1
|
Pan K, Li Q, Guo Z, Li Z. Healing action of Interleukin-4 (IL-4) in acute and chronic inflammatory conditions: Mechanisms and therapeutic strategies. Pharmacol Ther 2025; 265:108760. [PMID: 39615600 DOI: 10.1016/j.pharmthera.2024.108760] [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: 07/14/2024] [Revised: 11/02/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Interleukin-4 (IL-4), which is traditionally associated with inflammation, has emerged as a key player in tissue regeneration. Produced primarily by T-helper 2 (Th2) and other immune cells, IL-4 activates endogenous lymphocytes and promotes M2 macrophage polarization, both of which are crucial for tissue repair. Moreover, IL-4 stimulates the proliferation and differentiation of various cell types, contributing to efficient tissue regeneration, and shows promise for promoting tissue regeneration after injury. This review explores the multifaceted roles of IL-4 in tissue repair, summarizing its mechanisms and potential for clinical application. This review delves into the multifaceted functions of IL-4, including its immunomodulatory effects, its involvement in tissue regeneration, and its potential therapeutic applications. We discuss the mechanisms underlying IL-4-induced M2 macrophage polarization, a crucial process for tissue repair. Additionally, we explore innovative strategies for delivering IL-4, including gene therapy, protein-based therapies, and cell-based therapies. By leveraging the regenerative properties of IL-4, we can potentially develop novel therapies for various diseases, including chronic inflammatory disorders, autoimmune diseases, and organ injuries. While early research has shown promise for the application of IL-4 in regenerative medicine, further studies are needed to fully elucidate its therapeutic potential and optimize its use.
Collapse
Affiliation(s)
- Kai Pan
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou, China; Nankai University School of Medicine, Tianjin, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Qiong Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China; Sanquan Medical College, Xinxiang Medical University, Xinxiang, China.
| | - Zhikun Guo
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China.
| | - Zongjin Li
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou, China; Nankai University School of Medicine, Tianjin, China; Sanquan Medical College, Xinxiang Medical University, Xinxiang, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
2
|
Dworan J, Aellos F, Grauer JA, Fabbri G, Harder KG, Boccardo S, Cuevas PL, Dawid I, Vicini M, Helms JA. Dynamics of Mucosal Integration of Machined versus Anodized Titanium Implants. J Dent Res 2024:220345241296506. [PMID: 39704472 DOI: 10.1177/00220345241296506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
The long-term success of dental implants depends on the ability of soft tissues to form a protective barrier, limiting pathogen infiltration into peri-implant tissues. Here, we investigated the impact of an anodized surface modification on mucosal integration. Scanning electron microscopy and surface chemistry characterization were carried out on miniaturized implants. Following placement in fresh extraction sockets of mice, peri-implant tissues were examined at 4 time points. Histology along with quantitative immunohistochemistry for Keratin14, Vimentin, Laminin5, and CD68 were carried out on postimplant day (PID) 3 to assess early events in soft-tissue repair; on PID7, when peri-implant epithelialization was complete; at PID14, when osseointegration was complete; and at PID28, when soft-tissue maturation was nearing completion. In all cases, an intact junctional epithelium served as a reference. These analyses supported 3 conclusions: first, maturation of the peri-implant epithelium (PIE) is a protracted process, consistent with clinical observations. Second, maturation of the soft tissue-implant interface is slower than maturation of the bone-implant interface. Third, there is a benefit, albeit transient, to soft-tissue maturation around an anodized implant surface. Given its prolonged time course, strategies to improve and/or accelerate PIE maturation are likely to have significant clinical benefit.
Collapse
Affiliation(s)
- J Dworan
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Medical University of Vienna, Department of Anatomy, Center for Anatomy and Cell Biology, Vienna, Austria
| | - F Aellos
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - J A Grauer
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
| | - G Fabbri
- Private practice, Ban Mancini Fabbri Dental Clinic, Cattolica, Italy
| | - K G Harder
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - S Boccardo
- Nobel Biocare Services AG, Kloten, Switzerland
| | - P L Cuevas
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - I Dawid
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - M Vicini
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - J A Helms
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
3
|
Butucescu M, Imre M, Rus-Hrincu F, Voicu-Balasea B, Popa A, Moisa M, Ripszky A, Neculau C, Pituru SM, Pârvu S. Cell-Type-Specific ROS-AKT/mTOR-Autophagy Interplay-Should It Be Addressed in Periimplantitis? Diagnostics (Basel) 2024; 14:2784. [PMID: 39767145 PMCID: PMC11727345 DOI: 10.3390/diagnostics14242784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/09/2024] [Indexed: 01/03/2025] Open
Abstract
Periimplantitis represents an inflammatory disease of the soft and hard tissues surrounding the osseointegrated dental implant, triggering progressive damage to the alveolar bone. Cumulative data have revealed that periimplantitis plays a crucial part in implant failure. Due to the strategic roles of autophagy and its upstream coordinator, the AKT/mTOR pathway, in inflammatory responses, the crosstalk between them in the context of periimplantitis should become a key research target, as it opens up an area of interesting data with clinical significance. Therefore, in this article, we aimed to briefly review the existing data concerning the complex roles played by ROS in the interplay between the AKT/mTOR signaling pathway and autophagy in periimplantitis, in each of the main cell types involved in periimplantitis pathogenesis and evolution. Knowing how to modulate specifically the autophagic machinery in each of the cellular types involved in the healing and osseointegration steps post implant surgery can help the clinician to make the most appropriate post-surgery decisions. These decisions might be crucial in order to prevent the occurrence of periimplantitis and ensure the proper conditions for effective osseointegration, depending on patients' clinical particularities.
Collapse
Affiliation(s)
- Mihai Butucescu
- Department of Organization, Professional Legislation and Management of the Dental Office, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Marina Imre
- Department of Prosthodontics, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Calea Plevnei, 010221 Bucharest, Romania;
| | - Florentina Rus-Hrincu
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Bianca Voicu-Balasea
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Alexandra Popa
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Mihai Moisa
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Alexandra Ripszky
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Cristina Neculau
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Silviu Mirel Pituru
- Department of Organization, Professional Legislation and Management of the Dental Office, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Simona Pârvu
- National Institute of Public Health, General Medicine Faculty, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
4
|
Le Z, Ramos MC, Shou Y, Li RR, Cheng HS, Jang CJ, Liu L, Xue C, Li X, Liu H, Lim CT, Tan NS, White AD, Charles CJ, Chen Y, Liu Z, Tay A. Bioactive sucralfate-based microneedles promote wound healing through reprogramming macrophages and protecting endogenous growth factors. Biomaterials 2024; 311:122700. [PMID: 38996671 DOI: 10.1016/j.biomaterials.2024.122700] [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: 04/03/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024]
Abstract
Impaired wound healing due to insufficient cell proliferation and angiogenesis is a significant physical and psychological burden to patients worldwide. Therapeutic delivery of exogenous growth factors (GFs) at high doses for wound repair is non-ideal as GFs have poor stability in proteolytic wound environments. Here, we present a two-stage strategy using bioactive sucralfate-based microneedle (SUC-MN) for delivering interleukin-4 (IL-4) to accelerate wound healing. In the first stage, SUC-MN synergistically enhanced the effect of IL-4 through more potent reprogramming of pro-regenerative M2-like macrophages via the JAK-STAT pathway to increase endogenous GF production. In the second stage, sucralfate binds to GFs and sterically disfavors protease degradation to increase bioavailability of GFs. The IL-4/SUC-MN technology accelerated wound healing by 56.6 % and 46.5 % in diabetic mice wounds and porcine wounds compared to their respective untreated controls. Overall, our findings highlight the innovative use of molecular simulations to identify bioactive ingredients and their incorporation into microneedles for promoting wound healing through multiple synergistic mechanisms.
Collapse
Affiliation(s)
- Zhicheng Le
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Mayk Caldas Ramos
- Department of Chemical Engineering, University of Rochester, 14627, USA
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Renee R Li
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore; Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, 119228, Singapore
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Clarisse Jm Jang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Ling Liu
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore
| | - Chencheng Xue
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Hong Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Andrew D White
- Department of Chemical Engineering, University of Rochester, 14627, USA
| | - Christopher John Charles
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore; Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, 119228, Singapore; Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore.
| |
Collapse
|
5
|
Wei X, Lei L, Luo L, Zhou Y, Zheng Z, Chen W. Advances in osteoimmunomodulation of biomaterials after intrabone implantation: focus on surface hydrophilicity. J Mater Chem B 2024; 12:11089-11104. [PMID: 39387541 DOI: 10.1039/d4tb01907e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Biomaterials intended for intrabone implantation are extensively utilized in orthopedic and dental applications. Their surface properties, particularly hydrophilicity, significantly influence the biological interactions surrounding the implant, ultimately determining the implant's in vivo fate. Recently, the role of osteoimmunomodulation in these implantable biomaterials has been recognized for its importance in regulating biomaterial-mediated osteogenesis. Consequently, it is imperative to elucidate the correlation between hydrophilicity and the immune response for the development of osteoimmunomodulatory implants. Herein, this review highlights recent advances in osteoimmunomodulation of biomaterials after intrabone implantation from a novel perspective-surface hydrophilicity, and summarizes the series of immune reactions and subsequent bone remodeling that occur in response to hydrophilic implants, focusing on protein adsorption, the behaviors of major immune cells, and osteoimmunomodulation-enhanced angiogenesis and osteogenesis. Hydrophilic biomaterials have the capacity to alter the surrounding immune microenvironment and accelerate the process of material-tissue bonding, thereby facilitating the successful integration of biomaterials with tissue. Collectively, the authors hope that this article provides strategies for modulating hydrophilicity to achieve osteoimmunomodulatory performance and further promotes the development of novel implantable biomaterials for orthopedic and dental applications.
