1
|
Müller E, Schoberwalter T, Mader K, Seitz JM, Kopp A, Baranowsky A, Keller J. The Biological Effects of Magnesium-Based Implants on the Skeleton and Their Clinical Implications in Orthopedic Trauma Surgery. Biomater Res 2024; 28:0122. [PMID: 39717475 PMCID: PMC11665827 DOI: 10.34133/bmr.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Magnesium (Mg)-based implants have evolved as a promising innovation in orthopedic trauma surgery, with the potential to revolutionize the treatment of bone diseases, including osteoporotic fractures and bone defects. Available clinical studies mostly show excellent patient outcomes of resorbable Mg-based implants, without the need for subsequent implant removal. However, the occurrence of radiolucent zones around Mg-based implants seems to be a noticeable drawback for a more widespread clinical use. Mechanistically, both in vivo and in vitro studies demonstrated beneficial effects on the formation of new bone, a unique characteristic of Mg-based implants. In this regard, Mg has been shown to exert pleiotropic functions on osteogenic differentiation and migration of osteoblasts and their precursors. Additionally, collective evidence suggests that Mg-based implants promote angiogenesis in newly formed bone and exert immunomodulatory effects in the bone microenvironment. Likewise, Mg-based implants and their degradation products were shown to inhibit bone resorption by impairing osteoclastogenesis. The purpose of this review is to provide a state-of-the-art summary of the clinical and basic science evidence regarding the performance of currently used Mg-based implants. In addition to the status of in vivo and in vitro research and clinical applications, future challenges and perspectives of Mg-based orthopedic implants are discussed.
Collapse
Affiliation(s)
- Elena Müller
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Till Schoberwalter
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Konrad Mader
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | | - Alexander Kopp
- Medical Magnesium GmbH, 52068 Aachen, Germany
- Meotec GmbH, 52068 Aachen, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
2
|
Tian F, Zhao Y, Wang Y, Xu H, Liu Y, Liu R, Li H, Ning R, Wang C, Gao X, Luo R, Jia S, Zhu L, Hao D. Magnesium-Based Composite Calcium Phosphate Cement Promotes Osteogenesis and Angiogenesis for Minipig Vertebral Defect Regeneration. ACS Biomater Sci Eng 2024; 10:7577-7593. [PMID: 39575879 DOI: 10.1021/acsbiomaterials.4c01521] [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] [Indexed: 12/10/2024]
Abstract
Calcium phosphate cement (CPC) is an injectable bone cement with excellent biocompatibility, widely used for filling bone defects of various shapes. However, its slow degradation, insufficient mechanical strength, and poor osteoinductivity limit its further clinical applications. In this study, we developed a novel composite magnesium-based calcium phosphate cement by integrating magnesium microspheres into PLGA fibers obtained through wet spinning and incorporating these fibers into CPC. The inclusion of magnesium-based PLGA fibers enhanced the compressive strength and degradation rate of CPC, with the degradation rate of the magnesium microspheres being controllable to allow for the sustained release of magnesium ions. In vitro experiments showed that magnesium-based CPC enhanced the proliferation and migration of MC3T3-E1 and HUVECs. Additionally, the magnesium-based composite CPC not only enhanced osteogenic differentiation of MC3T3-E1 cells but also promoted angiogenesis in HUVECs. In vivo experiments using a vertebral bone defect model in Bama miniature pigs showed that the magnesium-based composite CPC significantly increased new bone formation. Additionally, compared to the CPC group, this composite exhibited significantly higher levels of osteogenic and angiogenic markers, with no inflammation or necrosis observed in the heart, liver, or kidneys, indicating good biocompatibility. These results suggest that magnesium-based composite CPC, with its superior compressive strength, biodegradability, and ability to promote vascularized bone regeneration, holds promise as a minimally invasive injectable material for bone regeneration.
Collapse
Affiliation(s)
- Fang Tian
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Yuqi Zhao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Yuhao Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Hailiang Xu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Youjun Liu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Renfeng Liu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Hui Li
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Ruojie Ning
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Chengwen Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Xinlin Gao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Rongjin Luo
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Shuaijun Jia
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Lei Zhu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| | - Dingjun Hao
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Youyi East Road No.555, Beilin District, Xi'an, Shaanxi 710001, China
| |
Collapse
|
3
|
Oladele IO, Adekola SA, Agbeboh NI, Isola-Makinde BA, Adewuyi BO. Synthesis and Application of Sustainable Tricalcium Phosphate Based Biomaterials From Agro-Based Materials: A Review. Biomed Eng Comput Biol 2024; 15:11795972241293525. [PMID: 39524096 PMCID: PMC11544672 DOI: 10.1177/11795972241293525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Trends in health care delivery systems have shifted as a result of the modern uses of biomaterials in medicine. Contrary to traditional medicine, modern healthcare are now useful in solving problems that were considered impossible some years back. One of the most significant factors to the most recent advancements in implant development has been the use of calcium based materials in the creation of necessary implants in the form of soft and hard tissues. With the advent of naturally sourced materials in the manufacturing of biomaterials, lots of attention are now focused on the different sources of agro-based resources that can be used for the product developments. These agro-based materials are now been considered for sustainable and ecological purposes in several areas of applications globally in the recent times. Hence, the review was carried out with focus on the sources, relevance, processing techniques and applications of tricalcium phosphate based biomaterials in modern day healthcare delivery. This review provides a historical and prospective picture of the crucial functions that materials based on tricalcium phosphate will play in fulfilling human requirements for medication.
Collapse
Affiliation(s)
- Isiaka Oluwole Oladele
- Department of Metallurgical and Materials Engineering, Federal University of Technology, Akure, Ondo, Nigeria
| | - Samson Ademola Adekola
- Department of Metallurgical and Materials Engineering, Federal University of Technology, Akure, Ondo, Nigeria
- Biomedical Engineering Department, Achievers University Owo, Ondo Sate, Nigeria
| | - Newton Itua Agbeboh
- Department of Metallurgical and Materials Engineering, Federal University of Technology, Akure, Ondo, Nigeria
- Department of Mechanical and Mechatronics Engineering, Federal University Otuoke, Otuoke, Bayelsa State, Nigeria
| | | | - Benjamin Omotayo Adewuyi
- Department of Metallurgical and Materials Engineering, Federal University of Technology, Akure, Ondo, Nigeria
| |
Collapse
|
4
|
Zhang Z, He D, Wang X, Ma X, Zheng Y, Gu X, Li Y. In vitro and in vivo evaluation of osteogenesis and antibacterial activity of MgGa alloys. Acta Biomater 2024; 185:85-97. [PMID: 39025394 DOI: 10.1016/j.actbio.2024.07.021] [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: 05/13/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
MgGa alloys are considered highly potential biodegradable materials, owing to its good mechanical properties and appropriate corrosion resistance. However, it is still far from application due to the lack of biological evaluation. In the present study, biocompatibility, osteogenesis and antibacterial activity of extruded Mg-xGa (x = 1 and 5 wt%) alloys were investigated by in vitro cell culture experiments and in vivo implantation. The cell adhesion and proliferation of osteoblast precursor cells (MC3T3-E1) showed the excellent cytocompatibility of Mg-1Ga and poor cytocompatibility of Mg-5Ga. The osteogenic activity was evaluated and revealed that Ga3+ in the Mg-1Ga extract had the ability to enhance osteogenic differentiation through the facilitation of its early stages. In vivo studies in a rat femoral condyle model revealed that both Mg-1Ga and Mg-5Ga significantly promoted new bone formation without causing any adverse effects. Mg-5Ga exhibited a much higher corrosion rate in vivo than Mg-1Ga. Its osteogenic activity was better due to the rapid release of Mg2+ and Ga3+, but this caused premature structural integrity loss. Mg-1Ga and Mg-5Ga released Ga3+ to inhibit E. coli and S. aureus, with antibacterial rate increasing with Ga content. Our studies demonstrate that Mg-Ga alloys have the potential to be used as osteogenic and antibacterial implant materials. STATEMENT OF SIGNIFICANCE: This study evaluates the biocompatibility, osteogenesis, and antibacterial activity of Mg-Ga alloys, which are promising biodegradable materials for medical applications. The study finds that Mg-1Ga exhibits excellent cytocompatibility and promotes osteogenic differentiation, facilitating the early stages of osteoblast precursor cell development. In vivo studies in a rat femoral condyle model reveal that Mg-1Ga and Mg-5Ga significantly promote new bone formation without causing any adverse effects. The antibacterial activity of both alloys is evaluated against E. coli and S. aureus, with the inhibition rate increasing with Ga content. These findings suggest that Mg-Ga alloys have the potential to serve as osteogenic and antibacterial implant materials, providing significant insights into the development of novel biomedical implants.
