1
|
Chen Y, Luo Z, Meng W, Liu K, Chen Q, Cai Y, Ding Z, Huang C, Zhou Z, Jiang M, Zhou L. Decoding the "Fingerprint" of Implant Materials: Insights into the Foreign Body Reaction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310325. [PMID: 38191783 DOI: 10.1002/smll.202310325] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Foreign body reaction (FBR) is a prevalent yet often overlooked pathological phenomenon, particularly within the field of biomedical implantation. The presence of FBR poses a heavy burden on both the medical and socioeconomic systems. This review seeks to elucidate the protein "fingerprint" of implant materials, which is generated by the physiochemical properties of the implant materials themselves. In this review, the activity of macrophages, the formation of foreign body giant cells (FBGCs), and the development of fibrosis capsules in the context of FBR are introduced. Additionally, the relationship between various implant materials and FBR is elucidated in detail, as is an overview of the existing approaches and technologies employed to alleviate FBR. Finally, the significance of implant components (metallic materials and non-metallic materials), surface CHEMISTRY (charge and wettability), and physical characteristics (topography, roughness, and stiffness) in establishing the protein "fingerprint" of implant materials is also well documented. In conclusion, this review aims to emphasize the importance of FBR on implant materials and provides the current perspectives and approaches in developing implant materials with anti-FBR properties.
Collapse
Affiliation(s)
- Yangmengfan Chen
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zeyu Luo
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weikun Meng
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiqing Chen
- Department of Ultrasound, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Yongrui Cai
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zichuan Ding
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chao Huang
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongke Zhou
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Jiang
- Emergency and Trauma Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liqiang Zhou
- MOE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| |
Collapse
|
2
|
Chen M, Ren M, Shi Y, Liu X, Wei H. State-of-the-art polyetheretherketone three-dimensional printing and multifunctional modification for dental implants. Front Bioeng Biotechnol 2023; 11:1271629. [PMID: 37929192 PMCID: PMC10621213 DOI: 10.3389/fbioe.2023.1271629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Polyetheretherketone (PEEK) is a high-performance thermoplastic polymer with an elastic modulus close to that of the jawbone. PEEK has the potential to become a new dental implant material for special patients due to its radiolucency, chemical stability, color similarity to teeth, and low allergy rate. However, the aromatic main chain and lack of surface charge and chemical functional groups make PEEK hydrophobic and biologically inert, which hinders subsequent protein adsorption and osteoblast adhesion and differentiation. This will be detrimental to the deposition and mineralization of apatite on the surface of PEEK and limit its clinical application. Researchers have explored different modification methods to effectively improve the biomechanical, antibacterial, immunomodulatory, angiogenic, antioxidative, osteogenic and anti-osteoclastogenic, and soft tissue adhesion properties. This review comprehensively summarizes the latest research progress in material property advantages, three-dimensional printing synthesis, and functional modification of PEEK in the fields of implant dentistry and provides solutions for existing difficulties. We confirm the broad prospects of PEEK as a dental implant material to promote the clinical conversion of PEEK-based dental implants.
Collapse
Affiliation(s)
- Meiqing Chen
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mei Ren
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yingqi Shi
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiuyu Liu
- Hospital of Stomatogy, Jilin University, Changchun, China
| | - Hongtao Wei
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Li W, Xu F, Dai F, Deng T, Ai Y, Xu Z, He C, Ai F, Song L. Hydrophilic surface-modified 3D printed flexible scaffolds with high ceramic particle concentrations for immunopolarization-regulation and bone regeneration. Biomater Sci 2023; 11:3976-3997. [PMID: 37115001 DOI: 10.1039/d3bm00362k] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Bioceramic scaffolds used in bone tissue engineering suffer from a low concentration of ceramic particles (<50 wt%), because the high concentration of ceramic particles increases the brittleness of the composite. 3D printed flexible PCL/HA scaffolds with high ceramic particle concentrations (84 wt%) were successfully fabricated in this study. However, the hydrophobicity of PCL weakens the composite scaffold hydrophilicity, which may limit the osteogenic ability to some extent. Thus, as a less time-consuming, less labour intensive, and more cost-effective treatment method, alkali treatment (AT) was employed to modify the surface hydrophilicity of the PCL/HA scaffold, and its regulation of immune responses and bone regeneration were investigated in vivo and in vitro. Initially, several concentrations of NaOH (0.5, 1, 1.5, 2, 2.5, and 5 mol L-1) were employed in tests to determine the appropriate concentration for AT. Based on the comprehensive consideration of the results of mechanical experiments and hydrophilicity, 2 mol L-1 and 2.5 mol L-1 of NaOH were selected for further investigation in this study. The PCL/HA-AT-2 scaffold dramatically reduced foreign body reactions as compared to the PCL/HA and PCL/HA-AT-2.5 scaffolds, promoted macrophage polarization towards the M2 phenotype and enhanced new bone formation. The Wnt/β-catenin pathway might participate in the signal transduction underlying hydrophilic surface-modified 3D printed scaffold-regulated osteogenesis, according to the results of immunohistochemical staining. In conclusion, hydrophilic surface-modified 3D printed flexible scaffolds with high ceramic particle concentrations can regulate the immune reactions and macrophage polarization to promote bone regeneration, and the PCL/HA-AT-2 scaffold is a potential candidate for bone tissue repair.
