lamellar structural biomaterial as a potential biomaterial for guided bone regeneration, we describe the preparation of a collagen membrane with high mechanical strength and anti-enzyme degradation ability by using the multi-level structure of Ctenopharyngodon idella scales. The physical and chemical properties, in vitro degradation, biocompatibility, and in vivo osteogenic activity were preliminarily evaluated. In conclusion, it was shown that the multi-layered collagen structure material had sufficient mechanical properties, biocompatibility, and osteogenic ability. Meanwhile, it is also shown that there is a gap in current clinical needs, between the guided tissue regeneration membrane and the one being used. Therefore, this study provides useful insights into the efforts being made to design and adjust the microstructure to balance its mechanical properties, degradation rate, and osteogenic activity. To evaluate a novel lamellar structural biomaterial for guided bone regeneration, we describe the preparation of a collagen membrane with high mechanical strength and anti-enzyme degradation ability using Ctenopharyngodon idella scales.![]()
Collapse
Affiliation(s)
- Guoping Cheng
- Department of Periodontics, West China College of Stomatology, Sichuan University Chengdu 610041 P. R. China +86-28-85501439.,State Key Laboratory of Oral Diseases, Sichuan University Chengdu 610041 P. R. China
| | - Shujuan Guo
- Department of Periodontics, West China College of Stomatology, Sichuan University Chengdu 610041 P. R. China +86-28-85501439.,State Key Laboratory of Oral Diseases, Sichuan University Chengdu 610041 P. R. China
| | - Ningxin Wang
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610065 P. R. China +86-28-85412848 +86-28-85415977
| | - Shimeng Xiao
- Department of Periodontics, West China College of Stomatology, Sichuan University Chengdu 610041 P. R. China +86-28-85501439.,State Key Laboratory of Oral Diseases, Sichuan University Chengdu 610041 P. R. China
| | - Bo Jiang
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610065 P. R. China +86-28-85412848 +86-28-85415977
| | - Yi Ding
- Department of Periodontics, West China College of Stomatology, Sichuan University Chengdu 610041 P. R. China +86-28-85501439.,State Key Laboratory of Oral Diseases, Sichuan University Chengdu 610041 P. R. China
| |
Collapse
|
29
|
Zeiter S, Koschitzki K, Alini M, Jakob F, Rudert M, Herrmann M. Evaluation of Preclinical Models for the Testing of Bone Tissue-Engineered Constructs. Tissue Eng Part C Methods 2020; 26:107-117. [PMID: 31808374 DOI: 10.1089/ten.tec.2019.0213] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Autologous bone grafting is the clinical gold standard for the treatment of large bone defects, but it can only be obtained in limited amounts and is associated with donor site morbidity. These challenges might be overcome by tissue engineering (TE). Although promising results have been reported, translation into clinics often fails. Lack of reproducibility in preclinical studies may be one of the reasons. We evaluated preclinical models for testing of novel TE strategies, as well as the perception of researchers and clinicians toward the models. Therefore, a review of publications on preclinical models of the past 10 years was performed. A survey addressed to both clinicians and scientists was conducted to assess the clinical need for bone tissue engineering (BTE) constructs and researchers were asked about their satisfaction with the currently available preclinical models. A literature review revealed 169 articles on in vivo studies in the field of BTE, including 26 studies utilizing large animal models and 143 studies in small animals, with rabbits and rats presenting the most commonly used species. Only a few studies used skeletally mature animals, which is in large contrast to the patients targeted. The localization of the bone defects varied, but the vast majority (60%) were segmental bone defects with various fixation techniques. Results of 70 surveys confirmed a great clinical need for TE constructs and positive perceptions of all participants toward its future clinical application. Nevertheless, the need for optimization of preclinical models and limitations when it comes to translation of results to the clinical situation were indicated. No clear trends were detected with regards to the preclinical model, leading to most satisfying results despite the trend that scientists rated generally large animal models higher than small animal models. Results of the literature review and the survey reveal the lack of standardized methods. Despite the affirmed clinical need as well as a very positive perception of clinicians toward the use of TE, results indicate a critical need to optimize preclinical models and, in particular, improve translational aspects of the models. A consensus in the field on a limited number of well-standardized models should be reached.