Collapse
Affiliation(s)
- Ziyue Zhang
- School of Materials Science and Engineering, Beihang University, Beijing 100191, China; Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
| | - Donglei He
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China; State Key Laboratory of Tribology, Tsinghua University, Beijing, China
| | - Xueying Wang
- School of Materials Science and Engineering, Beihang University, Beijing 100191, China
| | - Xiaolong Ma
- School of Materials Science and Engineering, Beihang University, Beijing 100191, China
| | - Yang Zheng
- School of Aeronautics and Astronautics, Tiangong University, Tianjin 300387, China.
| | - Xuenan Gu
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Yan Li
- School of Materials Science and Engineering, Beihang University, Beijing 100191, China; Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China; Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
5
|
Huang Y, Xu Y, Huang Z, Mao J, Hui Y, Rui M, Jiang X, Wu J, Ding Z, Feng Y, Gu Y, Chen L. Melatonin and calcium phosphate crystal-loaded poly(L-lactic acid) porous microspheres reprogram macrophages to improve bone repair. J Mater Chem B 2024. [PMID: 38940905 DOI: 10.1039/d3tb02965d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
The bone immune microenvironment can influence the occurrence and progression of bone defects. To date, research on promoting macrophage M2 polarization to improve bone injury repair has been insufficient. In this study, we designed an injectable poly(L-lactic acid) (PLLA) porous microsphere that forms calcium phosphate crystals on its surface by binding to melatonin, followed by bionanomimetic mineralization in vitro. The microsphere is injectable and degradable, and its release of melatonin (MT) and calcium phosphate (CaP) crystals promotes macrophage M2 polarization, reprogramming of macrophages, and enhanced osteogenesis. After LPS stimulation, the proportion of M2-polarized macrophages in the MS@CaP@MT group was 39.2 ± 2.7%, significantly higher than that in other groups (P < 0.05). Further, in the MS@CaP@MT group, rats exhibited bone mineral densities of 129.4 ± 12.8 mg cc-1 at 2 weeks and 171.6 ± 13.6 mg cc-1 at 4 weeks in the defect area, which were significantly higher than those in other groups (P < 0.05). Using an animal model of femoral condylar defects, we demonstrated that MT PLLA porous microspheres loaded with calcium phosphate crystals can improve the immune microenvironment and form a microsphere-centered osteogenesis model. This significantly accelerates bone defect repair and provides a potential strategy for bone defect treatment.
Collapse
Affiliation(s)
- Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Ziyan Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Jiannan Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
- Department of Orthopaedics, Jiangyin Clinical College, Xuzhou Medical University, No. 163 Shoushan Road, Jiang Yin 214400, China
| | - Yujian Hui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
- Department of Orthopaedics, Jiangyin Clinical College, Xuzhou Medical University, No. 163 Shoushan Road, Jiang Yin 214400, China
| | - Min Rui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
- Department of Orthopaedics, Jiangyin Clinical College, Xuzhou Medical University, No. 163 Shoushan Road, Jiang Yin 214400, China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Jie Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Zhouye Ding
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Yu Feng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China.
| |
Collapse
|
6
|
Tanvir MAH, Khaleque MA, Kim GH, Yoo WY, Kim YY. The Role of Bioceramics for Bone Regeneration: History, Mechanisms, and Future Perspectives. Biomimetics (Basel) 2024; 9:230. [PMID: 38667241 PMCID: PMC11048714 DOI: 10.3390/biomimetics9040230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Osteoporosis is a skeletal disorder marked by compromised bone integrity, predisposing individuals, particularly older adults and postmenopausal women, to fractures. The advent of bioceramics for bone regeneration has opened up auspicious pathways for addressing osteoporosis. Research indicates that bioceramics can help bones grow back by activating bone morphogenetic protein (BMP), mitogen-activated protein kinase (MAPK), and wingless/integrated (Wnt)/β-catenin pathways in the body when combined with stem cells, drugs, and other supports. Still, bioceramics have some problems, such as not being flexible enough and prone to breaking, as well as difficulties in growing stem cells and discovering suitable supports for different bone types. While there have been improvements in making bioceramics better for healing bones, it is important to keep looking for new ideas from different areas of medicine to make them even better. By conducting a thorough scrutiny of the pivotal role bioceramics play in facilitating bone regeneration, this review aspires to propel forward the rapidly burgeoning domain of scientific exploration. In the end, this appreciation will contribute to the development of novel bioceramics that enhance bone regrowth and offer patients with bone disorders alternative treatments.
Collapse
Affiliation(s)
| | | | | | | | - Young-Yul Kim
- Department of Orthopedic Surgery, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943, Republic of Korea; (M.A.H.T.); (M.A.K.); (G.-H.K.); (W.-Y.Y.)
| |
Collapse
|
7
|
Yang M, Chen C, Wang D, Shao Y, Zhou W, Shuai C, Yang Y, Ning X. Biomedical rare-earth magnesium alloy: Current status and future prospects. JOURNAL OF MAGNESIUM AND ALLOYS 2024; 12:1260-1282. [DOI: 10.1016/j.jma.2024.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
8
|
Luo X, Li M, Cai H, Yang S, Hao L, Ebel T, Helmholz H, Huang B, Wang X. Degradation Adaptability Assessment of Semisolid Powder Molded Mg-Zn-Mn Alloys for Orthopedic Applications. ACS APPLIED BIO MATERIALS 2023; 6:5515-5530. [PMID: 37982492 DOI: 10.1021/acsabm.3c00710] [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] [Indexed: 11/21/2023]
Abstract
Semisolid powder molding was used to prepare the medical Mg-6Zn alloy; in order to further improve its degradation adaptability, 0.5 and 1 wt % Mn were added. Then, the effect of the forming temperature (540, 560, 580, and 600 °C) on the in vitro degradation behavior of the prepared Mg-6Zn-xMn (x = 0.5, 1 wt %) was analyzed, and the optimized alloy was obtained. Finally, the biocompatibility and in vivo degradation performance of the optimized and Mn-free alloys were evaluated. Importantly, single-photon emission tomographic imaging (SPECT/CT) was first applied to monitor the in vivo degradation process. The results show that the corrosion mechanism of the Mn-free alloy is microgalvanic corrosion control with corrosive pitting. After adding Mn, the in vitro degradation rate decreases by half (0.17 ± 0.01 mm/year) as the forming temperature increases to 600 °C, and Mg-6Zn-1Mn prepared at 600 °C is the optimized alloy. Mn addition improves the corrosion product film protection and discontinuous secondary phases, and thus, the corrosion mechanism is changed to corrosive pitting control. Additionally, semisolid powder molding is an easy method to prepare alloys with low average pore interconnectivity (<10%), which is helpful for slowing down the degradation rate. The Mn-containing alloy has better biocompatibility, with a cytotoxicity of grade 0-1, due to its lower degradation rate. The in vivo corrosion rate of the Mn-free alloy is 0.19 mm/year after 28 days of implantation, which was precisely detected by SPECT/CT in real-time. The long-term in vivo degradation adaptability of Mn-free and Mn-containing alloys was not correctly presented, which may be due to the unreasonable bone defect model causing implant displacement. However, both of these alloys cause no obvious inflammation and show good healing. In summary, semisolid powder molding is a potentially promising technique to prepare medical Mg alloys, and nuclear imaging is an effective in vivo degradation evaluation method.