Collapse
Affiliation(s)
- Xinpeng Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Linshan Lei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ling Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Ying Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zheng Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Wenchuan Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
6
|
Li S, Lin Y, Mo C, Bi J, Liu C, Lu Y, Jia B, Xu S, Liu Z. Application of metal-organic framework materials in regenerative medicine. J Mater Chem B 2024; 12:8543-8576. [PMID: 39136436 DOI: 10.1039/d4tb00226a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
In the past few decades, scaffolds manufactured from composite or hybrid biomaterials of natural or synthetic origin have made great strides in enhancing wound healing and repairing fractures and pathological bone loss. However, the prevailing use of such scaffolds in tissue engineering is accompanied by numerous constraints, including low mechanical stability, poor biological activity, and impaired cell proliferation and differentiation. The performance of scaffolds in wound and bone tissue engineering may be enhanced by some modifications in the synthesis of nanoscale metal-organic framework (nano-MOF) scaffolds. Nano-MOFs have attracted researchers' attention in recent years due to their distinctive features, which include tenability, biocompatibility, good mechanical stability, and ultrahigh surface area. The biological properties of scaffolds are enhanced and tissue regeneration is facilitated by the introduction of nano-MOFs. Moreover, the physicochemical characteristics, drug loading, and ion release capacities of the scaffolds are improved by the nanoscale structure and topological features of nano-MOFs, which also control stem cell differentiation, proliferation, and attachment. This review provides further comprehensive detail about the most recent uses of nano-MOFs in tissue engineering. The distinct characteristics of nano-MOFs are explored in enhancing tissue repair, wound healing, osteoinduction, and bone conductivity. Significant attributes include high antibacterial activity, substantial drug-loading capacity, and the ability to regulate drug release. Finally, this discussion addresses the obstacles, clinical impediments, and considerations encountered in the application of these nanomaterials to diverse scaffolds, tissue-mimicking structures, dressings, fillers, and implants for bone tissue repair and wound healing.
Collapse
Affiliation(s)
- Siwei Li
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Yunhe Lin
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Jiaming Bi
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Chengxia Liu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Yu Lu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Li J, Liu X, Jiang H, Yang M. Interleukin-4-Loaded Gelatin Methacryloyl Hydrogel Promotes Subcutaneous Chondrogenesis of Engineered Auricular Cartilage in a Rabbit Model. J Biomed Mater Res B Appl Biomater 2024; 112:e35473. [PMID: 39198004 DOI: 10.1002/jbm.b.35473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/13/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024]
Abstract
Tissue engineering technology offers a promising solution for ear reconstruction; however, it faces the challenge of foreign body reaction and neocartilage malformation. This study investigates the impact of interleukin-4 (IL-4), an anti-inflammatory factor, on cartilage regeneration of hydrogel encapsulating autologous auricular chondrocytes in a rabbit subcutaneous environment. Initially, we assessed the influence of IL-4 on chondrocyte proliferation and determined the appropriate concentration using the CCK-8 test in vitro. Subsequently, we loaded IL-4 into gelatin methacryloyl (GelMA) hydrogel containing chondrocytes and measured its release profile through ELISA. The constructs were then implanted autologously into rabbits' subcutis, and after 3, 7, 14, and 28 days, cartilage matrix formation was evaluated by histological examinations, and gene expression levels were detected by qRT-PCR. Results demonstrated that IL-4 promotes chondrocyte proliferation in vitro, and maximum release from constructs occurred during the first week. In the rabbit subcutaneous implantation model, IL-4-loaded constructs (20 ng/mL) maintained a superior chondrocytic phenotype compared to controls with increased expression of anti-inflammatory factors. These findings highlight IL-4 as a potential strategy for promoting chondrogenesis in a subcutaneous environment and improving ear reconstruction.
Collapse
Affiliation(s)
- Jinqiao Li
- Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xia Liu
- Research Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Haiyue Jiang
- Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Mingyong Yang
- Department of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
8
|
Abdalla MM, Sayed O, Lung CYK, Rajasekar V, Yiu CKY. Applications of Bioactive Strontium Compounds in Dentistry. J Funct Biomater 2024; 15:216. [PMID: 39194654 DOI: 10.3390/jfb15080216] [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: 07/08/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Divalent cations have captured the interest of researchers in biomedical and dental fields due to their beneficial effects on bone formation. These metallic elements are similar to trace elements found in human bone. Strontium is a divalent cation commonly found in various biomaterials. Since strontium has a radius similar to calcium, it has been used to replace calcium in many calcium-containing biomaterials. Strontium has the ability to inhibit bone resorption and increase bone deposition, making it useful in the treatment of osteoporosis. Strontium has also been used as a radiopacifier in dentistry and has been incorporated into a variety of dental materials to improve their radiopacity. Furthermore, strontium has been shown to improve the antimicrobial and mechanical properties of dental materials, promote enamel remineralization, alleviate dentin hypersensitivity, and enhance dentin regeneration. The objective of this review is to provide a comprehensive review of the applications of strontium in dentistry.
Collapse
Affiliation(s)
- Mohamed Mahmoud Abdalla
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
- Dental Biomaterials, Faculty of Dental Medicine, Al-Azhar University, Cairo 11651, Egypt
| | - Osama Sayed
- Faculty of Dentistry, Fayoum University, Faiyum 63514, Egypt
| | - Christie Ying Kei Lung
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Vidhyashree Rajasekar
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
9
|
Wu Y, Chen X, Song P, Li R, Zhou Y, Wang Q, Shi J, Qiao W, Dong N. Functional Oxidized Hyaluronic Acid Cross-Linked Decellularized Heart Valves for Improved Immunomodulation, Anti-Calcification, and Recellularization. Adv Healthc Mater 2024; 13:e2303737. [PMID: 38560921 DOI: 10.1002/adhm.202303737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/09/2024] [Indexed: 04/04/2024]
Abstract
Tissue engineering heart valves (TEHVs) are expected to address the limitations of mechanical and bioprosthetic valves used in clinical practice. Decellularized heart valve (DHV) is an important scaffold of TEHVs due to its natural three-dimensional structure and bioactive extracellular matrix, but its mechanical properties and hemocompatibility are impaired. In this study, DHV is cross-linked with three different molecular weights of oxidized hyaluronic acid (OHA) by a Schiff base reaction and presented enhanced stability and hemocompatibility, which could be mediated by the molecular weight of OHA. Notably, DHV cross-linked with middle- and high-molecular-weight OHA could drive the macrophage polarization toward the M2 phenotype in vitro. Moreover, DHV cross-linked with middle-molecular-weight OHA scaffolds are further modified with RGD-PHSRN peptide (RPF-OHA/DHV) to block the residual aldehyde groups of the unreacted OHA. The results show that RPF-OHA/DHV not only exhibits anti-calcification properties, but also facilitates endothelial cell adhesion and proliferation in vitro. Furthermore, RPF-OHA/DHV shows excellent performance under an in vivo hemodynamic environment with favorable recellularization and immune regulation without calcification. The optimistic results demonstrate that OHA with different molecular weights has different cross-linking effects on DHV and that RPF-OHA/DHV scaffold with enhanced immune regulation, anti-calcification, and recellularization properties for clinical transformation.
Collapse
Affiliation(s)
- Yunlong Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, China
| | - Peng Song
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Rui Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Ying Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Qin Wang
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| |
Collapse
|
10
|
Zhu S, Chen Y, Lu Z, Kong S, Zhang Y, Jia X, Xin H, Zhang X, Zhang W, Liu F, Kong L. Bacteroid cerium oxide particles promote macrophage polarization to achieve early vascularization and subsequent osseointegration around implants. Biochem Biophys Res Commun 2024; 703:149647. [PMID: 38350211 DOI: 10.1016/j.bbrc.2024.149647] [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: 12/15/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024]
Abstract
The establishment of an osseointegration is crucial for the long-term stability and functionality of implant materials, and early angiogenesis is the key to successful osseointegration. However, the bioinertness of titanium implants affects osseointegration, limiting their clinical application. In this study, inspired by the rapid polarization of macrophages following the phagocytosis of bacteria, we developed bacteroid cerium oxide particles; these particles were composed of CeO2 and had a size similar to that of Bacillus (0.5 μ m). These particles were constructed on the implant surfaces using a hydrothermal method. In vitro experiments demonstrated that the particles effectively decreased the reactive oxygen species (ROS) levels in macrophages (RAW264.7). Furthermore, these particles exerted effects on M1 macrophage polarization, enhanced nitric oxide (NO) secretion to promote vascular regeneration, and facilitated rapid macrophage transition to the M2 phenotype. Subsequently, the particles facilitated human umbilical vein endothelial cell (HUVEC) migration. In vivo studies showed that these particles rapidly stimulated innate immune responses in animal models, leading to enhanced angiogenesis around the implant and improved osseointegration. In summary, the presence of bacteroid cerium oxide particles on the implant surface regulated and accelerated macrophage polarization, thereby enhancing angiogenesis during the immune response and improving peri-implant osseointegration.