Collapse
Affiliation(s)
| | - Kim Koschitzki
- IZKF Research Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | - Franz Jakob
- Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - Maximilian Rudert
- Department of Orthopaedic Surgery, König-Ludwig-Haus, University of Wuerzburg, Wuerzburg, Germany
| | - Marietta Herrmann
- IZKF Research Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
30
|
Dias RB, Guimarães JAM, Cury MB, Rocha LR, da Costa ES, Nogueira LP, Hochman-Mendez C, Fortuna-Costa A, Silva AKF, Cunha KS, de Souza SAL, Duarte MEL, Sartore RC, Bonfim DC. The Manufacture of GMP-Grade Bone Marrow Stromal Cells with Validated In Vivo Bone-Forming Potential in an Orthopedic Clinical Center in Brazil. Stem Cells Int 2019; 2019:2608482. [PMID: 31781235 PMCID: PMC6875385 DOI: 10.1155/2019/2608482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/26/2019] [Accepted: 09/18/2019] [Indexed: 12/30/2022] Open
Abstract
In vitro-expanded bone marrow stromal cells (BMSCs) have long been proposed for the treatment of complex bone-related injuries because of their inherent potential to differentiate into multiple skeletal cell types, modulate inflammatory responses, and support angiogenesis. Although a wide variety of methods have been used to expand BMSCs on a large scale by using good manufacturing practice (GMP), little attention has been paid to whether the expansion procedures indeed allow the maintenance of critical cell characteristics and potency, which are crucial for therapeutic effectiveness. Here, we described standard procedures adopted in our facility for the manufacture of clinical-grade BMSC products with a preserved capacity to generate bone in vivo in compliance with the Brazilian regulatory guidelines for cells intended for use in humans. Bone marrow samples were obtained from trabecular bone. After cell isolation in standard monolayer flasks, BMSC expansion was subsequently performed in two cycles, in 2- and 10-layer cell factories, respectively. The average cell yield per cell factory at passage 1 was of 21.93 ± 12.81 × 106 cells, while at passage 2, it was of 83.05 ± 114.72 × 106 cells. All final cellular products were free from contamination with aerobic/anaerobic pathogens, mycoplasma, and bacterial endotoxins. The expanded BMSCs expressed CD73, CD90, CD105, and CD146 and were able to differentiate into osteogenic, chondrogenic, and adipogenic lineages in vitro. Most importantly, nine out of 10 of the cell products formed bone when transplanted in vivo. These validated procedures will serve as the basis for in-house BMSC manufacturing for use in clinical applications in our center.
Collapse
Affiliation(s)
- Rhayra B. Dias
- Master Program in Musculoskeletal Sciences, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - João A. M. Guimarães
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
- Trauma Center, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Marco B. Cury
- Hip Surgery Center, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Leonardo R. Rocha
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
- Trauma Center, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Elaine S. da Costa
- Institute of Paediatrics and Puericulture Martagão Gesteira, Federal University of Rio de Janeiro, Rio de Janeiro 21941-912, Brazil
| | | | - Camila Hochman-Mendez
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Texas Heart Institute, Regenerative Medicine Research, Texas 77030, USA
| | - Anneliese Fortuna-Costa
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Anna Karoline F. Silva
- Graduate Program in Pathology, Fluminense Federal University, Rio de Janeiro 24030-215, Brazil
| | - Karin S. Cunha
- Graduate Program in Pathology, Fluminense Federal University, Rio de Janeiro 24030-215, Brazil
| | - Sergio A. L. de Souza
- Department of Radiology, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Maria Eugênia L. Duarte
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Rafaela C. Sartore
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Danielle C. Bonfim
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| |
Collapse
|
31
|