Collapse
Affiliation(s)
- Wenfeng Li
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Fancheng Xu
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Fang Dai
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Tian Deng
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Yufeng Ai
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Zhiyong Xu
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Chenjiang He
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| | - Fanrong Ai
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
- School of Advanced Manufacturing, Nanchang University, Nanchang, China.
| | - Li Song
- The Center of Stomatology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China.
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, China
| |
Collapse
|
4
|
Zeng X, Chen B, Wang L, Sun Y, Jin Z, Liu X, Ouyang L, Liao Y. Chitosan@Puerarin hydrogel for accelerated wound healing in diabetic subjects by miR-29ab1 mediated inflammatory axis suppression. Bioact Mater 2023; 19:653-665. [PMID: 35600974 PMCID: PMC9109129 DOI: 10.1016/j.bioactmat.2022.04.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Wound healing is one of the major global health concerns in patients with diabetes. Overactivation of pro-inflammatory M1 macrophages is associated with delayed wound healing in diabetes. miR-29ab1 plays a critical role in diabetes-related macrophage inflammation. Hence, inhibition of inflammation and regulation of miR-29 expression have been implicated as new points for skin wound healing. In this study, the traditional Chinese medicine, puerarin, was introduced to construct an injectable and self-healing chitosan@puerarin (C@P) hydrogel. The C@P hydrogel promoted diabetic wound healing and accelerated angiogenesis, which were related to the inhibition of the miR-29 mediated inflammation response. Compared to healthy subjects, miR-29a and miR-29b1 were ectopically increased in the skin wound of the diabetic model, accompanied by upregulated M1-polarization, and elevated levels of IL-1β and TNF-α. Further evaluations by miR-29ab1 knockout mice exhibited superior wound healing and attenuated inflammation. The present results suggested that miR-29ab1 is essential for diabetic wound healing by regulating the inflammatory response. Suppression of miR-29ab1 by the C@P hydrogel has the potential for improving medical approaches for wound repair. A chitosan based hydrogel containing puerarin was constructed for promoting diabetic wound healing. Chitosan@Puerarin hydrogel accelerated skin repair through inhibiting M1-polarization and reducing IL-1β and TNF-α. miR-29 a/b1 was found to be ectopic increased in the skin-wound of diabetic model. miR-29 a/b1 was inhibited by Chitosan@Puerarin in diabetic wound healing.