Collapse
Affiliation(s)
- Xia Luo
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, P. R. China
| | - Mingyu Li
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, P. R. China
- Department of Nuclear Medicine & Laboratary of Clinical Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Huawei Cai
- Department of Nuclear Medicine & Laboratary of Clinical Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Shanghui Yang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, P. R. China
| | - Lijing Hao
- National Engineering Research Center for Tissue Restoration and Reconstruction, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Thomas Ebel
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon GmbH, 21502 Geesthacht, Germany
| | - Heike Helmholz
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon GmbH, 21502 Geesthacht, Germany
| | - Bensheng Huang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, P. R. China
| | - Xiaohong Wang
- School of New Energy and Materials, Southwest Petroleum University, Chengdu 610500, P. R. China
| |
Collapse
|
9
|
Manescu (Paltanea) V, Antoniac I, Antoniac A, Laptoiu D, Paltanea G, Ciocoiu R, Nemoianu IV, Gruionu LG, Dura H. Bone Regeneration Induced by Patient-Adapted Mg Alloy-Based Scaffolds for Bone Defects: Present and Future Perspectives. Biomimetics (Basel) 2023; 8:618. [PMID: 38132557 PMCID: PMC10742271 DOI: 10.3390/biomimetics8080618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Treatment of bone defects resulting after tumor surgeries, accidents, or non-unions is an actual problem linked to morbidity and the necessity of a second surgery and often requires a critical healthcare cost. Although the surgical technique has changed in a modern way, the treatment outcome is still influenced by patient age, localization of the bone defect, associated comorbidities, the surgeon approach, and systemic disorders. Three-dimensional magnesium-based scaffolds are considered an important step because they can have precise bone defect geometry, high porosity grade, anatomical pore shape, and mechanical properties close to the human bone. In addition, magnesium has been proven in in vitro and in vivo studies to influence bone regeneration and new blood vessel formation positively. In this review paper, we describe the magnesium alloy's effect on bone regenerative processes, starting with a short description of magnesium's role in the bone healing process, host immune response modulation, and finishing with the primary biological mechanism of magnesium ions in angiogenesis and osteogenesis by presenting a detailed analysis based on a literature review. A strategy that must be followed when a patient-adapted scaffold dedicated to bone tissue engineering is proposed and the main fabrication technologies are combined, in some cases with artificial intelligence for Mg alloy scaffolds, are presented with examples. We emphasized the microstructure, mechanical properties, corrosion behavior, and biocompatibility of each study and made a basis for the researchers who want to start to apply the regenerative potential of magnesium-based scaffolds in clinical practice. Challenges, future directions, and special potential clinical applications such as osteosarcoma and persistent infection treatment are present at the end of our review paper.
Collapse
Affiliation(s)
- Veronica Manescu (Paltanea)
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (G.P.); (I.V.N.)
| | - Iulian Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, RO-050094 Bucharest, Romania
| | - Aurora Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
| | - Dan Laptoiu
- Department of Orthopedics and Trauma I, Colentina Clinical Hospital, 19-21 Soseaua Stefan cel Mare, RO-020125 Bucharest, Romania;
| | - Gheorghe Paltanea
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (G.P.); (I.V.N.)
| | - Robert Ciocoiu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
| | - Iosif Vasile Nemoianu
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (G.P.); (I.V.N.)
| | - Lucian Gheorghe Gruionu
- Faculty of Mechanics, University of Craiova, 13 Alexandru Ioan Cuza, RO-200585 Craiova, Romania;
| | - Horatiu Dura
- Faculty of Medicine, Lucian Blaga University of Sibiu, RO-550169 Sibiu, Romania;
| |
Collapse
|
10
|
Wen X, Wang J, Pei X, Zhang X. Zinc-based biomaterials for bone repair and regeneration: mechanism and applications. J Mater Chem B 2023; 11:11405-11425. [PMID: 38010166 DOI: 10.1039/d3tb01874a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Zinc (Zn) is one of the most important trace elements in the human body and plays a key role in various physiological processes, especially in bone metabolism. Zn-containing materials have been reported to enhance bone repair through promoting cell proliferation, osteogenic activity, angiogenesis, and inhibiting osteoclast differentiation. Therefore, Zn-based biomaterials are potential substitutes for traditional bone grafts. In this review, the specific mechanisms of bone formation promotion by Zn-based biomaterials were discussed, and recent developments in their application in bone tissue engineering were summarized. Moreover, the challenges and perspectives of Zn-based biomaterials were concluded, revealing their attractive potential and development directions in the future.
Collapse
Affiliation(s)
- Xinyu Wen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xin Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
11
|
Luo Y, Liu H, Zhang Y, Liu Y, Liu S, Liu X, Luo E. Metal ions: the unfading stars of bone regeneration-from bone metabolism regulation to biomaterial applications. Biomater Sci 2023; 11:7268-7295. [PMID: 37800407 DOI: 10.1039/d3bm01146a] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
In recent years, bone regeneration has emerged as a remarkable field that offers promising guidance for treating bone-related diseases, such as bone defects, bone infections, and osteosarcoma. Among various bone regeneration approaches, the metal ion-based strategy has surfaced as a prospective candidate approach owing to the extensive regulatory role of metal ions in bone metabolism and the diversity of corresponding delivery strategies. Various metal ions can promote bone regeneration through three primary strategies: balancing the effects of osteoblasts and osteoclasts, regulating the immune microenvironment, and promoting bone angiogenesis. In the meantime, the complex molecular mechanisms behind these strategies are being consistently explored. Moreover, the accelerated development of biomaterials broadens the prospect of metal ions applied to bone regeneration. This review highlights the potential of metal ions for bone regeneration and their underlying mechanisms. We propose that future investigations focus on refining the clinical utilization of metal ions using both mechanistic inquiry and materials engineering to bolster the clinical effectiveness of metal ion-based approaches for bone regeneration.
Collapse
Affiliation(s)
- Yankun Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Emergency, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xian Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| |
Collapse
|
12
|
Zheng W, Wu D, Zhang Y, Luo Y, Yang L, Xu X, Luo F. Multifunctional modifications of polyetheretherketone implants for bone repair: A comprehensive review. BIOMATERIALS ADVANCES 2023; 154:213607. [PMID: 37651963 DOI: 10.1016/j.bioadv.2023.213607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Polyetheretherketone (PEEK) has emerged as a highly promising orthopedic implantation material due to its elastic modulus which is comparable to that of natural bone. This polymer exhibits impressive properties for bone implantation such as corrosion resistance, fatigue resistance, self-lubrication and chemical stability. Significantly, compared to metal-based implants, PEEK implants have mechanical properties that are closer to natural bone, which can mitigate the "stress shielding" effect in bone implantation. Nevertheless, PEEK is incapable of inducing osteogenesis due to its bio-inert molecular structure, thereby hindering the osseointegration process. To optimize the clinical application of PEEK, researchers have been working on promoting its bioactivity and endowing this polymer with beneficial properties, such as antibacterial, anti-inflammatory, anti-tumor, and angiogenesis-promoting capabilities. Considering the significant growth of research on PEEK implants over the past 5 years, this review aims to present a timely update on PEEK's modification methods. By highlighting the latest advancements in PEEK modification, we hope to provide guidance and inspiration for researchers in developing the next generation bone implants and optimizing their clinical applications.
Collapse
Affiliation(s)
- Wenzhuo Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dongxu Wu
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yankun Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiangrui Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
13
|
Lin H, Zhang L, Zhang Q, Wang Q, Wang X, Yan G. Mechanism and application of 3D-printed degradable bioceramic scaffolds for bone repair. Biomater Sci 2023; 11:7034-7050. [PMID: 37782081 DOI: 10.1039/d3bm01214j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Bioceramics have attracted considerable attention in the field of bone repair because of their excellent osteogenic properties, degradability, and biocompatibility. To resolve issues regarding limited formability, recent studies have introduced 3D printing technology for the fabrication of bioceramic bone repair scaffolds. Nevertheless, the mechanisms by which bioceramics promote bone repair and clinical applications of 3D-printed bioceramic scaffolds remain elusive. This review provides an account of the fabrication methods of 3D-printed degradable bioceramic scaffolds. In addition, the types and characteristics of degradable bioceramics used in clinical and preclinical applications are summarized. We have also highlighted the osteogenic molecular mechanisms in biomaterials with the aim of providing a basis and support for future research on the clinical applications of degradable bioceramic scaffolds. Finally, new developments and potential applications of 3D-printed degradable bioceramic scaffolds are discussed with reference to experimental and theoretical studies.