Collapse
Affiliation(s)
- Simin Zhu
- College of Life Sciences, Northwest University, Xi'an, 710069, People's Republic of China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yicheng Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Zihan Lu
- College of Life Sciences, Northwest University, Xi'an, 710069, People's Republic of China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Shaolingzhuo Kong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yufan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Xuelian Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - He Xin
- The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, Fujian, 350001, People's Republic of China
| | - Xiao Zhang
- College of Life Sciences, Northwest University, Xi'an, 710069, People's Republic of China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Wenhui Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Fuwei Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| | - Liang Kong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| |
Collapse
|
11
|
Zhou L, Chen D, Wu R, Li L, Shi T, Shangguang Z, Lin H, Chen G, Wang Z, Liu W. An injectable and photocurable methacrylate-silk fibroin/nano-hydroxyapatite hydrogel for bone regeneration through osteoimmunomodulation. Int J Biol Macromol 2024; 263:129925. [PMID: 38311129 DOI: 10.1016/j.ijbiomac.2024.129925] [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: 12/06/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024]
Abstract
Tissue engineering has emerged as a promising approach for addressing bone defects. Most of the traditional 3D printing materials predominantly relying on polymers and ceramics. Although these materials exhibit superior osteogenic effects, their gradual degradation poses a limitation. Digital light processing (DLP) 3D bioprinting that uses natural biomaterials as bioinks has become one of the promising strategies for bone regeneration. In this study, we introduce a hydrogel biomaterial derived from silk fibroin (SF). Notably, we present the novel integration of nano-hydroxyapatite (nHA) into the hydrogel, forming a composite hydrogel that rapidly cross-links upon initiation. Moreover, we demonstrate the loading of nHA through non-covalent bonds in SilMA. In vitro experiments reveal that composite hydrogel scaffolds with 10 % nHA exhibit enhanced osteogenic effects. Transcriptomic analysis indicates that the composite hydrogel promotes bone regeneration by inducing M2 macrophage polarization. Furthermore, rat femoral defect experiments validate the efficacy of SilMA/nHA10 in bone regeneration. This study synthesis of a simple and effective composite hydrogel bioink for bone regeneration, presenting a novel strategy for the future implementation of digital 3D printing technology in bone tissue engineering.
Collapse
Affiliation(s)
- Linquan Zhou
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Dehui Chen
- Fujian Medical University, Fuzhou 350000, China
| | - Rongcan Wu
- Fujian Medical University, Fuzhou 350000, China
| | - Lan Li
- Key Laboratory of Optoelectronic Materials Chemistry and Physics, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Tengbin Shi
- Fujian Medical University, Fuzhou 350000, China
| | - Zhitao Shangguang
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Hailin Lin
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Gang Chen
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhenyu Wang
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | - Wenge Liu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
12
|
Zhong C, Zhu H, Sheng Y, Wo J, You D, Sun G, Yu Z, Li W, Wang X. Biocompatibility and osteogenic potential of choline phosphate chitosan-coated biodegradable Zn1Mg. Acta Biomater 2024; 175:395-410. [PMID: 38096961 DOI: 10.1016/j.actbio.2023.12.014] [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: 09/12/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/25/2023]
Abstract
Zinc alloys have demonstrated considerable potentials as implant materials for biodegradable vascular and orthopedic applications. However, the high initial release of Zn2+ can trigger intense immune responses that impede tissue healing. To address this challenge and enhance the osteogenic capacity of zinc alloys, the surface of Zn1Mg was subjected to CO2 plasma modification (Zn1Mg-PP) followed by grafting with choline phosphate chitosan (Zn1Mg-PP-PCCs). This study aims to investigate the in vitro and in vivo biocompatibility of the surface-modified Zn1Mg. The effect of the surface modification on the inflammatory response and osteogenic repair process was investigated. Compared with unmodified Zn1Mg, the degradation rate of Zn1Mg-PP-PCCs was significantly decreased, avoiding the cytotoxicity triggered by the release of large amounts of Zn2+. Moreover, PCCs significantly enhanced the cell-material adhesion, promoted the proliferation of osteoblasts (MC3T3-E1) and upregulated the expression of key osteogenic factors in vitro. Notably, the in vivo experiments revealed that the surface modification of Zn1Mg suppressed inhibited the expression of inflammatory cytokines, promoting the secretion of anti-inflammatory factors, thereby reducing inflammation and promoting bone tissue repair. Furthermore, histological analysis of tissue sections exhibited strong integration between the material and the bone, along with well-defined new bone formation and reduced osteoclast aggregation on the surface. This was attributed to the improved immune microenvironment by PCCs, which promoted osteogenic differentiation of osteoblasts. These findings highlight that the preparation of PCCs coatings on zinc alloy surfaces effectively inhibited ion release and modulated the immune environment to promote bone tissue repair. STATEMENT OF SIGNIFICANCE: Surface modification of biodegradable Zn alloys facilitates the suppression of intense immune responses caused by excessive ion release concentrations from implants. We modified the surface of Zn1Mg with choline phosphate chitosan (PCCs) and investigated the effects of surface modification on the inflammatory response and osteogenic repair process. In vitro results showed that the PCCs coating effectively reduced the degradation rate of Zn1Mg to avoid cytotoxicity caused by high Zn2+ concentration, favoring the proliferation of osteoblasts. In addition, in vivo results indicated that Zn1Mg-PP-PCCs attenuated inflammation to promote bone repair by modulating the release of inflammation-related factors. The surface-modified Zn1Mg implants demonstrated strong osseointegration, indicating that the PCCs coating effectively modulated the immune microenvironment and promoted bone healing.
Collapse
Affiliation(s)
- Chen Zhong
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou 510632, China
| | - Haoran Zhu
- Guandgong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Heyuan 517000, China
| | - Yinying Sheng
- Institute of Corrosion Science and Technology, Guangzhou 510530, China.
| | - Jin Wo
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Deqiang You
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou 510632, China
| | - Guodong Sun
- Guandgong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Heyuan 517000, China; Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China.
| | - Zhentao Yu
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou 510632, China
| | - Wei Li
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou 510632, China
| | - Xiaojian Wang
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou 510632, China; Shaoguan Research Institute of Jinan University, 168 Muxi Avenue, Shaoguan 512029, China.
| |
Collapse
|
13
|
Tao Z, Tao M, Zhou M, Wu XJ. Niacin treatment prevents bone loss in iron overload osteoporotic rats via activation of SIRT1 signaling pathway. Chem Biol Interact 2024; 388:110827. [PMID: 38081572 DOI: 10.1016/j.cbi.2023.110827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 10/31/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Recently, more and more studies have revealed that iron overload can lead to osteoporosis by inducing oxidative stress. Niacin (NAN), also known as nicotinate or vitamin B3, has been confirmed to possess potent antioxidative effects. In addition, very little is currently known about the protective effects of NAN on iron overload in osteoporotic bone tissue. Therefore, we aimed to evaluate the protective effect of niacin on iron overload-induced bone injury and to investigate the effect and underlying mechanisms of the niacin and iron overload on intracellular antioxidant properties. When MC3T3-E1 and RAW264.7 cells were cultured in the presence of ammonium ferric citrate(FAC), NAN therapy could increase the matrix mineralization and promote expression of osteogenic markers in MC3T3-E1, inhibit osteoclastic differentiation of RAW264.7 cells, while increasing intracellular reactive oxygen species (ROS) levels and strengthening mitochondrial membrane potential (MMP). In the ovariectomized (OVX) rat model, NAN had an obvious protective effect against iron-overloaded injury. Meanwhile, superoxide dismutase 2 (SOD2), intracellular antioxidant enzymes and silent information regulator type 1 (SIRT1), were up-regulated in response to NAN exposures in MC3T3-E1. Furthermore, SIRT1 inhibitor EX527 attenuated the protective effects of NAN. Results revealed that NAN could stimulate osteogenic differentiation, inhibit osteoclastic differentiation and markedly increased antioxidant properties in cells through the induction of SIRT1. Studies suggest that niacin is a promising agent for preventing bone loss in iron overload conditions.