Collapse
|
5
|
Bai X, Wang J, Zhang X, Tang Y, He Y, Zhao J, Han L, Fang R, Liu Z, Dong H, Li Q, Ge J, Ma Y, Yu M, Sun R, Wang J, Fei J, Huang F. Deficiency of miR-29a/b1 leads to premature aging and dopaminergic neuroprotection in mice. Front Mol Neurosci 2022; 15:978191. [PMID: 36277485 PMCID: PMC9582353 DOI: 10.3389/fnmol.2022.978191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by progressive degeneration of midbrain dopaminergic neurons. The miR-29s family, including miR-29a and miR-29b1 as well as miR-29b2 and miR-29c, are implicated in aging, metabolism, neuronal survival, and neurological disorders. In this study, the roles of miR-29a/b1 in aging and PD were investigated. miR-29a/b1 knockout mice (named as 29a KO hereafter) and their wild-type (WT) controls were used to analyze aging-related phenotypes. After challenged with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), dopaminergic injuries, glial activation, and mouse behaviors were evaluated. Primary glial cells were further cultured to explore the underlying mechanisms. Additionally, the levels of miR-29s in the cerebrospinal fluid (CSF) of PD patients (n = 18) and healthy subjects (n = 17) were quantified. 29a KO mice showed dramatic weight loss, kyphosis, and along with increased and deepened wrinkles in skins, when compared with WT mice. Moreover, both abdominal and brown adipose tissues reduced in 29a KO mice, compared to their WT counterpart. However, in MPTP-induced PD mouse model, the deficiency of miR-29a/b1 led to less severe damages of dopaminergic system and mitigated glial activation in the nigrostriatal pathway, and subsequently alleviated the motor impairments in 3-month-old mice. Eight-month-old mutant mice maintained such a resistance to MPTP intoxication. Mechanistically, the deficiency of miR-29a/b-1 promoted the expression of neurotrophic factors in 1-Methyl-4-phenylpyridinium (MPP+)-treated primary mixed glia and primary astrocytes. In lipopolysaccharide (LPS)-treated primary microglia, knockout of miR-29a/b-1 inhibited the expression of inflammatory factors, and promoted the expression of anti-inflammatory factors and neurotrophic factors. Knockout of miR-29a/b1 increased the activity of AMP-activated protein kinase (AMPK) and repressed NF-κB/p65 signaling in glial cells. Moreover, we found miR-29a level was increased in the CSF of patients with PD. Our results suggest that 29a KO mice display the peripheral premature senility. The combined effects of less activated glial cells might contribute to the mitigated inflammatory responses and elicit resistance to MPTP intoxication in miR-29a/b1 KO mice.
Collapse
Affiliation(s)
- Xiaochen Bai
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jinghui Wang
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yilin Tang
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongtao He
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jiayin Zhao
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Linlin Han
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Rong Fang
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Zhaolin Liu
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Hongtian Dong
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Qing Li
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
- Shanghai Engineering Research Center for Model Organisms, SMOC, Shanghai, China
| | - Jingyu Ge
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Mei Yu
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Ruilin Sun
- Shanghai Engineering Research Center for Model Organisms, SMOC, Shanghai, China
| | - Jian Wang
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Jian Wang,
| | - Jian Fei
- Shanghai Engineering Research Center for Model Organisms, SMOC, Shanghai, China
- School of Life Science and Technology, Tongji University, Shanghai, China
- *Correspondence: Jian Fei,
| | - Fang Huang
- Department of Translational Neuroscience, MOE Frontiers Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
- Fang Huang,
| |
Collapse
|
6
|
Javaid S, Dey M, Matzke C, Gupta S. Synthesis and characterization of engineered
PEEK
‐based composites for enhanced tribological and mechanical performance. J Appl Polym Sci 2022. [DOI: 10.1002/app.52886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sabah Javaid
- Department of Mechanical Engineering University of North Dakota Grand Forks North Dakota USA
| | - Maharshi Dey
- Department of Mechanical Engineering University of North Dakota Grand Forks North Dakota USA
| | - Caleb Matzke
- Department of Mechanical Engineering University of North Dakota Grand Forks North Dakota USA
| | - Surojit Gupta
- Department of Mechanical Engineering University of North Dakota Grand Forks North Dakota USA
| |
Collapse
|
7
|
Chen J, Cao G, Li L, Cai Q, Dunne N, Li X. Modification of polyether ether ketone for the repairing of bone defects. Biomed Mater 2022; 17. [PMID: 35395651 DOI: 10.1088/1748-605x/ac65cd] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/08/2022] [Indexed: 11/12/2022]
Abstract
Bone damage as a consequence of disease or trauma is a common global occurrence. For bone damage treatment - bone implant materials are necessary across three classifications of surgical intervention (i.e. fixation, repair, and replacement). Many types of bone implant materials have been developed to meet the requirements of bone repair. Among them, polyether ether ketone (PEEK) has been considered as one of the next generation of bone implant materials, owing to its advantages related to good biocompatibility, chemical stability, X-ray permeability, elastic modulus comparable to natural bone, as well as the ease of processing and modification. However, as PEEK is a naturally bioinert material, some modification is needed to improve its integration with adjacent bones after implantation. Therefore, it has become a very hot topic of biomaterials research and various strategies for the modification of PEEK including blending, 3D printing, coating, chemical modification and the introduction of bioactive and/or antibacterial substances have been proposed. In this systematic review, the recent advances in modification of PEEK and its application prospect as bone implants are summarized, and the remaining challenges are also discussed.