Collapse
Affiliation(s)
- Hui Lin
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Liyun Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiyue Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Xue Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| | - Guangqi Yan
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| |
Collapse
|
14
|
Wang L, Li Y, Jiang S, Zhang Z, Zhao S, Song Y, Liu J, Tan F. Alginate hydrogels containing different concentrations of magnesium-containing poly(lactic-co-glycolic acid) microspheres for bone tissue engineering. Biomed Mater 2023; 18:055022. [PMID: 37478839 DOI: 10.1088/1748-605x/ace9a5] [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: 05/06/2023] [Accepted: 07/21/2023] [Indexed: 07/23/2023]
Abstract
The easy loss of crosslinking ions in alginate can result in structural collapse and loss of its characteristics as a bone scaffold. A novel injectable tissue engineering scaffold containing poly(lactic-co-glycolic acid) (PLGA) microspheres and alginate was fabricated to improve alginate's physiochemical and biological properties. MgCO3and MgO were loaded at a 1:1 ratio into PLGA microspheres to form biodegradable PLGA microspheres containing magnesium (PMg). Subsequently, different concentrations of PMg were mixed into a Ca2+suspension and employed as crosslinking agents for an alginate hydrogel. A pure Ca2+suspension was used as the alginate crosslinking agent in the control group. The influence of PMg on the physiochemical properties of the injectable scaffolds, including the surface morphology, degradation rate, Mg2+precipitation concentration, and the swelling rate, was investigated. MC3T3-E1 cells were seeded onto the hydrogels to evaluate the effect of the resultant alginate on osteoblastic attachment, proliferation, and differentiation. The physicochemical properties of the hydrogels, including morphology, degradation rate, and swelling ratio, were effectively tuned by PMg. Inductively coupled plasma-optical emission spectroscopy results showed that, in contrast to those in pure PMg, the magnesium ions (Mg2+) in alginate hydrogel containing PMg microspheres (Alg-PMg) were released in a dose-dependent and slow-releasing manner. Additionally, Alg-PMg with an appropriate concentration of PMg not only improved cell attachment and proliferation but also upregulated alkaline phosphatase activity, gene expression of osteogenic markers, and related growth factors. These findings indicate that PMg incorporation can regulate the physicochemical properties of alginate hydrogels. The resultant hydrogel promoted cell attachment, matrix mineralization, and bone regeneration. The hydrogel described in this study can be considered a promising injectable scaffold for bone tissue engineering.
Collapse
Affiliation(s)
- Lizhe Wang
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, People's Republic of China
- Qingdao Stomatological Hospital Affiliated to Qingdao University, 17 Dexian Road, Qingdao 266001, People's Republic of China
| | - Yaxin Li
- Qingdao Stomatological Hospital Affiliated to Qingdao University, 17 Dexian Road, Qingdao 266001, People's Republic of China
| | - Shuai Jiang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, 17 Dexian Road, Qingdao 266001, People's Republic of China
| | - Zhihao Zhang
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, People's Republic of China
| | - Sinan Zhao
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, People's Republic of China
| | - Yuru Song
- Huantai Country People Hospital, 2198 Huantai Road, Zibo 256400, People's Republic of China
| | - Jie Liu
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, People's Republic of China
| | - Fei Tan
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, People's Republic of China
| |
Collapse
|
15
|
Bao Z, Yang J, Shen J, Wang C, Li Y, Zhang Y, Yang G, Zhong C, Xu S, Xie L, Shen M, Gou Z. Core-shell bioceramic fiber-derived biphasic granules with adjustable core compositions for tuning bone regeneration efficacy. J Mater Chem B 2023; 11:2417-2430. [PMID: 36809396 DOI: 10.1039/d2tb02702j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Silicate-based biomaterials-clinically applied fillers and promising candidates-can act as a highly biocompatible substrate for osteostimulative osteogenic cell growth in vitro and in vivo. These biomaterials have been proven to exhibit a variety of conventional morphologies in bone repair, including scaffolds, granules, coatings and cement pastes. Herein, we aim to develop a series of novel bioceramic fiber-derived granules with core-shell structures which have a hardystonite (HT) shell layer and changeable core components-that is, the chemical compositions of a core layer can be tuned to include a wide range of silicate candidates (e.g., wollastonite (CSi)) with doping of functional ions (e.g., Mg, P, and Sr). Meanwhile, it is versatile to control the biodegradation and bioactive ion release sufficiently for stimulating new bone growth after implantation. Our method employs rapidly gelling ultralong core-shell CSi@HT fibers derived from different polymer hydrosol-loaded inorganic powder slurries through the coaxially aligned bilayer nozzles, followed by cutting and sintering treatments. It was demonstrated that the nonstoichiometric CSi core component could contribute to faster bio-dissolution and biologically active ion release in tris buffer in vitro. The rabbit femoral bone defect repair experiments in vivo indicated that core-shell bioceramic granules with an 8% P-doped CSi-core could significantly stimulate osteogenic potential favorable for bone repair. It is worth concluding that such a tunable component distribution strategy in fiber-type bioceramic implants may develop new-generation composite biomaterials endowed with time-dependent biodegradation and high osteostimulative activities for a range of bone repair applications in situ.
Collapse
Affiliation(s)
- Zhaonan Bao
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China.
| | - Jun Yang
- Department of Orthopaedic Surgery, Rui'an People's Hospital & the 3rd Hospital Affiliated to Wenzhou Medical University, Rui'an 325200, China
| | - Jian Shen
- Department of Orthopedics, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Cong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310008, China
| | - Yifan Li
- Department of Orthopedics, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Yan Zhang
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China.
| | - Guojing Yang
- Department of Orthopaedic Surgery, Rui'an People's Hospital & the 3rd Hospital Affiliated to Wenzhou Medical University, Rui'an 325200, China
| | - Cheng Zhong
- Department of Orthopedics, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Sanzhong Xu
- Department of Orthopedics, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Lijun Xie
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China.
| | - Miaoda Shen
- Department of Orthopedics, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Zhongru Gou
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
16
|
Yang Y, Chen M, Qiu Y, Li X, Huang Y, Zhang W. The Apelin/APLNR system modulates tumor immune response by reshaping the tumor microenvironment. Gene X 2022; 834:146564. [PMID: 35598689 DOI: 10.1016/j.gene.2022.146564] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 11/04/2022] Open
Abstract
Apelin is an endogenous ligand of the Apelin receptor (APLNR), a seven-transmembrane G protein-coupled receptor, which is widely distributed in human tissue. The Apelin/APLNR system is involved in regulating several physiological and pathological processes. The Apelin expression is increased in a variety of cancer and the Apelin/APLNR system could regulate the development of tumors through mediating autophagy, apoptosis, pyroptosis, and other biological processes to regulate tumor cell proliferation, migration, and invasion. The Apelin/APLNR system also participates in immune response and immune regulation through PI3K-Akt, ERK-MAPK, and other signal pathways. The latest research points out that there is a negative regulatory relationship between APLNR and immune checkpoint PD-L1. In this review, we outline the significance of the Apelin/APLNR signaling pathway in tumorigenesis and its immune regulation. These endeavors provide new insights into the translational application of Apelin/APLNR in cancer and may contribute to the promotion of more effective treatments for cancers.
Collapse
Affiliation(s)
- Yuqin Yang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Meilin Chen
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Yanbing Qiu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Xiaoxu Li
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Yumei Huang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Wenling Zhang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China.
| |
Collapse
|
17
|
Jing G, Yang L, Wang H, Niu J, Li Y, Wang S. Interference of layered double hydroxide nanoparticles with pathways for biomedical applications. Adv Drug Deliv Rev 2022; 188:114451. [PMID: 35843506 DOI: 10.1016/j.addr.2022.114451] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/18/2022] [Accepted: 07/09/2022] [Indexed: 11/01/2022]
Abstract
Recent decades have witnessed a surge of explorations into the application of multifarious materials, especially biomedical applications. Among them, layered double hydroxides (LDHs) have been widely developed as typical inorganic layer materials to achieve remarkable advancements. Multiple physicochemical properties endow LDHs with excellent merits in biomedical applications. Moreover, LDH nanoplatforms could serve as "molecular switches", which are capable of the controlled release of payloads under specific physiological pH conditions but are stable during circulation in the bloodstream. In addition, LDHs themselves are composed of several specific cations and possess favorable biological effects or regulatory roles in various cellular functions. These advantages have caused LDHs to become increasingly of interest in the area of nanomedicine. Recent efforts have been devoted to revealing the potential factors that interfere with the biological pathways of LDH-based nanoparticles, such as their applications in shaping the functions of immune cells and in determining the fate of stem cells and tumor treatments, which are comprehensively described herein. In addition, several intracellular signaling pathways interfering with by LDHs in the above applications were also systematically expatiated. Finally, the future development and challenges of LDH-based nanomedicine are discussed in the context of the ultimate goal of practical clinical application.