Collapse
Affiliation(s)
- Zhoushan Tao
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China; Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China.
| | - Ma Tao
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China
| | - Maosheng Zhou
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China
| | - Xing-Jing Wu
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe Shan Xi Road, Wuhu, 241001, Anhui, PR China
| |
Collapse
|
14
|
Bai L, Song P, Su J. Bioactive elements manipulate bone regeneration. BIOMATERIALS TRANSLATIONAL 2023; 4:248-269. [PMID: 38282709 PMCID: PMC10817798 DOI: 10.12336/biomatertransl.2023.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 01/30/2024]
Abstract
While bone tissue is known for its inherent regenerative abilities, various pathological conditions and trauma can disrupt its meticulously regulated processes of bone formation and resorption. Bone tissue engineering aims to replicate the extracellular matrix of bone tissue as well as the sophisticated biochemical mechanisms crucial for effective regeneration. Traditionally, the field has relied on external agents like growth factors and pharmaceuticals to modulate these processes. Although efficacious in certain scenarios, this strategy is compromised by limitations such as safety issues and the transient nature of the compound release and half-life. Conversely, bioactive elements such as zinc (Zn), magnesium (Mg) and silicon (Si), have garnered increasing interest for their therapeutic benefits, superior stability, and reduced biotic risks. Moreover, these elements are often incorporated into biomaterials that function as multifaceted bioactive components, facilitating bone regeneration via release on-demand. By elucidating the mechanistic roles and therapeutic efficacy of the bioactive elements, this review aims to establish bioactive elements as a robust and clinically viable strategy for advanced bone regeneration.
Collapse
Affiliation(s)
- Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
15
|
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: 13] [Impact Index Per Article: 6.5] [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
|
16
|
Lukin I, Erezuma I, Desimone MF, Zhang YS, Dolatshahi-Pirouz A, Orive G. Nanomaterial-based drug delivery of immunomodulatory factors for bone and cartilage tissue engineering. BIOMATERIALS ADVANCES 2023; 154:213637. [PMID: 37778293 DOI: 10.1016/j.bioadv.2023.213637] [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: 07/06/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
As life expectancy continues to increase, so do disorders related to the musculoskeletal system. Orthopedics-related impairments remain a challenge, with nearly 325 thousand and 120 thousand deaths recorded in 2019. Musculoskeletal system, including bone and cartilage tissue, is a living system in which cells constantly interact with the immune system, which plays a key role in the tissue repair process. An alternative to bridge the gap between these two systems is exploiting nanomaterials, as they have proven to serve as delivery agents of an array of molecules, including immunomodulatory agents (anti-inflammatory drugs, cytokines), as well as having the ability to mimic tissue by their nanoscopic structure and promote tissue repair per se. Therefore, this review outlooks nanomaterials and immunomodulatory factors widely employed in the area of bone and cartilage tissue engineering. Emerging developments in nanomaterials for delivery of immunomodulatory agents for bone and cartilage tissue engineering applications have also been discussed. It can be concluded that latest progress in nanotechnology have enabled to design intricate systems with the ability to deliver biologically active agents, promoting tissue repair and regeneration; thus, nanomaterials studied herein have shown great potential to serve as immunomodulatory agents in the area of tissue engineering.
Collapse
Affiliation(s)
- Izeia Lukin
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itsasne Erezuma
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Martin F Desimone
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | | | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria 01007, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore.
| |
Collapse
|
17
|
Zhang B, Yin X, Zhang F, Hong Y, Qiu Y, Yang X, Li Y, Zhong C, Yang H, Gou Z. Customized bioceramic scaffolds and metal meshes for challenging large-size mandibular bone defect regeneration and repair. Regen Biomater 2023; 10:rbad057. [PMID: 37359729 PMCID: PMC10287912 DOI: 10.1093/rb/rbad057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/20/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Large-size mandible graft has huge needs in clinic caused by infection, tumor, congenital deformity, bone trauma and so on. However, the reconstruction of large-size mandible defect is challenged due to its complex anatomical structure and large-range bone injury. The design and fabrication of porous implants with large segments and specific shapes matching the native mandible remain a considerable challenge. Herein, the 6% Mg-doped calcium silicate (CSi-Mg6) and β- and α-tricalcium phosphate (β-TCP, α-TCP) bioceramics were fabricated by digital light processing as the porous scaffolds of over 50% in porosity, while the titanium mesh was fabricated by selective laser melting. The mechanical tests showed that the initial flexible/compressive resistance of CSi-Mg6 scaffolds was markedly higher than that of β-TCP and α-TCP scaffolds. Cell experiments showed that these materials all had good biocompatibility, while CSi-Mg6 significantly promoted cell proliferation. In the rabbit critically sized mandible bone defects (∼13 mm in length) filled with porous bioceramic scaffolds, the titanium meshes and titanium nails were acted as fixation and load bearing. The results showed that the defects were kept during the observation period in the blank (control) group; in contrast, the osteogenic capability was significantly enhanced in the CSi-Mg6 and α-TCP groups in comparison with the β-TCP group, and these two groups not only had significantly increased new bone formation but also had thicker trabecular and smaller trabecular spacing. Besides, the CSi-Mg6 and α-TCP groups showed appreciable material biodegradation in the later stage (from 8 to 12 weeks) in comparison with the β-TCP scaffolds while the CSi-Mg6 group showed much outstanding mechanical capacity in vivo in the early stage compared to the β-TCP and α-TCP groups. Totally, these findings suggest that the combination of customized strength-strong bioactive CSi-Mg6 scaffolds together with titanium meshes is a promising way for repairing the large-size load-bearing mandible defects.
Collapse
Affiliation(s)
- Bin Zhang
- Correspondence address. E-mail: (B.Z.); (Z.G.)
| | - Xiaohong Yin
- State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhang
- Department of Stomatology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yirong Hong
- State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yuesheng Qiu
- Department of Stomatology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xianyan Yang
- Bio-Nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou 310058, China
| | - Yifan Li
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Cheng Zhong
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhongru Gou
- Correspondence address. E-mail: (B.Z.); (Z.G.)
| |
Collapse
|
18
|
Qiang H, Hou C, Zhang Y, Luo X, Li J, Meng C, Liu K, Lv Z, Chen X, Liu F. CaP-coated Zn-Mn-Li alloys regulate osseointegration via influencing macrophage polarization in the osteogenic environment. Regen Biomater 2023; 10:rbad051. [PMID: 37324238 PMCID: PMC10267298 DOI: 10.1093/rb/rbad051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 06/17/2023] Open
Abstract
Immune response is an important factor in determining the fate of bone replacement materials, in which macrophages play an important role. It is a new idea to design biomaterials with immunomodulatory function to reduce inflammation and promote bone integration by regulating macrophages polarization. In this work, the immunomodulatory properties of CaP Zn-Mn-Li alloys and the specific mechanism of action were investigated. We found that the CaP Zn0.8Mn0.1Li alloy promoted the polarization of macrophages toward M2 and reduced inflammation, which could effectively upregulate osteogenesis-related factors and promote new bone formation, indicating the important role of macrophages polarization in biomaterial induction of osteogenesis. In vivo studies further demonstrated that CaP Zn0.8Mn0.1Li alloy could stimulate osteogenesis better than other Zn-Mn-Li alloys implantations by regulating macrophages polarization and reducing inflammation. In addition, transcriptome results showed that CaP Zn0.8Mn0.1Li played an important regulatory role in the life process of macrophages, activating Toll-like receptor signaling pathway, which participated in the activation and attenuation of inflammation, and accelerated bone integration. Thus, by preparing CaP coatings on the surface of Zn-Mn-Li alloys and combining the bioactive ingredient with controlled release, the biomaterial will be imbibed with beneficial immunomodulatory properties that promote bone integration.
Collapse
Affiliation(s)
| | | | - Yujue Zhang
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
- School of Materials Science and Engineering, University of Science and Technology, Beijing 100083, China
| | - Xin Luo
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Jun Li
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Chunxiu Meng
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Kun Liu
- Liaocheng People’s Hospital, Liaocheng Dongchangfu People’s Hospital, Liaocheng 252000, China
| | - Zhaoyong Lv
- Correspondence address. E-mail: (Z.L.); (X.C.); (F.L.)
| | - Ximeng Chen
- Correspondence address. E-mail: (Z.L.); (X.C.); (F.L.)
| | - Fengzhen Liu
- Correspondence address. E-mail: (Z.L.); (X.C.); (F.L.)
| |
Collapse
|
19
|
Zhang Z, Wang F, Huang X, Sun H, Xu J, Qu H, Yan X, Shi W, Teng W, Jin X, Shao Z, Zhang Y, Zhao S, Wu Y, Ye Z, Yu X. Engineered Sensory Nerve Guides Self-Adaptive Bone Healing via NGF-TrkA Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206155. [PMID: 36725311 PMCID: PMC10074090 DOI: 10.1002/advs.202206155] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/10/2023] [Indexed: 06/17/2023]
Abstract
The upstream role of sensory innervation during bone homeostasis is widely underestimated in bone repairing strategies. Herein, a neuromodulation approach is proposed to orchestrate bone defect healing by constructing engineered sensory nerves (eSN) in situ to leverage the adaptation feature of SN during tissue formation. NGF liberated from ECM-constructed eSN effectively promotes sensory neuron differentiation and enhances CGRP secretion, which lead to improved RAOECs mobility and osteogenic differentiation of BMSC. In turn, such eSN effectively drives ossification in vivo via NGF-TrkA signaling pathway, which substantially accelerates critical size bone defect healing. More importantly, eSN also adaptively suppresses excessive bone formation and promotes bone remodeling by activating osteoclasts via CGRP-dependent mechanism when combined with BMP-2 delivery, which ingeniously alleviates side effects of BMP-2. In sum, this eSN approach offers a valuable avenue to harness the adaptive role of neural system to optimize bone homeostasis under various clinical scenario.