Collapse
Affiliation(s)
- Junfeng Chen
- Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, Beijing, 100083, CHINA
| | - Guangxiu Cao
- Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, Beijing, 100083, CHINA
| | - Linhao Li
- Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, CHINA
| | - Qiang Cai
- Tsinghua University Department of Materials Science and Engineering, 30 shuangqing Rd, Haidian District, Beijing, Beijing, 100084, CHINA
| | - Nicholas Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Dublin, D09, IRELAND
| | - Xiaoming Li
- Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, Beijing, 100083, CHINA
| |
Collapse
|
8
|
Ouyang L, Sun Y, Lv D, Peng X, Liu X, Ci L, Zhang G, Yuan B, Li L, Fei J, Ma J, Liu X, Liao Y. miR-29cb2 promotes angiogenesis and osteogenesis by inhibiting HIF-3α in bone. iScience 2022; 25:103604. [PMID: 35005549 PMCID: PMC8718933 DOI: 10.1016/j.isci.2021.103604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
Coordination between osteogenesis and angiogenesis is required for bone homeostasis. Here, we show that miR-29cb2 is a bone-specific miRNA and plays critical roles on angiogenesis-osteogenesis coupling during bone remodeling. Mice with deletion of miR-29cb2 exhibit osteopenic phenotypes and osteoblast impairment, accompanied by pronounced decreases in specific H vessels. The decrease in bone miR-29cb2 was associated with pathological ovariectomy stimuli. Mechanistically, hypoxia-inducible factor (HIF)-3α, as a target for miR-29cb2, inhibits HIF-1α activity by competitively bonding with HIF-1β. Notably, miR-29cb2 in peripheral blood (PB) nearly is undetectable in sham and significantly increases in ovariectomy mice. Further evaluation from osteoporosis patients demonstrates similar signatures. ROC analysis shows miR-29cb2 in PB has higher sensitivity and specificity for diagnosing osteoporosis when compared with four clinical biomarkers. Collectively, these findings reveal that miR-29cb2 is essential for bone remodeling by inhibiting HIF-3α and elevated bone-specific miR-29cb2 in PB, which may be a promising biomarker for bone loss.
Collapse
Affiliation(s)
- Liping Ouyang
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
- Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yingxiao Sun
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Dan Lv
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xiaochun Peng
- Department of Orthopaedics, The Sixth Affiliated People's Hospital, Shanghai Jiaotong University, Shanghai 200233, China
| | - Xiaoming Liu
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Lei Ci
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai 201203, China
| | - Guoning Zhang
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Bo Yuan
- 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
| | - Jian Fei
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai 201203, China
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jun Ma
- Hongqiao International Institute of Medicine, 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, Shanghai 200050, China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yun Liao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
9
|
Liu X, Ouyang L, Chen L, Qiao Y, Ma X, Xu G, Liu X. Hydroxyapatite composited PEEK with 3D porous surface enhances osteoblast differentiation through mediating NO by macrophage. Regen Biomater 2021; 9:rbab076. [PMID: 35480864 PMCID: PMC9039504 DOI: 10.1093/rb/rbab076] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/21/2021] [Accepted: 11/30/2021] [Indexed: 11/14/2022] Open
Abstract
The adverse immune response mediated by macrophages is one of the main factors that are prone to lead poor osseointegration of polyetheretherketone (PEEK) implants in clinic. Hence, endowing PEEK with immunomodulatory ability to avoid the adverse immune response becomes a promising strategy to promote bone repair. In this work, sulfonation and hydrothermal treatment were used to fabricate a 3D porous surface on PEEK and hydroxyapatite (HA) composited PEEK. The HA composited PEEK with 3D porous surface inhibited macrophages polarizing to M1 phenotype and downregulated inducible nitric oxide synthase protein expression, which led to a nitric oxide concentration reduction in culture medium of mouse bone marrow mesenchymal stem cells (mBMSCs) under co-culture condition. The decrease of nitric oxide concentration could help to increase bone formation-related OSX and ALP genes expressions and decrease bone resorption-related MMP-9 and MMP-13 genes expressions via cAMP-PKA-RUNX2 pathway in mBMSCs. In summary, the HA composited PEEK with 3D porous surface has the potential to promote osteogenesis of PEEK through immunomodulation, which provides a promising strategy to improve the bone repair ability of PEEK.