Collapse
Affiliation(s)
- Guoxin Jing
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Linnan Yang
- Central Laboratory, First Affiliated Hospital, Anhui Medical University, Hefei, PR China
| | - Hong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Jintong Niu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Youyuan Li
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Shilong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China.
| |
Collapse
|
18
|
Zhu Y, Zhao S, Cheng L, Lin Z, Zeng M, Ruan Z, Sun B, Luo Z, Tang Y, Long H. Mg 2+ -mediated autophagy-dependent polarization of macrophages mediates the osteogenesis of bone marrow stromal stem cells by interfering with macrophage-derived exosomes containing miR-381. J Orthop Res 2022; 40:1563-1576. [PMID: 34727384 DOI: 10.1002/jor.25189] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/02/2021] [Accepted: 09/30/2021] [Indexed: 02/04/2023]
Abstract
Magnesium ion (Mg2+ ) has received increased attention due to the roles it plays in promoting osteogenesis and preventing inflammation. This study was designed to investigate the mechanism by which Mg2+ influences the osteoblastic differentiation of bone marrow stromal stem cells (BMSCs). The polarization of Mø (macrophages) was measured after treatment with Mg2+ . Meanwhile, autophagy in Mø was measured by detecting LC3B expression. Mø-derived exosomes were isolated and cocultured with BMSCs; after which, osteogenic differentiation was evaluated by Alizarin Red staining and detection of alkaline phosphatase (ALP). Our results showed that Mg2+ could induce autophagy in macrophages and modulate the M1/M2 polarization of macrophages. Mg2+ -mediated macrophages could facilitate the osteogenic differentiation of BMSCs by regulating autophagy, and this facilitation by Mg2+ -mediated macrophages was closely related to macrophage-derived exosomes, and especially exosomes containing miR-381. However, miR-381 in macrophages did not influence autophagy or the polarization of Mg2+ -mediated macrophages. Furthermore, macrophage-derived exosomes containing miR-381 mainly determined the osteogenic differentiation of BMSCs. Mg2+ -mediated macrophages were shown to promote the osteogenic differentiation of BMSCs via autophagy through reducing miR-381 in macrophage-derived exosomes. In conclusion, our results suggest Mg2+ -mediated macrophage-derived exosomes containing miR-381 as novel vehicles for promoting the osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Shushan Zhao
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Liang Cheng
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Zhangyuan Lin
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Min Zeng
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Zhe Ruan
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Buhua Sun
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Zhongwei Luo
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Yifu Tang
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China.,Department of Orthopaedics, Xiangya Third Hospital of Central South University, Changsha, Hunan, PR China
| | - Haitao Long
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| |
Collapse
|
19
|
Li H, Wang C, Jin Y, Cai Y, Sun H, Liu M. The integrative analysis of competitive endogenous RNA regulatory networks in osteoporosis. Sci Rep 2022; 12:9549. [PMID: 35680981 PMCID: PMC9184474 DOI: 10.1038/s41598-022-13791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/27/2022] [Indexed: 11/09/2022] Open
Abstract
Osteoporosis (OP) is a common bone disease of old age resulting from the imbalance between bone resorption and bone formation. CircRNAs are a class of endogenous non-coding RNAs (ncRNAs) involved in gene regulation and may play important roles in the development of OP. Here, we aimed to discover the OP‑related circRNA-miRNA-mRNA (ceRNA) network and the potential mechanisms. Six microarray datasets were obtained from the GEO database and the OP‑related differentially expressed genes (DEGs), circRNAs (DECs), and miRNAs (DEMs) were screened out from these datasets. Then, combined with the prediction of the relationships between DEGs, DEMs, and DECs, a ceRNA network containing 7 target circRNAs, 5 target miRNAs, and 38 target genes was constructed. Then the RNA-seq verification by using total RNAs isolated from the femurs of normal and ovariectomized Wistar rats indicated that MFAP5, CAMK2A, and RGS4 in the ceRNA network were closely associated with osteoporosis. Function enrichment analysis indicated that the target circRNAs, miRNAs, and genes were involved in the process of MAPK cascade, hormone stimulus, cadherin binding, rRNA methyltransferase, PI3K-Akt signaling pathway, and Vitamin digestion and absorption, etc. Then a circRNA-miRNA-hub gene subnetwork was constructed and the qRT-PCR analysis of human bone tissues from the femoral head was used to confirm that the transcription of hsa_circR_0028877, hsa_circR_0082916, DIRAS2, CAMK2A, and MAPK4 showed a significant correlation with osteogenic genes. Besides, the two axes of hsa_circR_0028877/hsa-miR-1273f/CAMK2A and hsa_circR_0028877/hsa-miR-1273f/DIRAS2 conformed to be closely associated with OP. Additionally, by constructing a drug-target gene network, RKI-1447, FRAX486, Hyaluronic, and Fostamatinib were identified as therapeutic options for OP. Our study revealed the potential links between circRNAs, miRNAs, and mRNAs in OP, suggesting that the ceRNA mechanism might contribute to the occurrence of OP.
Collapse
Affiliation(s)
- Hao Li
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Yue Jin
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Yuanqing Cai
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China.
| | - Mozhen Liu
- Department of Orthopaedics, The First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China.
| |
Collapse
|
20
|
Connexin 43 affects thoracic ossification of ligamentum flavum by regulating the p38 MAPK-RUNX2 signaling pathway. Tissue Cell 2022; 76:101760. [PMID: 35220127 DOI: 10.1016/j.tice.2022.101760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/24/2022] [Accepted: 02/18/2022] [Indexed: 01/14/2023]
Abstract
This study aimed to investigate the role of connexin 43 (CX43) in thoracic ossification of ligamentum flavum (TOLF) based on the p38 mitogen-activated protein kinase (p38MAPK)-runt-related transcription factor 2 (RUNX2) pathway. Immunohistochemistry was used to detect CX43 expression in TOLF and non-TOLF patients, fibroblasts of TOLF were isolated and induced osteogenic differentiation, and CX43 expression was detected by western blot analysis (WB). In addition, si-CX43 was used to intervene CX43, and SB203580 was used to inhibit the p38MAPK. The expressions of bone differentiation marker protein were detected by WB, and the ossification ability was analyzed by alizarin red staining. The interaction between RUNX2 and CX43 was identified by dual-luciferase reporter assay. Results found that CX43 was highly expressed during TOLF, and si-CX43 could inhibit the expression of alkaline phosphatase (ALP) and osteopontin (OPN), as well as inhibit TOLF and the p38MAPK-RUNX2 pathway. In addition, SB203580 showed a synergistic effect with si-CX43 to further inhibit TOLF and the expression of RUNX2. The dual-luciferase reporter assay confirmed that RUNX2 could bind to the CX43 promoter. In conclusion, CX43 promotes TOLF, which may be mediated by p38MAPK-RUNX2, and RUNX2 binds to the CX43 promoter to form a positive feedback regulatory loop during TOLF.
Collapse
|
21
|
Osteoblastic microRNAs in skeletal diseases: Biological functions and therapeutic implications. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
22
|
Influence of Synthesis Conditions on Gadolinium-Substituted Tricalcium Phosphate Ceramics and Its Physicochemical, Biological, and Antibacterial Properties. NANOMATERIALS 2022; 12:nano12050852. [PMID: 35269340 PMCID: PMC8912835 DOI: 10.3390/nano12050852] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 01/27/2023]
Abstract
Gadolinium-containing calcium phosphates are promising contrast agents for various bioimaging modalities. Gadolinium-substituted tricalcium phosphate (TCP) powders with 0.51 wt% of gadolinium (0.01Gd-TCP) and 5.06 wt% of (0.1Gd-TCP) were synthesized by two methods: precipitation from aqueous solutions of salts (1) (Gd-TCP-pc) and mechano-chemical activation (2) (Gd-TCP-ma). The phase composition of the product depends on the synthesis method. The product of synthesis (1) was composed of β-TCP (main phase, 96%), apatite/chlorapatite (2%), and calcium pyrophosphate (2%), after heat treatment at 900 °C. The product of synthesis (2) was represented by β-TCP (main phase, 73%), apatite/chlorapatite (20%), and calcium pyrophosphate (7%), after heat treatment at 900 °C. The substitution of Ca2+ ions by Gd3+ in both β-TCP (main phase) and apatite (admixture) phases was proved by the electron paramagnetic resonance technique. The thermal stability and specific surface area of the Gd-TCP powders synthesized by two methods were significantly different. The method of synthesis also influenced the size and morphology of the prepared Gd-TCP powders. In the case of synthesis route (1), powders with particle sizes of tens of nanometers were obtained, while in the case of synthesis (2), the particle size was hundreds of nanometers, as revealed by transmission electron microscopy. The Gd-TCP ceramics microstructure investigated by scanning electron microscopy was different depending on the synthesis route. In the case of (1), ceramics with grains of 1–50 μm, pore sizes of 1–10 µm, and a bending strength of about 30 MPa were obtained; in the case of (2), the ceramics grain size was 0.4–1.4 μm, the pore size was 2 µm, and a bending strength of about 39 MPa was prepared. The antimicrobial activity of powders was tested for four bacteria (S. aureus, E. coli, S. typhimurium, and E. faecalis) and one fungus (C. albicans), and there was roughly 30% of inhibition of the micro-organism’s growth. The metabolic activity of the NCTC L929 cell and viability of the human dental pulp stem cell study demonstrated the absence of toxic effects for all the prepared ceramic materials doped with Gd ions, with no difference for the synthesis route.