Collapse
Affiliation(s)
- Zengjie Zhang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Fangqian Wang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xin Huang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Hangxiang Sun
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Jianxiang Xu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Hao Qu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xiaobo Yan
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Wei Shi
- Department of OrthopedicTaizhou First People's HospitalWenzhou Medical University218 Hengjie Road, Huangyan DistrictTaizhou CityZhejiang Province318020P. R. China
| | - Wangsiyuan Teng
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xiaoqiang Jin
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Zhenxuan Shao
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Yongxing Zhang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Shenzhi Zhao
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Yan Wu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Zhaoming Ye
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xiaohua Yu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| |
Collapse
|
20
|
Peng H, Ling T, Zhang Y, Xie T, Pei X, Zhou K, Chen A, Chen J, Zhu X, Zhang X, Zhou Z. Nanowhiskers Orchestrate Bone Formation and Bone Defect Repair by Modulating Immune Cell Behavior. ACS APPLIED MATERIALS & INTERFACES 2023; 15:9120-9134. [PMID: 36755394 DOI: 10.1021/acsami.2c21865] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Immunomodulatory biomaterials have emerged as promising treatment agents for bone defects. However, it is unclear how such biomaterials control immune cell behaviors to facilitate large-segment bone defect repair. Herein, we fabricated biphasic calcium phosphate ceramics with nanowhisker structures to explore the immunoregulation features and influence on large-segment bone defect repair. We found that the nanowhisker structures markedly facilitated large-segment bone defect repair by promoting bone regeneration and scaffold resorption. Our in vitro experiment and transcriptomic analysis showed that mechanical stress derived from nanowhisker structures may activate the transcription of Egr-1 to induce early switch of macrophage phenotype to M2, which could not only facilitate osteogenic differentiation of BMSCs but also enhance the expression of osteoclast differentiation-regulating genes of M2 macrophage. In vivo study showed that the nanowhisker structures relieved local inflammatory responses by inducing early switch of macrophage phenotype from M1 to M2, which resulted in accelerated osteoclastogenesis for biomaterial resorption and osteogenesis for ectopic bone formation. Hence, we presume that nanowhisker structures may orchestrate bone formation and material resorption coupling to facilitate large-segment bone defect repair by controlling the switch of macrophage phenotype. This study provides new insight into the designing of immunomodulatory tissue engineering biomaterials for treating large-segment bone defects.
Collapse
Affiliation(s)
- Haitao Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Tingxian Ling
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Zhang
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Tianhang Xie
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xuan Pei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Kai Zhou
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Anjing Chen
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiali Chen
- West China School of Nursing, Sichuan University/Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Zongke Zhou
- Orthopedic Research Institute & Department of Orthopedics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
21
|
Liu Z, Zhu J, Li Z, Liu H, Fu C. Biomaterial scaffolds regulate macrophage activity to accelerate bone regeneration. Front Bioeng Biotechnol 2023; 11:1140393. [PMID: 36815893 PMCID: PMC9932600 DOI: 10.3389/fbioe.2023.1140393] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Bones are important for maintaining motor function and providing support for internal organs. Bone diseases can impose a heavy burden on individuals and society. Although bone has a certain ability to repair itself, it is often difficult to repair itself alone when faced with critical-sized defects, such as severe trauma, surgery, or tumors. There is still a heavy reliance on metal implants and autologous or allogeneic bone grafts for bone defects that are difficult to self-heal. However, these grafts still have problems that are difficult to circumvent, such as metal implants that may require secondary surgical removal, lack of bone graft donors, and immune rejection. The rapid advance in tissue engineering and a better comprehension of the physiological mechanisms of bone regeneration have led to a new focus on promoting endogenous bone self-regeneration through the use of biomaterials as the medium. Although bone regeneration involves a variety of cells and signaling factors, and these complex signaling pathways and mechanisms of interaction have not been fully understood, macrophages undoubtedly play an essential role in bone regeneration. This review summarizes the design strategies that need to be considered for biomaterials to regulate macrophage function in bone regeneration. Subsequently, this review provides an overview of therapeutic strategies for biomaterials to intervene in all stages of bone regeneration by regulating macrophages.
Collapse
Affiliation(s)
- Zongtai Liu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, China
- Department of Orthopedics, Affiliated Hospital of Beihua University, Jilin, China
| | - Jiabo Zhu
- Department of Orthopedics, Affiliated Hospital of Beihua University, Jilin, China
| | - Zhuohan Li
- Department of Gynecology, Affiliated Hospital of Beihua University, Jilin, China
| | - Hanyan Liu
- Department of Orthopedics, Baicheng Central Hospital, Baicheng, China
| | - Changfeng Fu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
22
|
Zhou G, Wang F, Lin G, Tang B, Li X, Ding X, Wang W, Zhang J, Shi Y. Novel coatings for the continuous repair of human bone defects. Colloids Surf B Biointerfaces 2023; 222:113127. [PMID: 36610365 DOI: 10.1016/j.colsurfb.2023.113127] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
Bone defects are the second most common tissue grafts after blood. However, bone grafts face several problems, such as bone scaffolds, which have low bioactivity and are prone to corrosion. Much of the current research on bone scaffolds is focused on the mechanical aspects such as structure and strength. Surface modification of the bone scaffold is carried out in terms of the mechanical structure or structural design of the bone scaffold with reference to a bionic structure. However, with the development of mechanical designs, materials science, and medicine, many studies have reported that promoting bone growth by modifying the structure of the scaffold or coating is not possible. Therefore, the application of a bioactive coating to the surface of the bone scaffold is particularly important to generate a synergistic effect between the structure and active coating. In this article, we present several perspectives to improve the bioactivity of bone scaffolds, including corrosion resistance, loading of bioactive coatings or drugs on bone scaffolds, improved adhesion to the surface of the bone scaffolds, immune response modulation, and drawing on bionic structures during manufacturing.
Collapse
Affiliation(s)
- Guangzhen Zhou
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; Shandong Institute of Mechanical Design and Research, Jinan 250031, China.
| | - Fei Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; Shandong Institute of Mechanical Design and Research, Jinan 250031, China.
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China.
| | - Bingtao Tang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; Shandong Institute of Mechanical Design and Research, Jinan 250031, China.
| | - Xuelin Li
- School of Arts and Design, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Xinbing Ding
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; Shandong Institute of Mechanical Design and Research, Jinan 250031, China.
| | - Wenguang Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; Shandong Institute of Mechanical Design and Research, Jinan 250031, China.
| | - Jing Zhang
- Key Laboratory of Modern Preparation of TCM, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Yanbin Shi
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; Shandong Institute of Mechanical Design and Research, Jinan 250031, China; School of Arts and Design, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| |
Collapse
|
23
|
Li L, Li Q, Gui L, Deng Y, Wang L, Jiao J, Hu Y, Lan X, Hou J, Li Y, Lu D. Sequential gastrodin release PU/n-HA composite scaffolds reprogram macrophages for improved osteogenesis and angiogenesis. Bioact Mater 2023; 19:24-37. [PMID: 35415312 PMCID: PMC8980440 DOI: 10.1016/j.bioactmat.2022.03.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
Wound healing is a highly orchestrated process involving a variety of cells, including immune cells. Developing immunomodulatory biomaterials for regenerative engineering applications, such as bone regeneration, is an appealing strategy. Herein, inspired by the immunomodulatory effects of gastrodin (a bioactive component in traditional Chinese herbal medicine), a series of new immunomodulatory gastrodin-comprising biodegradable polyurethane (gastrodin-PU) and nano-hydroxyapatite (n-HA) (gastrodin-PU/n-HA) composites were developed. RAW 264.7 macrophages, rat bone marrow mesenchymal stem cells (rBMSCs), and human umbilical vein endothelial cells (HUVECs) were cultured with gastrodin-PU/n-HA containing different concentrations of gastrodin (0.5%, 1%, and 2%) to decipher their immunomodulatory effects on osteogenesis and angiogenesis in vitro. Results demonstrated that, compared with PU/n-HA, gastrodin-PU/n-HA induced macrophage polarization toward the M2 phenotype, as evidenced by the higher expression level of pro-regenerative cytokines (CD206, Arg-1) and the lower expression of pro-inflammatory cytokines (iNOS). The expression levels of osteogenesis-related factors (BMP-2 and ALP) in the rBMSCs and angiogenesis-related factors (VEGF and BFGF) in the HUVECs were significantly up-regulated in gastrodin-PU/n-HA/macrophage-conditioned medium. The immunomodulatory effects of gastrodin-PU/n-HA to reprogram macrophages from a pro-inflammatory (M1) phenotype to an anti-inflammatory and pro-healing (M2) phenotype were validated in a rat subcutaneous implantation model. And the 2% gastrodin-PU/n-HA significantly decreased fibrous capsule formation and enhanced angiogenesis. Additionally, 2% gastrodin-PU/n-HA scaffolds implanted in the rat femoral condyle defect model showed accelerated osteogenesis and angiogenesis. Thus, the novel gastrodin-PU/n-HA scaffold may represent a new and promising immunomodulatory biomaterial for bone repair and regeneration. A new immunomodulatory gastrodin-PU/n-HA biomaterial has been developed. The gastrodin-PU/n-HA triggered M2 macrophage polarization. The osteogenesis and angiogenesis were enhanced in response to the local immune microenvironment. The findings prove a therapeutic strategy in bone defect and other inflammatory osteoimmune disorders.