Collapse
Affiliation(s)
- Xingdan Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China
| | - Liping Ouyang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Lan Chen
- School of Materials Science, and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), Zhengzhou University, Science Avenue 100, Zhengzhou 450001, China
| | - Yuqin Qiao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China
| | - Xiaohan Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, China
- Cixi Center of Biomaterials Surface Engineering, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Wenwei Road 345, Ningbo 315300, China
| | - Guohua Xu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai 200003, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China
- Cixi Center of Biomaterials Surface Engineering, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Wenwei Road 345, Ningbo 315300, China
| |
Collapse
|
10
|
Liu W, Feng Q, Liao W, Li E, Wu L. TUG1 promotes the expression of IFITM3 in hepatocellular carcinoma by competitively binding to miR-29a. J Cancer 2021; 12:6905-6920. [PMID: 34659578 PMCID: PMC8517998 DOI: 10.7150/jca.57477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/13/2021] [Indexed: 02/07/2023] Open
Abstract
Purpose: Numerous studies have demonstrated the important relationship of TUG1 with tumorigenesis. The present study investigated the role of TUG1 and its downstream genes miR-29a and IFITM3 in the occurrence and development of hepatocellular carcinoma (HCC). We found that both TUG1 and IFITM3 genes are highly expressed in HCC, whereas the expression of miR-29a is low in HCC. Downregulation of TUG1 reduces cell invasion, metastasis, and cell proliferation ability and promotes cell apoptosis. Simultaneous downregulation of miR-29a reverses this effect. Moreover, IFITM3, as the target gene of miR-29a, is positively regulated by TUG1. However, the adjustment relationship between these three components is still unknown and thus warrants further investigation. The objective of this study was to investigate the regulatory relationship between TUG1, miR-29a, and IFITM3 in human liver cancer. Patients and methods: The expression of TUG1 and miR-29a in tumor tissues and adjacent non-tumor tissues of 65 patients with HCC was detected by real-time quantitative polymerase chain reaction (RT-qPCR). The migration and invasion of liver cancer cells were studied by the wound healing assay and the Transwell method, respectively. The apoptosis rate of HCC cells was detected by flow cytometry, and the proliferation rate of hepatoma cells was detected by the 5-ethynyl-2'-deoxyuridine (EdU) method. Immunofluorescence was used to detect the expression of TUG1 and IFITM3 in HCC-LM3 and HL-7702 cell lines. The relationship between TUG1 and miR-29a was detected using a double luciferase reporter assay and fluorescence in situ hybridization (FISH). Tumors were established in vivo by subcutaneous injection of HCC cells into nude mice and injection of these cells into the tail vein. Western blotting was used to quantify the biomarkers. Results: The expression of TUG1 increased significantly in tumor tissues and HCC cells. Moreover, the expression of miR-29a in liver cancer tissues was significantly lower than that in normal human liver tissues. The expression of TUG1 in liver cancer tissue was negatively correlated with miR-29a. Knockdown of TUG1 weakened the invasion, migration, and proliferation of HCC cells, and enhanced their apoptosis. A simultaneous knockdown of miR-29a enhanced cell invasion, metastasis, and cell proliferation, whereas the apoptosis ability decreased. As a target gene of miR-29a, IFITM3 is not only negatively regulated by miR-29a, but also positively regulated by TUG1. Therefore, TUG1 regulates IFITM3 in HCC cells by competitively binding to miR-29a, thus affecting cell invasion, migration, proliferation, and apoptosis. Conclusion: As a CeRNA, TUG1 competitively binds to miR-29a to regulate IFITM3 and promote the development of liver cancer. Downregulation of TUG1 can significantly inhibit the migration, invasion, and proliferation of liver cancer cells. Based on these results, we conclude that TUG1 could serve as a key gene to improve the prognosis of patients with HCC.