Adipose-Derived Stem Cells in Bone Tissue Engineering: Useful Tools with New Applications. Stem Cells Int 2019; 2019:3673857. [PMID: 31781238 PMCID: PMC6875209 DOI: 10.1155/2019/3673857] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose stem cells (ASCs) are a crucial element in bone tissue engineering (BTE). They are easy to harvest and isolate, and they are available in significative quantities, thus offering a feasible and valid alternative to other sources of mesenchymal stem cells (MSCs), like bone marrow. Together with an advantageous proliferative and differentiative profile, they also offer a high paracrine activity through the secretion of several bioactive molecules (such as growth factors and miRNAs) via a sustained exosomal release which can exert efficient conditioning on the surrounding microenvironment. BTE relies on three key elements: (1) scaffold, (2) osteoprogenitor cells, and (3) bioactive factors. These elements have been thoroughly investigated over the years. The use of ASCs has offered significative new advancements in the efficacy of each of these elements. Notably, the phenotypic study of ASCs allowed discovering cell subpopulations, which have enhanced osteogenic and vasculogenic capacity. ASCs favored a better vascularization and integration of the scaffolds, while improvements in scaffolds' materials and design tried to exploit the osteogenic features of ASCs, thus reducing the need for external bioactive factors. At the same time, ASCs proved to be an incredible source of bioactive, proosteogenic factors that are released through their abundant exosome secretion. ASC exosomes can exert significant paracrine effects in the surroundings, even in the absence of the primary cells. These paracrine signals recruit progenitor cells from the host tissues and enhance regeneration. In this review, we will focus on the recent discoveries which have involved the use of ASCs in BTE. In particular, we are going to analyze the different ASCs' subpopulations, the interaction between ASCs and scaffolds, and the bioactive factors which are secreted by ASCs or can induce their osteogenic commitment. All these advancements are ultimately intended for a faster translational and clinical application of BTE.
Collapse
|
32
|
Fu T, Liang P, Song J, Wang J, Zhou P, Tang Y, Li J, Huang E. Matrigel Scaffolding Enhances BMP9-induced Bone Formation in Dental Follicle Stem/Precursor Cells. Int J Med Sci 2019; 16:567-575. [PMID: 31171908 PMCID: PMC6535656 DOI: 10.7150/ijms.30801] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
Bone tissue engineering requires a combination of cells, efficient biochemical and physicochemical factors, and biocompatible scaffolds. In this study, we evaluated the potential use of injectable Matrigel as a scaffold for the delivery of rat dental follicle stem/precursor cells (rDFSCs) transduced by bone morphogenetic protein (BMP) 9 to enhance osteogenic differentiation in vitro and promote ectopic bone formation in vivo. Recombinant adenovirus was used to overexpress BMP9 in rDFSCs. Alkaline phosphatase activity was measured using a histochemical staining assay and a chemiluminescence assay kit. Quantitative real-time polymerase chain reaction was used to determine mRNA expression levels of bone-related genes including distal-less homeobox 5 (DLX5), osteopontin (OPN), osterix (Osx), and runt-related transcription factor 2 (Runx2). Matrix mineralization was examined by Alizarin Red S staining. rDFSCs proliferation was analyzed using the Cell Counting Kit-8 assay. Subcutaneous implantation of rDFSCs-containing Matrigel scaffolds was used, and micro-computed tomography analysis, histological evaluation, and trichrome staining of implants extracted at 6 weeks were performed. We found that BMP9 enhanced alkaline phosphatase activity and mineralization in rDFSCs. The expression of bone-related genes (DLX5, OPN, Osx, and Runx2) was also increased as a result of BMP9 stimulation. Micro-computed tomography analysis and histological evaluation revealed that the bone masses retrieved from BMP9-overexpressing rDFSCs were significantly more pronounced in those with than in those without Matrigel. Our results suggest that BMP9 effectively promote osteogenic differentiation of rDFSCs, and Matrigel facilitate BMP9-induced osteogenesis of rDFSCs in vivo.
Collapse
Affiliation(s)
- Tiwei Fu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Panpan Liang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jinlin Song
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jinhua Wang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Pengfei Zhou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yinhong Tang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jing Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Enyi Huang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, P.R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|