Collapse
Affiliation(s)
- Limei Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Li Gui
- Department of Endocrinology, The Third People's Hospital of Yunnan Province, Kunming, 650011, China
| | - Yi Deng
- School of Chemical Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Lu Wang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jianlin Jiao
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Yingrui Hu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Xiaoqian Lan
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650000, China
| | - Jianhong Hou
- Department of Orthopaedics, The Third People's Hospital of Yunnan Province, Kunming, 650011, China
- Corresponding author.
| | - Yao Li
- Department of Stomatology, The First People's Hospital of Yunnan Province, Kunming, 650032, China
- Corresponding author.
| | - Di Lu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
- Corresponding author.
| |
Collapse
|
24
|
Freitag L, Spinell T, Kröger A, Würfl G, Lauseker M, Hickel R, Kebschull M. Dental implant material related changes in molecular signatures in peri-implantitis - A systematic review and integrative analysis of omics in-vitro studies. Dent Mater 2023; 39:101-113. [PMID: 36526446 DOI: 10.1016/j.dental.2022.11.022] [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: 07/09/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Since peri-implantitis differs clinically and histopathologically from periodontitis, implant wear debris is considered to play a role in the destructive processes. This work aims to systematically review if titanium particles affect oral-related cells through changes in molecular signatures (e.g., transcriptome, proteome, epigenome), thereby promoting peri-implantitis. METHODS Leveraging three literature databases (Medline, Embase, Cochrane) a systematic search based on a priori defined PICOs was conducted: '-omics' studies examining titanium exposure in oral-related cells. After risk of bias assessments, lists of differentially expressed genes, proteins, and results of functional enrichment analyses were compiled. The significance of overlapping genes across multiple studies was assessed via Monte Carlo simulation and their ranking was verified using rank aggregation. RESULTS Out of 2104 screened articles we found 12 eligible publications. A significant overlap of gene expression in oral-related cells exposed to titanium particles was found in four studies. Furthermore, changes in biological processes like immune/inflammatory or stress response as well as toll-like receptor (TLR) and mitogen-activated protein kinase (MAPK) signaling pathways were linked to titanium in transcriptome and proteome studies. Epigenetic changes caused by titanium were detected but inconsistent. CONCLUSION An influence of titanium implant wear debris on the development and progression of peri-implantitis is plausible but needs to be proven in further studies. Limitations arise from small sample sizes of included studies and insufficient publication of re-analyzable data.
Collapse
Affiliation(s)
- Lena Freitag
- Department of Conservative Dentistry and Periodontology, University Hospital, Ludwig-Maximilians-University, Goethestr. 70, D-80336 Munich, Germany
| | - Thomas Spinell
- Department of Conservative Dentistry and Periodontology, University Hospital, Ludwig-Maximilians-University, Goethestr. 70, D-80336 Munich, Germany.
| | - Annika Kröger
- School of Dentistry, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK; Birmingham Community Healthcare NHS Foundation Trust, Birmingham, UK
| | | | - Michael Lauseker
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | - Reinhard Hickel
- Department of Conservative Dentistry and Periodontology, University Hospital, Ludwig-Maximilians-University, Goethestr. 70, D-80336 Munich, Germany
| | - Moritz Kebschull
- School of Dentistry, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK; Birmingham Community Healthcare NHS Foundation Trust, Birmingham, UK; Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, Columbia University College of Dental Medicine, New York, NY, USA
| |
Collapse
|
25
|
Wu Y, Zhang S, Sun L, Lu Y, Jiang Y, Xiao G. Strontium doping stimulates the phase composition and evolution of β-tricalcium phosphate prepared by wet chemical method. J SOLID STATE CHEM 2022. [DOI: 10.1016/j.jssc.2022.123732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Zhao DW, Fan XC, Zhao YX, Zhao W, Zhang YQ, Zhang RH, Cheng L. Biocompatible Nano-Hydroxyapatites Regulate Macrophage Polarization. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15196986. [PMID: 36234325 PMCID: PMC9573195 DOI: 10.3390/ma15196986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/06/2022] [Accepted: 10/05/2022] [Indexed: 05/27/2023]
Abstract
Research on regulation of the immune microenvironment based on bioactive materials is important to osteogenic regeneration. Hydroxyapatite (HAP) is believed to be a promising scaffold material for dental and orthopedic implantation due to its ideal biocompatibility and high osteoconductivity. However, any severe inflammation response can lead to loosening and fall of implantation, which cause implant failures in the clinic. Morphology modification has been widely studied to regulate the host immune environment and to further promote bone regeneration. Here, we report the preparation of nHAPs, which have uniform rod-like shape and different size (200 nm and 400 nm in length). The morphology, biocompatibility, and anti-inflammatory properties were evaluated. The results showed that the 400 nm nHAPs exhibited excellent biocompatibility and osteoimmunomodulation, which can not only induce M2-phenotype macrophages (M2) polarization to decrease the production of inflammatory cytokines, but also promote the production of osteogenic factor. The reported 400 nm nHAPs are promising for osteoimmunomodulation in bone regeneration, which is beneficial for clinical application of bone defects.
Collapse
Affiliation(s)
- Da-Wang Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, China
- Institute of Stomatology, Shandong University, Jinan 250012, China
| | - Xin-Cheng Fan
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, China
- Department of Orthopaedics, Taian City Central Hospital, Tai’an 271000, China
| | - Yi-Xiang Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, China
- Institute of Stomatology, Shandong University, Jinan 250012, China
| | - Wei Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, China
- Institute of Stomatology, Shandong University, Jinan 250012, China
| | - Yuan-Qiang Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Ren-Hua Zhang
- Outpatient Department, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Lei Cheng
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
27
|
Bai B, Hao J, Hou M, Wang T, Wu X, Liu Y, Wang Y, Dai C, Hua Y, Ji G, Zhou G. Repair of Large-Scale Rib Defects Based on Steel-Reinforced Concrete-Designed Biomimetic 3D-Printed Scaffolds with Bone-Mineralized Microenvironments. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42388-42401. [PMID: 36094886 DOI: 10.1021/acsami.2c08422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Tissue engineering technology provides a promising approach for large-scale bone reconstruction in cases of extensive chest wall defects. However, previous studies did not consider meticulous scaffold design specific to large-scale rib regeneration in terms of three-dimensional (3D) shape, proper porous structures, enough mechanical strength, and osteogenic microenvironments. Thus, there is an urgent need to develop an appropriate bone biomimetic scaffold (BBS) to address this problem. In this study, a BBS with controllable 3D morphology, appropriate mechanical properties, good biocompatibility and biodegradability, porous structure suitable for cell loading, and a biomimetic osteogenic inorganic salt (OIS) microenvironment was successfully prepared by integrating computer-aided design, 3D-printing, cast-molding, and freeze-drying technologies. The addition of the OIS in the scaffold substantially promoted ectopic bone regeneration in vivo, which might be attributed to the activation of osteogenic and angiogenic signaling pathways as well as upregulated expression of osteogenic genes. More importantly, dual long rib defects could be successfully repaired and medullary cavity recanalized by the rib-shaped mature cortical bone, which might be mediated by the activation of osteoclast signaling pathways. Thus, this paper presents a reliable BBS and proposes a new strategy for the repair of large-scale bone defects.