Collapse
Affiliation(s)
| | | | | | - Enliang Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Nanchang University, 1 Mindle Road, Nanchang Jiangxi 330006, P.R. China
| | - Linquan Wu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Nanchang University, 1 Mindle Road, Nanchang Jiangxi 330006, P.R. China
| |
Collapse
|
11
|
Li M, Tian F, Jiang J, Zhou M, Chen Q, Zhao D, Zhai W. Robust and Multifunctional Porous Polyetheretherketone Fiber Fabricated via a Microextrusion CO 2 Foaming. Macromol Rapid Commun 2021; 42:e2100463. [PMID: 34490937 DOI: 10.1002/marc.202100463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/03/2021] [Indexed: 01/12/2023]
Abstract
Fabrication of multifunctional porous fibers with excellent mechanical properties has attracted abundant attention in the fields of personal thermal management textiles and smart wearable devices. However, the high cost and harsh preparation environment of the traditional solution-solvent phase separation method for making porous fibers aggravates the problems of resource consumption and environmental pollution. Herein, a microextrusion process that combines environmentally friendly CO2 physical foaming with fused deposition modeling technology is proposed, via the dual features of high gas uptake and restricted cell growth, to implement the continuous production of porous polyetheretherketone (PEEK) fibers with a production efficiency of 10.5 cm s-1 . The porous PEEK fiber exhibits excellent stretchability (234.8% strain) and good high-temperature thermal insulation property. The open-cell structure on the surface is favorable for the adsorption to achieve superhydrophobicity (154.4°) and high-efficiency photocatalytic degradation of rhodamine B (90.4%). Moreover, the parameterized controllability of the cell structure is beneficial to widening the multifunctional window. In short, the first porous PEEK physical foaming fiber, which opens up a new avenue for the application expansion, especially in the medical field, is realized.
Collapse
Affiliation(s)
- Mengya Li
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Fangwei Tian
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China.,College of Science, China University of Petroleum, Beijing, 102249, P. R. China
| | - Junjie Jiang
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China.,Ningbo Key Lab of Polymer Materials, Ningbo Institute of Material Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, P. R. China
| | - Mengnan Zhou
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Qiyuan Chen
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Dan Zhao
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Wentao Zhai
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China.,Sun Yat-sen University Nanchang Research Institute, Nanchang, 330224, P. R. China
| |
Collapse
|
12
|
Implant Fibrosis and the Underappreciated Role of Myofibroblasts in the Foreign Body Reaction. Cells 2021; 10:cells10071794. [PMID: 34359963 PMCID: PMC8304203 DOI: 10.3390/cells10071794] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Body implants and implantable medical devices have dramatically improved and prolonged the life of countless patients. However, our body repair mechanisms have evolved to isolate, reject, or destroy any object that is recognized as foreign to the organism and inevitably mounts a foreign body reaction (FBR). Depending on its severity and chronicity, the FBR can impair implant performance or create severe clinical complications that will require surgical removal and/or replacement of the faulty device. The number of review articles discussing the FBR seems to be proportional to the number of different implant materials and clinical applications and one wonders, what else is there to tell? We will here take the position of a fibrosis researcher (which, coincidentally, we are) to elaborate similarities and differences between the FBR, normal wound healing, and chronic healing conditions that result in the development of peri-implant fibrosis. After giving credit to macrophages in the inflammatory phase of the FBR, we will mainly focus on the activation of fibroblastic cells into matrix-producing and highly contractile myofibroblasts. While fibrosis has been discussed to be a consequence of the disturbed and chronic inflammatory milieu in the FBR, direct activation of myofibroblasts at the implant surface is less commonly considered. Thus, we will provide a perspective how physical properties of the implant surface control myofibroblast actions and accumulation of stiff scar tissue. Because formation of scar tissue at the surface and around implant materials is a major reason for device failure and extraction surgeries, providing implant surfaces with myofibroblast-suppressing features is a first step to enhance implant acceptance and functional lifetime. Alternative therapeutic targets are elements of the myofibroblast mechanotransduction and contractile machinery and we will end with a brief overview on such targets that are considered for the treatment of other organ fibroses.