Collapse
|
23
|
Tang W, Liu Q, Tan W, Sun T, Deng Y. LncRNA expression profile analysis of Mg 2+-induced osteogenesis by RNA-seq and bioinformatics. Genes Genomics 2021; 43:1247-1257. [PMID: 34427873 DOI: 10.1007/s13258-021-01140-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND In recent years, magnesium (Mg) has been extensively studied for manufacturing biodegradable orthopedic devices. Besides other advantages, researches have shown that magnesium-based implants can stimulate osteogenesis thus accelerating orthopedic trauma recovery, but its molecular mechanism is not fully understood. Meanwhile, long non-coding RNA (lncRNA) has been found to play vital role in regulating osteogenic differentiation. OBJECTIVE To explore the role of lncRNA in Mg2+ (magnesium ions)-induced osteogenesis. METHODS The effect of Mg2+ on mBMSCs proliferation was detected by the CCK-8 assay. The optimum concentration of Mg2+ (7.5 mM) in promoting mBMSCs osteogenesis was determined by ALP staining and Alizarin red staining, western blot and RT-qPCR were performed to detect osteogenic markers expressions. The lncRNAs and mRNAs expression profiles of mBMSCs were assessed by RNA-Seq and processed by bioinformatics analysis. The selected lncRNAs expression level was validated by RT-qPCR. RESULTS The effect of Mg2+ in promoting osteogenesis was confirmed and the optimum concentration was determined as 7.5 mM. The lncRNAs and mRNAs differentially expressed between 7.5 mM Mg2+-treated group and control group was detected and functional analysis revealed that their function were associated with osteogenesis. The ceRNA networks were constructed for H19 and Dubr that aberrantly expressed in two groups. The ceRNA networks of selected lncRNAs (H19 and Dubr) were constructed. CONCLUSIONS This study identified H19 and Dubr as osteogenic associated lncRNAs involved in Mg2+-induced osteogenesis, and they might play their roles through lncRNA-miRNA-mRNA axis.
Collapse
Affiliation(s)
- Wen Tang
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Qing Liu
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Wei Tan
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Tianshi Sun
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Youwen Deng
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
24
|
Yang Z, Zhuo Q, Qin W, Wang J, Wang L, Tien P. MicroRNAs miR-18a and miR-452 regulate the replication of enterovirus 71 by targeting the gene encoding VP3. Virus Genes 2021; 57:318-326. [PMID: 34002325 DOI: 10.1007/s11262-021-01842-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 05/05/2021] [Indexed: 11/29/2022]
Abstract
MicroRNAs (miRNAs) are crucial in the process of host-pathogen interaction. In this study, we established a screening system for miRNAs of target genes to detect the effect of miRNAs on Enterovirus 71 (EV71) replication in rhabdomyosarcoma (RD) cells. A 3'-untranslated region (UTR) dual-luciferase assay was performed to confirm putative miRNA targets in EV71 genome. Firstly, 13 fragments of EV71 genome were inserted into the vector pMIR, and luciferase activities were analyzed to identify the putative miRNAs of target genes. The expression of the reporter protein was significantly downregulated in cells transfected with the vector containing gene VP3. Then we screened for miRNAs that might target to VP3 through online analysis software. In addition, Western blot, real-time PCR, virus titration, and morphological changes were considered to examine the effects of miRNAs on virus replication. The results suggested that miR-18a and miR-452 repress the reproduction of EV71 virus by binding to VP3. Moreover, EV71 infection also affected the expression of endogenous miR-18a and miR-452. In addition, no significant cytotoxic effects were observed. The results from this study suggest that the intracellular miRNAs may play vital roles in the host-virus interaction.
Collapse
Affiliation(s)
- Zhuo Yang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China.
| | - Qin Zhuo
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Wen Qin
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Jingbo Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Liyuan Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Po Tien
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
25
|
Feng KC, Wu YJ, Wang CY, Tu CS, Lin YL, Chen CS, Lai PL, Huang YT, Chen PY. Enhanced mechanical and biological performances of CaO-MgO-SiO 2 glass-ceramics via the modulation of glass and ceramic phases. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112060. [PMID: 33947554 DOI: 10.1016/j.msec.2021.112060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/26/2021] [Accepted: 03/20/2021] [Indexed: 12/23/2022]
Abstract
This work reports a new CaO-MgO-SiO2 (CMS) bioactive glass-ceramic, using ZrO2 as a nucleus to modulate the ratios of glass and ceramic phases as a function of sintering temperature. Mg-rich bioactive CMS glass-ceramics exhibit advantages regarding mechanical strength (flexural strength ~190 MPa and compressive strength ~555 MPa), in-vitro and in-vivo biocompatibilities, and bone ingrowth. The high mechanical strengths could be attributed to the CaMgSi2O6 glass-ceramic and lower porosity. X-ray absorption spectra indicate an increased SiO covalent bond via the development of CaMgSi2O6 glass-ceramics. From the in-vitro cytotoxicity and BMSC differentiation assays, the CMS samples sintered above 800 °C exhibited better cell attachment and differentiation, possibly due to structural stability, appropriate pore, and ion release to boost osteogenesis. Compared to hydroxyapatite (HA) ceramics, the CMS glass-ceramics display higher mechanical strengths, biocompatibility, and osteoconductivity. An in-vivo experiment demonstrated a fine bone-ingrowth profile around the CMS implant. This study may further the application of CMS glass-ceramics in bone implants.
Collapse
Affiliation(s)
- Kuei-Chih Feng
- Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Yu-Jie Wu
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Chi-Yun Wang
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
| | - Chi-Shun Tu
- Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Physics, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Yu-Ling Lin
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Cheng-Sao Chen
- Department of Mechanical Engineering, Hwa Hsia University of Technology, New Taipei City 23567, Taiwan
| | - Po-Liang Lai
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan
| | - Yu-Tzu Huang
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Pin-Yi Chen
- Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan.
| |
Collapse
|
26
|
Peng W, Ren S, Zhang Y, Fan R, Zhou Y, Li L, Xu X, Xu Y. MgO Nanoparticles-Incorporated PCL/Gelatin-Derived Coaxial Electrospinning Nanocellulose Membranes for Periodontal Tissue Regeneration. Front Bioeng Biotechnol 2021; 9:668428. [PMID: 33842452 PMCID: PMC8026878 DOI: 10.3389/fbioe.2021.668428] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/02/2021] [Indexed: 12/19/2022] Open
Abstract
Electrospinning technique has attracted considerable attention in fabrication of cellulose nanofibrils or nanocellulose membranes, in which polycaprolactone (PCL) could be used as a promising precursor to prepare various cellulose nanofibril membranes for periodontal tissue regeneration. Conventional bio-membranes and cellulose films used in guided tissue regeneration (GTR) can prevent the downgrowth of epithelial cells, fibroblasts, and connective tissue in the area of tooth root but have limitations related to osteogenic and antimicrobial properties. Cellulose nanofibrils can be used as an ideal drug delivery material to encapsulate and carry some drugs. In this study, magnesium oxide (MgO) nanoparticles-incorporated PCL/gelatin core-shell nanocellulose periodontal membranes were fabricated using coaxial electrospinning technique, which was termed as Coaxial-MgO. The membranes using single-nozzle electrospinning technique, namely Blending-MgO and Blending-Blank, were used as control. The morphology and physicochemical property of these nanocellulose membranes were characterized by scanning electron microscopy (SEM), energy-dispersive spectrum of X-ray (EDS), transmission electron microscopy (TEM), contact angle, and thermogravimetric analysis (TGA). The results showed that the incorporation of MgO nanoparticles barely affected the morphology and mechanical property of nanocellulose membranes. Coaxial-MgO with core-shell fiber structure had better hydrophilic property and sustainable release of magnesium ion (Mg2+). CCK-8 cell proliferation and EdU staining demonstrated that Coaxial-MgO membranes showed better human periodontal ligament stem cells (hPDLSCs) proliferation rates compared with the other group due to its gelatin shell with great biocompatibility and hydrophilicity. SEM and immunofluorescence assay results illustrated that the Coaxial-MgO scaffold significantly enhanced hPDLSCs adhesion. In vitro osteogenic and antibacterial properties showed that Coaxial-MgO membrane enhanced alkaline phosphatase (ALP) activity, formation of mineralized nodules, osteogenic-related genes [ALP, collagen type 1 (COL1), runt-related transcription factor 2 (Runx2)], and high antibacterial properties toward Escherichia coli (E. coli) and Actinobacillus actinomycetemcomitans (A. a) when compared with controls. Our findings suggested that MgO nanoparticles-incorporated coaxial electrospinning PCL-derived nanocellulose periodontal membranes might have great prospects for periodontal tissue regeneration.