Collapse
Affiliation(s)
- Baoshuai Bai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Research Institute of Plastic Surgery, Weifang Medical University, Weifang, Shandong 261000, China
- National Tissue Engineering Center of China, Shanghai 200001, China
| | - Junxiang Hao
- Research Institute of Plastic Surgery, Weifang Medical University, Weifang, Shandong 261000, China
- National Tissue Engineering Center of China, Shanghai 200001, China
| | - Mengjie Hou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- National Tissue Engineering Center of China, Shanghai 200001, China
| | - Tao Wang
- Research Institute of Plastic Surgery, Weifang Medical University, Weifang, Shandong 261000, China
- National Tissue Engineering Center of China, Shanghai 200001, China
| | - Xiaodi Wu
- Research Institute of Plastic Surgery, Weifang Medical University, Weifang, Shandong 261000, China
- National Tissue Engineering Center of China, Shanghai 200001, China
| | - Yanhan Liu
- Shanghai JiaoTong University School of Medicine, Shanghai 200240, China
| | - Yiyang Wang
- National Tissue Engineering Center of China, Shanghai 200001, China
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Chengxiang Dai
- Cellular Biomedicine Group, Incorporated, No. 85 Faladi Road, Building 3, Pudong New Area, Shanghai 201210, China
| | - Yujie Hua
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- National Tissue Engineering Center of China, Shanghai 200001, China
- Shanghai JiaoTong University School of Medicine, Shanghai 200240, China
| | - Guangyu Ji
- National Tissue Engineering Center of China, Shanghai 200001, China
- Shanghai JiaoTong University School of Medicine, Shanghai 200240, China
- Department of Thoracic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
- Research Institute of Plastic Surgery, Weifang Medical University, Weifang, Shandong 261000, China
- National Tissue Engineering Center of China, Shanghai 200001, China
- Shanghai JiaoTong University School of Medicine, Shanghai 200240, China
| |
Collapse
|
28
|
Qiang HF, Lv ZY, Hou CY, Luo X, Li J, Liu K, Meng CX, Du WQ, Zhang YJ, Chen XM, Liu FZ. Development of biodegradable Zn-Mn-Li and CaP coatings on Zn-Mn-Li alloys and cytocompatibility evaluation for bone graft. Front Bioeng Biotechnol 2022; 10:1013097. [PMID: 36185442 PMCID: PMC9515419 DOI: 10.3389/fbioe.2022.1013097] [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: 08/06/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Zn-based alloys are considered as new kind of potential biodegradable implanted biomaterials recently. The difficulty of metal implanted biomaterials and bone tissue integration seriously affects the applications of metal implanted scaffolds in bone tissue-related fields. Herein, we self-designed Zn0.8Mn and Zn0.8Mn0.1Li alloys and CaP coated Zn0.8Mn and Zn0.8Mn0.1Li alloys, then evaluated the degradation property and cytocompatibility. The results demonstrated that the Zn0.8Mn0.1Li alloys had profoundly modified the degradation property and cytocompatibility, but Zn0.8Mn0.1Li alloys had particularly adverse effects on the surface morphology of osteoblasts. The results furtherly showed that the CaP-coated Zn0.8Mn and Zn0.8Mn0.1Li alloys scaffold had better biocompatibility, which would further guarantee the biosafety of this new kind of biodegradable Zn-based alloys implants for future clinical applications.
Collapse
Affiliation(s)
- Hui-Fen Qiang
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Zhao-Yong Lv
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
| | - Cai-Yao Hou
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
| | - Xin Luo
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
| | - Jun Li
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
| | - Kun Liu
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
| | - Chun-Xiu Meng
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
| | - Wan-Qing Du
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
| | - Yu-Jue Zhang
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
- *Correspondence: Feng-Zhen Liu, ; Xi-Meng Chen, ; Yu-Jue Zhang,
| | - Xi-Meng Chen
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
- *Correspondence: Feng-Zhen Liu, ; Xi-Meng Chen, ; Yu-Jue Zhang,
| | - Feng-Zhen Liu
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng, China
- Liaocheng People’s Hospital, Dongchangfu Hospital of Liaocheng Hospital, Liaocheng, China
- *Correspondence: Feng-Zhen Liu, ; Xi-Meng Chen, ; Yu-Jue Zhang,
| |
Collapse
|
29
|
Chen B, Liang Y, Song Y, Liang Y, Jiao J, Bai H, Li Y. Photothermal-Controlled Release of IL-4 in IL-4/PDA-Immobilized Black Titanium Dioxide (TiO 2) Nanotubes Surface to Enhance Osseointegration: An In Vivo Study. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5962. [PMID: 36079344 PMCID: PMC9457063 DOI: 10.3390/ma15175962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Host immune response has gradually been accepted as a critical factor in achieving successful implant osseointegration. The aim of this study is to create a favorable immune microenvironment by the dominant release of IL-4 during the initial few days after implant insertion to mitigate early inflammatory reactions and facilitate osseointegration. Herein, the B-TNT/PDA/IL-4 substrate was established by immobilizing an interleukin-4 (IL-4)/polydopamine (PDA) coating on a black TiO2 nanotube (B-TNT) surface, achieving on-demand IL-4 release under near infrared (NIR) irradiation. Gene Ontology (GO) enrichment analyses based on high-throughput DNA microarray data revealed that IL-4 addition inhibited osteoclast differentiation and function. Animal experiment results suggested that the B-TNT/PDA/IL-4+Laser substrate induced the least inflammatory, tartrate-resistant acid phosphatase, inducible nitric oxide synthase and the most CD163 positive cells, compared to the Ti group at 7 days post-implantation. In addition, 28 days post-implantation, micro-computed tomography results showed the highest bone volume/total volume, trabecular thickness, trabecular number and the lowest trabecular separation, while Hematoxylin-eosin and Masson-trichrome staining revealed the largest amount of new bone formation for the B-TNT/PDA/IL-4+Laser group. This study revealed the osteoimmunoregulatory function of the novel B-TNT/PDA/IL-4 surface by photothermal release of IL-4 at an early period post-implantation, thus paving a new way for dental implant surface modification.
Collapse
Affiliation(s)
- Bo Chen
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Yu Liang
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Yunjia Song
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Yunkai Liang
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Jian Jiao
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Hong Bai
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Ying Li
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
30
|
Kim YH, Oreffo ROC, Dawson JI. From hurdle to springboard: The macrophage as target in biomaterial-based bone regeneration strategies. Bone 2022; 159:116389. [PMID: 35301163 DOI: 10.1016/j.bone.2022.116389] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 12/16/2022]
Abstract
The past decade has seen a growing appreciation for the role of the innate immune response in mediating repair and biomaterial directed tissue regeneration. The long-held view of the host immune/inflammatory response as an obstacle limiting stem cell regenerative activity, has given way to a fresh appreciation of the pivotal role the macrophage plays in orchestrating the resolution of inflammation and launching the process of remodelling and repair. In the context of bone, work over the past decade has established an essential coordinating role for macrophages in supporting bone repair and sustaining biomaterial driven osteogenesis. In this review evidence for the role of the macrophage in bone regeneration and repair is surveyed before discussing recent biomaterial and drug-delivery based approaches that target macrophage modulation with the goal of accelerating and enhancing bone tissue regeneration.
Collapse
Affiliation(s)
- Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Jonathan I Dawson
- Bone and Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
31
|
Wu S, Ma J, Liu J, Liu C, Ni S, Dai T, Wang Y, Weng Y, Zhao H, Zhou D, Zhao X. Immunomodulation of Telmisartan-Loaded PCL/PVP Scaffolds on Macrophages Promotes Endogenous Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15942-15955. [PMID: 35353482 DOI: 10.1021/acsami.1c24748] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Biomaterial-immune system interactions play an important role in postimplantation osseointegration to retain the functionality of healthy and intact bones. Therefore, appropriate osteoimmunomodulation of implants has been considered and validated as an efficient strategy to alleviate inflammation and enhance new bone formation. Here, we fabricated a nanostructured PCL/PVP (polycaprolactone/polyvinylpyrrolidone) electrospinning scaffold for cell adhesion, tissue ingrowth, and bone defect padding. In addition, telmisartan, an angiotensin 2 receptor blocker with distinct immune bioactivity, was doped into PCL-/PVP-electrospun scaffolds at different proportions [1% (TPP-1), 5% (TPP-5), and 10% (TPP-10)] to investigate its immunomodulatory effects and osteoinductivity/conductivity. Telmisartan-loaded scaffolds displayed in vitro anti-inflammatory bioactivity on lipopolysaccharide-induced M1 macrophages by polarizing them to an M2-like phenotype and exhibited pro-osteogenic properties on bone marrow-derived mesenchymal stem cells (BMSCs). Histological analysis and micro-CT results of a rat skull defect model also showed that the telmisartan-loaded scaffolds induced a higher M2/M1 ratio, less inflammatory infiltration, and better bone regenerative patterns. Furthermore, activation of the BMP2 (bone morphogenetic protein-2)-Smad signaling pathway was found to be dominant in telmisartan-loaded scaffold-mediated macrophage-BMSC interactions. These findings indicate that telmisartan incorporation with PCL/PVP nanofibrous scaffolds is a potential therapeutic strategy for promoting bone healing by modulating M1 macrophages to a more M2 phenotype at early stages of postimplantation.
Collapse
Affiliation(s)
- Siyu Wu
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
- Dalian Medical University, Dalian 116044, China
| | - Jiayi Ma
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
- Dalian Medical University, Dalian 116044, China
| | - Jun Liu
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
- Dalian Medical University, Dalian 116044, China
| | - Chun Liu
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
| | - Su Ni
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
| | - Ting Dai
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
| | - Yan Wang
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
| | - Yiping Weng
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
| | - Hongbin Zhao
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
| | - Dong Zhou
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213164, China
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, U.K
| |
Collapse
|
32
|
[Research progress of antibacterial modification of orthopaedic implants surface]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:511-516. [PMID: 35426294 PMCID: PMC9011072 DOI: 10.7507/1002-1892.202112109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To summarize the related research progress of antibacterial modification of orthopaedic implants surface in recent years. METHODS The domestic and foreign related literature in recent years was extensively consulted, the research progress on antibacterial modification of orthopaedic implants surface was discussed from two aspects of characteristics of infection in orthopedic implants and surface anti-infection modification. RESULTS The orthopaedic implants infections are mainly related to aspects of bacterial adhesion, decreased host immunity, and surface biofilm formation. At present, the main antimicrobial coating methods of orthopaedic implants are antibacterial adhesion coating, antibiotic coating, inorganic antimicrobial coating, composite antimicrobial coating, nitric oxide coating, immunomodulation, three-dimensional printing, polymer antimicrobial coating, and "smart" coating. CONCLUSION The above-mentioned antibacterial coating methods of orthopedic implants can not only inhibit bacterial adhesion, but also solve the problems of low immunity and biofilm formation. However, its mechanism of action and modification are still controversial and require further research.