Collapse
|
13
|
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
|
14
|
Guo Y, Wu Y, Shi J, Zhuang H, Ci L, Huang Q, Wan Z, Yang H, Zhang M, Tan Y, Sun R, Xu L, Wang Z, Shen R, Fei J. miR-29a/b1 Regulates the Luteinizing Hormone Secretion and Affects Mouse Ovulation. Front Endocrinol (Lausanne) 2021; 12:636220. [PMID: 34135859 PMCID: PMC8202074 DOI: 10.3389/fendo.2021.636220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
miR-29a/b1 was reportedly involved in the regulation of the reproductive function in female mice, but the underlying molecular mechanisms are not clear. In this study, female mice lacking miR-29a/b1 showed a delay in vaginal opening, irregular estrous cycles, ovulation disorder and subfertility. The level of luteinizing hormone (LH) was significantly lower in plasma but higher in pituitary of mutant mice. However, egg development was normal in mutant mice and the ovulation disorder could be rescued by the superovulation treatment. These results suggested that the LH secretion was impaired in mutant mice. Further studies showed that deficiency of miR-29a/b1 in mice resulted in an abnormal expression of a number of proteins involved in vesicular transport and exocytosis in the pituitary, indicating the mutant mice had insufficient LH secretion. However, the detailed mechanism needs more research.
Collapse
Affiliation(s)
- Yang Guo
- School of Life Science and Technology, Tongji University, Shanghai, China
- Shanghai Lab, Animal Research Center, Shanghai, China
| | - Youbing Wu
- Shanghai Model Organisms, Shanghai, China
| | - Jiahao Shi
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Hua Zhuang
- Shanghai Model Organisms, Shanghai, China
| | - Lei Ci
- School of Life Science and Technology, Tongji University, Shanghai, China
- Shanghai Model Organisms, Shanghai, China
| | - Qin Huang
- Shanghai Model Organisms, Shanghai, China
| | - Zhipeng Wan
- School of Life Science and Technology, Tongji University, Shanghai, China
- Shanghai Model Organisms, Shanghai, China
| | - Hua Yang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Mengjie Zhang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yutong Tan
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Ruilin Sun
- Shanghai Model Organisms, Shanghai, China
| | - Leon Xu
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Zhugang Wang
- Department of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ruling Shen
- School of Life Science and Technology, Tongji University, Shanghai, China
- Shanghai Lab, Animal Research Center, Shanghai, China
- *Correspondence: Jian Fei, ; Ruling Shen,
| | - Jian Fei
- School of Life Science and Technology, Tongji University, Shanghai, China
- Shanghai Model Organisms, Shanghai, China
- *Correspondence: Jian Fei, ; Ruling Shen,
| |
Collapse
|
15
|
Yu X, Yao S, Chen C, Wang J, Li Y, Wang Y, Khademhosseini A, Wan J, Wu Q. Preparation of Poly(ether-ether-ketone)/Nanohydroxyapatite Composites with Improved Mechanical Performance and Biointerfacial Affinity. ACS OMEGA 2020; 5:29398-29406. [PMID: 33225171 PMCID: PMC7676340 DOI: 10.1021/acsomega.0c04257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/19/2020] [Indexed: 06/11/2023]
Abstract
Poly(ether-ether-ketone) (PEEK) displays promising potential in hard tissue repair and orthopedic surgery due to its adaptable mechanical performance, good chemical resistance, and bioinertness. However, the low biointerfacial affinity of pure PEEK implants and the decrease of mechanical strength after processing greatly limit their clinical applications. In this work, the influences on mechanical performance and biointerfacial affinity of the PEEK/nanohydroxyapatite (nHA) composites are systematically investigated. Results show that the mechanical performance of PEEK/nHA composites was improved by adjusting the nHA content. The maximum values of the tensile, compressive, bending, and impact strength of the composites were increased by approximately 16.2, 25, 54, and 21%, respectively, when compared with that of pure PEEK. Studies in vitro show that PEEK/nHA composites display good cytocompatibility and promote the biomimic formation of HA, adhesion, and proliferation of L929 cells on the surface. Studies in vivo demonstrate that, compared to the pure PEEK, PEEK/nHA composites exhibit higher biointerfacial affinity, including the adhesion and encapsulation of muscle tissues on the surface of the implants and the suppression of inflammatory reaction around the implants. Our findings could pave the way for extensive applications of PEEK/nHA composites in hard tissue repair, particularly orthopedic surgery.