Collapse
Affiliation(s)
- Wenzao Peng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Shuangshuang Ren
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yibo Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ruyi Fan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xuanwen Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
27
|
Zou J, Du J, Tu H, Chen H, Cong K, Bi Z, Sun J. Resveratrol benefits the lineage commitment of bone marrow mesenchymal stem cells into osteoblasts via miR-320c by targeting Runx2. J Tissue Eng Regen Med 2021; 15:347-360. [PMID: 33481337 DOI: 10.1002/term.3176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 11/07/2022]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are a potential source of osteoblasts and have been widely used in clinical therapies due to their pluripotency. Recent publications have found that resveratrol (RSVL) played a crucial role in the proliferation and differentiation of BMSCs; however, the underlying molecular mechanism of RSVL-induced BMSCs osteogenic differentiation needs to be fully elucidated. The objective of this study was to explore functions of miRNAs in the RSVL-treated BMSCs and its effects on the differentiation potentials of BMSCs. The findings demonstrated that RSVL enhanced the osteogenesis and suppressed the adipogenesis of BMSCs in a dose-dependent manner. Besides, a novel regulatory axis containing miR-320c, and its target Runx2 was found during the differentiation process of BMSCs under RSVL treatment. Increase of miR-320c reduced the osteogenic potential of BMSCs, while knockdown of miR-320c played a positive role in the osteogenesis of BMSCs. In contrast, overexpression of miR-320c accelerated the adipogenic differentiation, while knockdown of miR-320c restrained the adipogenic differentiation of BMSCs. The results confirmed that Runx2 might be the direct target of miR-320c in RSVL-promoted osteogenic differentiation of BMSCs. This study revealed that RSVL might be used for the treatment of bone loss related diseases and miR-320c could be regarded as a novel and potential target to regulate the biological functions of BMSCs.
Collapse
Affiliation(s)
- Jilong Zou
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianyang Du
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hualei Tu
- Department of Burn, The Fifth Hospital in Harbin, Harbin, China
| | - Hongjun Chen
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kai Cong
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenggang Bi
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiabing Sun
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
28
|
Zhou W, Yan J, Li Y, Wang L, Jing L, Li M, Yu S, Cheng Y, Zheng Y. Based on the synergistic effect of Mg 2+ and antibacterial peptides to improve the corrosion resistance, antibacterial ability and osteogenic activity of magnesium-based degradable metals. Biomater Sci 2021; 9:807-825. [PMID: 33210105 DOI: 10.1039/d0bm01584a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
To overcome the restricted degradation, poor antiacterial and osteoindctive problems of magnesium and its alloys, this study presented the spinning coating of an antimicrobial peptide (AP)-loaded silk fibroin (SF) composite onto a corrosion-resistant MgO-coated AZ31 Mg alloy via anodization (aMgO) and electrodeposition (eMgO) methods. The composite coatings not only created a smooth and hydrophilic surface, but also obviously improved the corrosion resistance according to the test of corrosion potential and current density. The colonization of E. coli on MgO-AP composite coatings was significantly reduced as compared to the MgO layers, due to the potential synergetic effects of APs and Mg2+. Compared with the bare AZ31, the composite coating inhibited the corrosion of the substrate and the release of Mg2+, supported the adhesion, spreading and proliferation of osteoblasts, and presented a significantly improved osteogenic differentiation trend. Therefore, the MgO-AP composite coating, which had both antibacterial and bone-promoting abilities, was expected to be applied for surface modification of magnesium alloy implants to solve the clinical problems of bacterial infection and poor osseointegration.
Collapse
Affiliation(s)
- Wenhao Zhou
- Shaanxi Key Laboratory of biomedical metal materials, Northwest Institute for Non-ferrous Metal Research, Xi'an 710016, P R China.
| | - Jianglong Yan
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| | - Yangyang Li
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| | - Lan Wang
- Shaanxi Key Laboratory of biomedical metal materials, Northwest Institute for Non-ferrous Metal Research, Xi'an 710016, P R China.
| | - Lei Jing
- Shaanxi Key Laboratory of biomedical metal materials, Northwest Institute for Non-ferrous Metal Research, Xi'an 710016, P R China.
| | - Ming Li
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Sen Yu
- Shaanxi Key Laboratory of biomedical metal materials, Northwest Institute for Non-ferrous Metal Research, Xi'an 710016, P R China.
| | - Yan Cheng
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, China.
| |
Collapse
|
29
|
Sun Y, Liu X, Tan J, Lv D, Song W, Su R, Li L, Liu X, Ouyang L, Liao Y. Strontium ranelate incorporated 3D porous sulfonated PEEK simulating MC3T3-E1 cell differentiation. Regen Biomater 2021; 8:rbaa043. [PMID: 33732489 PMCID: PMC7947580 DOI: 10.1093/rb/rbaa043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 12/26/2022] Open
Abstract
Polyetheretherketone (PEEK) has been used as an implant material because it has similar mechanical properties to natural bone. However, inferior osseointegration and bioinertness hamper the clinical application of PEEK. In this study, the surfaces of sulfonated three-dimensional (3D) PEEK porous structures were loaded with different concentrations of strontium ranelate, a compound commonly used in the treatment or prevention of osteoporosis by promoting bone formation and inhibiting bone resorption. Field-emission scanning electron microscopy was used to characterize the topography of the structures, elemental carbon, oxygen and strontium contents were measured by X-ray photoelectron spectroscopy, and surface zeta potentials and water-contact angle were also measured. The results indicated that strontium ranelate was successfully loaded onto the 3D porous structures. In vitro cellular results showed that strontium ranelate-treated sulfonated PEEK (SP-SR) strengthened the adhesion of MC3T3-E1 cells. The activity of alkaline phosphatase, collagen secretion and extracellular matrix mineralization deposition of MC3T3-E1 cells were also improved on the surface of SP-SR. These results indicate that SP-SR could serve a new implant candidate for surgical treatment.
Collapse
Affiliation(s)
- Yingxiao Sun
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xingdan Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Changning District, Shanghai 200050, China
| | - Ji Tan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Changning District, Shanghai 200050, China
| | - Dan Lv
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Wengang Song
- Graduate School of Beihua University, Beihua University, Fengman District Jilin 132013, China
| | - Rui Su
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Ling Li
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Changning District, Shanghai 200050, China
| | - Liping Ouyang
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yun Liao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
30
|
He M, Gao X, Fan Y, Xie L, Yang M, Tian W. Tannic acid/Mg 2+-based versatile coating to manipulate the osteoimmunomodulation of implants. J Mater Chem B 2021; 9:1096-1106. [PMID: 33427278 DOI: 10.1039/d0tb01577f] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Instead of directly stimulating osteogenesis, endowing an implant surface with a favourable osteoimmunomodulatory (OIM) function has emerged as a new effective strategy to enhance osteointegration. Though metal-phenolic coatings have demonstrated to possess an immunomodulatory function, their potential application in manipulating an osteoimmune response has not been well explored. Herein, in order to develop a simple, rapid and universal coating method to impart excellent OIM to hard tissue implants, tannic acid (TA) and Mg2+ were selected to form a coating on Ti plate based on metal-phenolic chemistry. Besides its virtues of simplicity, ultrafastness, low-cost, and versatility, another merit for the coating method is that it can easily combine the unique functions of metal ions and phenolic ligands. The chelated Mg2+ can not only activate macrophage polarization towards the anti-inflammatory phenotype but also directly stimulate the osteogenic differentiation of bone marrow-derived stem cells (BMSCs). TA motifs rendered the coating with an excellent reactive oxygen species (ROS) scavenging capacity. TA and Mg2+ showed synergistic effects on regulating macrophage biological behaviour, suppressing its polarization towards the M1 phenotype, and promoting its polarization towards the M2 phenotype. In vivo histological analysis also demonstrated that the TA/Mg2+ coating could effectively inhibit the host response. Finally, the formed osteoimmune environment obviously enhanced the osteogenic differentiation of BMSCs. The above results demonstrated that the designed TA/Mg2+ coating not only possessed the function of directly stimulating osteogenesis but also the function of manipulating OIM to a desired one. Hence, it has great potential to be applied on advanced hard tissue implants to enhance osteointegration.