Collapse
|
33
|
Zhou L, You J, Wang Z, Gu Y, Chen D, Lin B, Zhao X, Lin J, Lin J, Liu W. 3D printing monetite-coated Ti-6Al-4V surface with osteoimmunomodulatory function to enhance osteogenesis. BIOMATERIALS ADVANCES 2022; 134:112562. [PMID: 35525756 DOI: 10.1016/j.msec.2021.112562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/18/2021] [Accepted: 11/21/2021] [Indexed: 06/14/2023]
Abstract
Titanium and its alloys are widely used in orthopedic implant surgery due to their good mechanical properties and biocompatibility. Recent studies have shown that the healing process of fractures involve not only the calcification of osteoblasts but also the regulation of the immune system. The functionalization of titanium surface coatings is one of the most important methods for solving implant failures. In this study, monetite (CaHPO4) was coated on the Ti-6Al-4V porous scaffold by hydrothermal method. SEM, XRD and EDS were used to characterize the morphology, phase constitutes, elemental content of the coating, respectively. The results indicated that a well bonded and uniformly distributed monetite coating obtained, and the degradation performance and Ca2+ release of the surface coating were also studied. In terms of biology, live/dead staining and CCK8 methods showed the coating had good biocompatibility and BMSCs can adhere and proliferate on the surface. Flow cytometry and ELISA indicated that the surface monetite-coating had good anti-inflammatory properties. Through RNA-seq analysis, it was shown in KEGG that the osteoclast-related pathway was inhibited. In vitro, monetite induced osteogenic gene expression in BMSCs and inhibited the activity of osteoclasts. In vivo experiments showed that the monetite-coating increased bone formation. In summary, monetite-coating can effectively promote the osteogenesis in BMSCs, which may be achieved through bone immune regulation.
Collapse
Affiliation(s)
| | - Jiacheng You
- Key Laboratory of Optoelectronic Materials Chemistry and Physics, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Zhenyu Wang
- Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yang Gu
- Department of Trauma Orthopedics Surgery, Ningbo No.6 Hospital, Medical School of Ningbo University, 315000, China
| | - Dehui Chen
- Fujian Medical University, Fuzhou 350001, China
| | - Bin Lin
- Fujian Medical University, Fuzhou 350001, China
| | - Xin Zhao
- Fujian Medical University, Fuzhou 350001, China
| | - Jiemin Lin
- Fujian Medical University, Fuzhou 350001, China
| | - Jinxin Lin
- Key Laboratory of Optoelectronic Materials Chemistry and Physics, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
| | - Wenge Liu
- Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
34
|
Zuo K, Wang L, Wang Z, Yin Y, Du C, Liu B, Sun L, Li X, Xiao G, Lu Y. Zinc-Doping Induces Evolution of Biocompatible Strontium-Calcium-Phosphate Conversion Coating on Titanium to Improve Antibacterial Property. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7690-7705. [PMID: 35114085 DOI: 10.1021/acsami.1c23631] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Implant-associated infections (IAI) remains a common and devastating complication in orthopedic surgery. To reduce the incidence of IAI, implants with intrinsic antibacterial activity have been proposed. The surface functionalization and structure optimization of metallic implants can be achieved by surface modification using the phosphate chemical conversion (PCC) technique. Zinc (Zn) has strong antibacterial behavior toward a broad-spectrum of bacteria. Herein, Zn was incorporated into strontium-calcium-phosphate (SrCaP) coatings on titanium (Ti) via PCC method, and the influence of its doping amount on the phase, microstructure, antibacterial activity, and biocompatibility of the composite coating was researched. The results indicated that traces of Zn doping produced grain refinement of SrCaP coating with no significant effect on its phase and surface properties, while a higher Zn content induced its phase and microstructure transformed into zinc-strontium-phosphate (SrZn2(PO4)2). SrCaP-Zn1 and SrCaP-Zn4 represented trace and high content Zn-doped coatings, respectively, which exhibited a similar bacterial attachment for a short time but showed inhibition of biofilm formation after continuous incubation up to 24 h. The killing rates of SrCaP-Zn1 coating for Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) reached 61.25% and 55.38%, respectively. While that data increased to 83.01% and 71.28% on SrCaP-Zn4 coating due to the more-releasing Zn2+. Furthermore, in vitro culture of MC3T3-E1 cells proved that the Zn-doped coatings also possessed excellent biocompatibility. This study provides a new perception for the phase and microstructural optimization of phosphate coatings on implant surfaces, as well as fabricating promising coatings with excellent biocompatibility and antimicrobial properties against IAI.
Collapse
Affiliation(s)
- Kangqing Zuo
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China
- School of Materials Science and Engineering, Shandong University, Jinan, 250061, China
| | - Lili Wang
- Department of Stomatology, The People's Hospital of Zhaoyuan City, Yantai 264500, China
| | - Zhanghan Wang
- Department of Endodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250012, China
| | - Yixin Yin
- Oral Implantology Center, Ji Nan Stomatological Hospital, Jinan 250001, China
| | - Chunmiao Du
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China
- School of Materials Science and Engineering, Shandong University, Jinan, 250061, China
| | - Bing Liu
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China
- School of Materials Science and Engineering, Shandong University, Jinan, 250061, China
| | - Lanying Sun
- Oral Implantology Center, Ji Nan Stomatological Hospital, Jinan 250001, China
| | - Xiaoyan Li
- Department of Endodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan 250012, China
| | - Guiyong Xiao
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China
- School of Materials Science and Engineering, Shandong University, Jinan, 250061, China
| | - Yupeng Lu
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan 250061, China
- School of Materials Science and Engineering, Shandong University, Jinan, 250061, China
| |
Collapse
|
35
|
Zhou A, Wu B, Yu H, Tang Y, Liu J, Jia Y, Yang X, Xiang L. Current Understanding of Osteoimmunology in Certain Osteoimmune Diseases. Front Cell Dev Biol 2021; 9:698068. [PMID: 34485284 PMCID: PMC8416088 DOI: 10.3389/fcell.2021.698068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/15/2021] [Indexed: 02/05/2023] Open
Abstract
The skeletal system and immune system seem to be two independent systems. However, there in fact are extensive and multiple crosstalk between them. The concept of osteoimmunology was created to describe those interdisciplinary events, but it has been constantly updated over time. In this review, we summarize the interactions between the skeletal and immune systems in the co-development of the two systems and the progress of certain typical bone abnormalities and bone regeneration on the cellular and molecular levels according to the mainstream novel study. At the end of the review, we also highlighted the possibility of extending the research scope of osteoimmunology to other systemic diseases. In conclusion, we propose that osteoimmunology is a promising perspective to uncover the mechanism of related diseases; meanwhile, a study from the point of view of osteoimmunology may also provide innovative ideas and resolutions to achieve the balance of internal homeostasis.
Collapse
Affiliation(s)
- Anqi Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingfeng Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yufei Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiayi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yinan Jia
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyu Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Fu M, Wang F, Lin G. Design and research of bone repair scaffold based on two-way fluid-structure interaction. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 204:106055. [PMID: 33784546 DOI: 10.1016/j.cmpb.2021.106055] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/12/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND AND OBJECTIVE Porous bone repair scaffolds are an important method of repairing bone defects. Fluid flow in the scaffold plays a vital role in tissue differentiation and permeability and fluid shear stress (FSS) are two important factors. The differentiation of bone tissue depends on the osteogenic differentiation of cells, FSS affects cell proliferation and differentiation, and permeability affects the transportation of nutrients and metabolic waste. Therefore, it is necessary to better understand and analyze the FSS on the cell surface and the permeability of the scaffold to obtain better osteogenic performance. METHODS In this study, computational fluid dynamics (CFD) was used to analyze fluid flow in the scaffold. Three structures and nine scaffold unit cell models were designed and the cell models were loaded onto the scaffold surface. Considering cell deformability, the two-way fluid-structure interaction (FSI) method was used to evaluate the FSS on the cell surface. RESULTS The simulation results showed that as the pore size of the scaffold increases, its permeability increases and the FSS decreases. The FSS received on the cell surface was much larger than scaffold surface. Moreover the FSS on the cell surface was distributed in steps. CONCLUSIONS The results showed the permeability of all models matches that of human bone tissue. Based on the cell surface FSS as the criterion, it was found that the spherical-560 scaffold exhibited the best osteogenic performance. This provided a strategy to design a better bone repair scaffold from biological aspects.
Collapse
Affiliation(s)
- Mengguang Fu
- School of Mechanical and Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Fei Wang
- School of Mechanical and Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China
| |
Collapse
|