Collapse
Affiliation(s)
- Xunzhi Yu
- State
Key Laboratory of Advanced Technology for Materials Synthesis and
Processing, Biomedical Material and Engineering Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Shun Yao
- Center
for Pituitary Tumor Surgery, Department of Neurosurgery, The First
Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, P. R. China
| | - Chang Chen
- State
Key Laboratory of Advanced Technology for Materials Synthesis and
Processing, Biomedical Material and Engineering Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Jin Wang
- State
Key Laboratory of Advanced Technology for Materials Synthesis and
Processing, Biomedical Material and Engineering Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Yaomin Li
- State
Key Laboratory of Advanced Technology for Materials Synthesis and
Processing, Biomedical Material and Engineering Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Youfa Wang
- State
Key Laboratory of Advanced Technology for Materials Synthesis and
Processing, Biomedical Material and Engineering Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Ali Khademhosseini
- Center
for Minimally Invasive Therapeutics (C-MIT), Department of Bioengineering, University of California-Los Angeles, Los Angeles, California 90095, United States
| | - Jiangling Wan
- National
Engineering Research Center for Nanomedicine, College of Life Science
and Technology, Huazhong University of Science
and Technology, Wuhan 430074, P. R. China
| | - Qingzhi Wu
- State
Key Laboratory of Advanced Technology for Materials Synthesis and
Processing, Biomedical Material and Engineering Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| |
Collapse
|
16
|
Williams DF. Specifications for Innovative, Enabling Biomaterials Based on the Principles of Biocompatibility Mechanisms. Front Bioeng Biotechnol 2019; 7:255. [PMID: 31649926 PMCID: PMC6794428 DOI: 10.3389/fbioe.2019.00255] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
In any engineering discipline, whenever products are designed, manufactured and ultimately utilized for the benefits of society, a series of specifications for the product are defined, and maybe refined, in order that they perform as effectively as possible, with due attention being paid to the safety, and economic aspects. These specifications are established with respect to all of the relevant properties, including those determined by mechanical, physical, chemical, manufacturing and environmental conditions, and the resulting design and materials selection reflects the optimal balance. In areas of medical technology, these specifications should be based on both functionality, which determines whether a device can actually perform as intended, and biocompatibility, which determines how the device interacts, both acutely and chronically, with the body. Unfortunately, whilst so much progress has been made with the development of superior functionality for the treatment and diagnosis of so many disease states, this is not the same for biocompatibility, where the single most-important currently adopted specification is that the device should do no harm, which falls far short of the ideal requirement. This paper addresses the profound need for biomaterials specifications to be based on the mechanisms of biocompatibility.
Collapse
Affiliation(s)
- David F. Williams
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States
- Strait Access Technologies, Cape Town, South Africa
| |
Collapse
|
17
|
Chen L, Wang D, Peng F, Qiu J, Ouyang L, Qiao Y, Liu X. Nanostructural Surfaces with Different Elastic Moduli Regulate the Immune Response by Stretching Macrophages. NANO LETTERS 2019; 19:3480-3489. [PMID: 31091110 DOI: 10.1021/acs.nanolett.9b00237] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A proper immune response is key for the successful implantation of biomaterials, and designing and fabricating biomaterials to regulate immune responses is the future trend. In this work, three different nanostructures were constructed on the surface of titanium using a hydrothermal method, and through a series of in vitro and in vivo experiments, we found that the aspect ratio of nanostructures can affect the elastic modulus of a material surface and further regulate immune cell behaviors. This work demonstrates that nanostructures with a higher aspect ratio can endow a material surface with a lower elastic modulus, which was confirmed by experiments and theoretical analyses. The deflection of nanostructures under the cell adsorption force is a substantial factor in stretching macrophages to enhance cell adhesion and spreading, further inducing macrophage polarization toward the M1 phenotype and leading to intense immune responses. In contrast, a nanostructure with a lower aspect ratio on a material surface leads to a higher surface elastic modulus, making deflection of the material difficult and creating a surface that is not conducive to macrophage adhesion and spreading, thus reducing the immune response. Moreover, molecular biology experiments indicated that regulation of the immune response by the elastic modulus is primarily related to the NF-κB signaling pathway. These findings suggest that the immune response can be regulated by constructing nanostructural surfaces with the proper elastic modulus through their influence on cell adhesion and spreading, which provides new insights into the surface design of biomaterials.
Collapse
Affiliation(s)
- Lan Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Donghui Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
| | - Feng Peng
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Jiajun Qiu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Liping Ouyang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yuqin Qiao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|