Collapse
Affiliation(s)
- Min He
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | | | | | | | | | | |
Collapse
|
31
|
Bosch-Rué E, Diez-Tercero L, Giordano-Kelhoffer B, Delgado LM, Bosch BM, Hoyos-Nogués M, Mateos-Timoneda MA, Tran PA, Gil FJ, Perez RA. Biological Roles and Delivery Strategies for Ions to Promote Osteogenic Induction. Front Cell Dev Biol 2021; 8:614545. [PMID: 33520992 PMCID: PMC7841204 DOI: 10.3389/fcell.2020.614545] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Bone is the most studied tissue in the field of tissue regeneration. Even though it has intrinsic capability to regenerate upon injury, several pathologies and injuries could hamper the highly orchestrated bone formation and resorption process. Bone tissue engineering seeks to mimic the extracellular matrix of the tissue and the different biochemical pathways that lead to successful regeneration. For many years, the use of extrinsic factors (i.e., growth factors and drugs) to modulate these biological processes have been the preferred choice in the field. Even though it has been successful in some instances, this approach presents several drawbacks, such as safety-concerns, short release profile and half-time life of the compounds. On the other hand, the use of inorganic ions has attracted significant attention due to their therapeutic effects, stability and lower biological risks. Biomaterials play a key role in such strategies where they serve as a substrate for the incorporation and release of the ions. In this review, the methodologies used to incorporate ions in biomaterials is presented, highlighting the osteogenic properties of such ions and the roles of biomaterials in controlling their release.
Collapse
Affiliation(s)
- Elia Bosch-Rué
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Leire Diez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Luis M. Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Begoña M. Bosch
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Mireia Hoyos-Nogués
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Phong A. Tran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Francisco Javier Gil
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Roman A. Perez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
32
|
Liu Q, Li M, Wang S, Xiao Z, Xiong Y, Wang G. Recent Advances of Osterix Transcription Factor in Osteoblast Differentiation and Bone Formation. Front Cell Dev Biol 2020; 8:601224. [PMID: 33384998 PMCID: PMC7769847 DOI: 10.3389/fcell.2020.601224] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
With increasing life expectations, more and more patients suffer from fractures either induced by intensive sports or other bone-related diseases. The balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption is the basis for maintaining bone health. Osterix (Osx) has long been known to be an essential transcription factor for the osteoblast differentiation and bone mineralization. Emerging evidence suggests that Osx not only plays an important role in intramembranous bone formation, but also affects endochondral ossification by participating in the terminal cartilage differentiation. Given its essentiality in skeletal development and bone formation, Osx has become a new research hotspot in recent years. In this review, we focus on the progress of Osx's function and its regulation in osteoblast differentiation and bone mass. And the potential role of Osx in developing new therapeutic strategies for osteolytic diseases was discussed.
Collapse
Affiliation(s)
- Qian Liu
- Key Laboratory of Brain and Neuroendocrine Diseases, College of Hunan Province, Hunan University of Medicine, Huaihua, China
- Biomedical Research Center, Hunan University of Medicine, Huaihua, China
| | - Mao Li
- Biomedical Research Center, Hunan University of Medicine, Huaihua, China
| | - Shiyi Wang
- XiangYa School of Medicine, Central South University, Changsha, China
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yuanyuan Xiong
- Key Laboratory of Brain and Neuroendocrine Diseases, College of Hunan Province, Hunan University of Medicine, Huaihua, China
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guangwei Wang
- Key Laboratory of Brain and Neuroendocrine Diseases, College of Hunan Province, Hunan University of Medicine, Huaihua, China
- Biomedical Research Center, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
33
|
Xu C, Liu H, He Y, Li Y, He X. Endothelial progenitor cells promote osteogenic differentiation in co-cultured with mesenchymal stem cells via the MAPK-dependent pathway. Stem Cell Res Ther 2020; 11:537. [PMID: 33308309 PMCID: PMC7731475 DOI: 10.1186/s13287-020-02056-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The role of bone tissue engineering is to regenerate tissue using biomaterials and stem cell-based approaches. Combination of two or more cell types is one of the strategies to promote bone formation. Endothelial progenitor cells (EPCs) may enhance the osteogenic properties of mesenchymal stem cells (MSCs) and promote bone healing; this study aimed to investigate the possible mechanisms of EPCs on promoting osteogenic differentiation of MSCs. METHODS MSCs and EPCs were isolated and co-cultured in Transwell chambers, the effects of EPCs on the regulation of MSC biological properties were investigated. Real-time PCR array, and western blotting were performed to explore possible signaling pathways involved in osteogenesis. The expression of osteogenesis markers and calcium nodule formation was quantified by qRT-PCR, western blotting, and Alizarin Red staining. RESULTS Results showed that MSCs exhibited greater alkaline phosphatase (ALP) activity and increased calcium mineral deposition significantly when co-cultured with EPCs. The mitogen-activated protein kinase (MAPK) signaling pathway was involved in this process. p38 gene expression and p38 protein phosphorylation levels showed significant upregulation in co-cultured MSCs. Silencing expression of p38 in co-cultured MSCs reduced osteogenic gene expression, protein synthesis, ALP activity, and calcium nodule formation. CONCLUSIONS These data suggest paracrine signaling from EPCs influences the biological function and promotes MSCs osteogenic differentiation. Activation of the p38MAPK pathway may be the key to enhancing MSCs osteogenic differentiation via indirect interactions with EPCs.
Collapse
Affiliation(s)
- Chu Xu
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China.,Department of General Dentistry, School of Stomatology, China Medical University, Shenyang, 110001, Liaoning, China
| | - Haijie Liu
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China
| | - Yuanjia He
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China
| | - Yuanqing Li
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China
| | - Xiaoning He
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China.
| |
Collapse
|
34
|
Jiang K, Teng GD, Chen YQ. MicroRNA-23 suppresses osteogenic differentiation of human bone marrow mesenchymal stem cells by targeting the MEF2C-mediated MAPK signaling pathway. J Gene Med 2020; 22:e3216. [PMID: 32410261 DOI: 10.1002/jgm.3216] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The present study aimed to determine the role and mechanism of miR-23 with respect to regulating the osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs). MATERIALS The expression of miR-23 and MEF2C was measured in osteoporosis (OP) patients and healthy controls by a quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). The correlation between miR-23 and MEF2C was determined by the Pearson correlation coefficient. Moreover, bioinformatic analysis was performed using public databases. Target gene function and potential pathways were further examined. Then, we used a miR-23 mimic or inhibitor to further explore the potential mechanism of miR-23. RESULTS miR-23 is found to be up-regulated and MEF2C is down-regulated in OP patients compared to healthy controls. miR-23 had a negative correlation with MEF2C (r = -0.937, p = 0.001). Bioinformatic analysis revealed that a total of 664 overlapping target genes were found in the TargetScan (http://www.targetscan.org), miRDB (http://mirdb.org) and miRanda (http://www.microrna.org/microrna/home.do) databases. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that miR-23 may regulate the mitogan-activated protein kinase (MAPK) signaling pathway. miR-23 is down-regulated and MEF2C is significantly up-regulated in the osteogenic differentiation of hBMSCs. MEF2C was significantly up-regulated in the osteogenic differentiation of hBMSCs. Overexpression of miR-23 significantly down-regulated alkaline phosphatase (ALP) activity and calcium deposition, whereas the miR-23 inhibitor had the opposite effects. Moreover, overexpression of miR-23 significantly decreased osteoblast-related markers (Runx2, Osx, ALP and OCN). Further experiments confirmed that MEF2C is a direct target of miR-23. Moreover, the miR-23 mimic enhanced the expression of p-p38 but had no effect on p-JNK. CONCLUSIONS miR-23 decreases the osteogenic differentiation of hBMSCs through the MEF2C/MAPK signaling pathway.
Collapse
Affiliation(s)
- Kai Jiang
- Hand Surgery, 971th Hospital of PLA, Qingdao, Shandong, China
| | - Guo-Dong Teng
- Hand Surgery, 971th Hospital of PLA, Qingdao, Shandong, China
| | - Yan-Qing Chen
- Hand Surgery, 971th Hospital of PLA, Qingdao, Shandong, China
| |
Collapse
|