1
|
Wang MF, Yan T, Gao MC, Han CW, Yan ZQ, Gao YZ, Zhang W, Yi Z. A review of the advances in implant technology: accomplishments and challenges for the design of functionalized surface structures. Biomed Mater 2025; 20:032003. [PMID: 40199334 DOI: 10.1088/1748-605x/adca7c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 04/08/2025] [Indexed: 04/10/2025]
Abstract
Biomedical implants are extensively utilized to replace hard-tissue defects owing to their biocompatibility and remarkable tissue-affinity. The materials and functional design are selected based on the resultant osseointegration level and resistance to infection, and these considerations constitute the dominant research topic in this field. However, high rates of implantation failure and peri-implantitis have been reported. Current research on biomedical-implant design encompasses enhancement of the implant surface properties, such as the roughness, nano/micro topography, and hydrophilicity, along with the realization of advanced features including antibacterial properties and cell and immunomodulation regulation. This review considers the two achievements of contemporary implant manufacturing; namely, osseointegration and the realization of antibacterial properties. Present mainstream surface modifications and coatings are discussed, along with functional design technologies and achievements. The impacts of direct surface-treatment techniques and osteogenic functional coatings on osseointegration performance and antibacterial surface structures are elucidated, considering inorganic and organic coatings with antibacterial properties as well as antibiotic-releasing coatings. Furthermore, this review highlights recent advancements in physically driven antimicrobial strategies. Expanding upon existing research, future directions for implant studies are proposed, including the realization of comprehensive functionality that integrates osseointegration and antibacterial properties, as well as patient-specific design. Our study presents a comprehensive review and offers a novel perspective on the design of biomedical implants for enhanced versatility. An in-depth exploration of future research directions will also stimulate subsequent investigations.
Collapse
Affiliation(s)
- Ming-Feng Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Tao Yan
- Joint Orthopedics, Xiangyang Hospital Affiliated to Hubei University of Chinese Medicine, Xiangyang, Hubei 441000, People's Republic of China
| | - Ming-Cen Gao
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Cheng-Wei Han
- Liaoning Upcera Co., Ltd, Benxi, Liaoning 117004, People's Republic of China
| | - Zhuo-Qun Yan
- Liaoning Upcera Co., Ltd, Benxi, Liaoning 117004, People's Republic of China
| | - Yu-Zhong Gao
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Wei Zhang
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, People's Republic of China
| | - Zhe Yi
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| |
Collapse
|
2
|
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
|
3
|
Wang Y, Chen Y, Zhou T, Li J, Zhang N, Liu N, Zhou P, Mao Y. A novel multifunctional nanocomposite hydrogel orchestrates the macrophage reprogramming-osteogenesis crosstalk to boost bone defect repair. J Nanobiotechnology 2024; 22:702. [PMID: 39533396 PMCID: PMC11558876 DOI: 10.1186/s12951-024-02996-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Repairing bone defects is a complex cascade reaction process, as immune system regulation, vascular growth, and osteogenic differentiation are essential. Thus, developing a tissue-engineered biomaterial that caters to the complex healing process of bone regeneration remains a major clinical challenge. In the study, Ca2+-TA-rGO (CTAG)/GelMA hydrogels were synthesized by binding Ca2+ using metal chelation to graphene oxide (GO) nanosheets reduced by tannic acid (TA-rGO) and doping them into gelatin methacrylate (GelMA) hydrogels. TA and rGO exhibited biocompatibility and immunomodulatory properties in this composite, while Ca2+ promoted bone formation and angiogenesis. This novel nanocomposite hydrogel demonstrated good mechanical properties, degradability, and conductivity, and it could achieve slow Ca2+ release during bone regeneration. Both in vitro and in vivo experiments revealed that CTAG/GelMA hydrogel modulated macrophage reprogramming and induced a shift from macrophages to healing-promoting M2 macrophages during the inflammatory phase, promoted vascular neovascularization, and facilitated osteoblast differentiation during bone formation. Moreover, CTAG/GelMA hydrogel could downregulate the NF-κB signaling pathway, offering new insights into regulating macrophage reprogramming-osteogenic crosstalk. Conclusively, this novel multifunctional nanocomposite hydrogel provides a multistage treatment for bone and orchestrates macrophage reprogramming-osteogenic crosstalk to boost bone repair.
Collapse
Affiliation(s)
- Ying Wang
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Yedan Chen
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Tao Zhou
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, 233030, China
| | - Jingze Li
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, 233030, China
| | - Na Zhang
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Na Liu
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Pinghui Zhou
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China.
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, 233030, China.
| | - Yingji Mao
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China.
- School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China.
- Anhui Nerve Regeneration Technology and Medical New Materials Engineering Research Center, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
4
|
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
|
5
|
Shi Y, Gu J, Zhang C, Mi R, Ke Z, Xie M, Jin W, Shao C, He Y, Shi J, Xie Z. A Janus Microsphere Delivery System Orchestrates Immunomodulation and Osteoinduction by Fine-tuning Release Profiles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403835. [PMID: 38984921 DOI: 10.1002/smll.202403835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Bone regeneration is a well-orchestrated process synergistically involving inflammation, angiogenesis, and osteogenesis. Therefore, an effective bone graft should be designed to target multiple molecular events and biological demands during the bone healing process. In this study, a biodegradable gelatin methacryloyl (GelMA)-based Janus microsphere delivery system containing calcium phosphate oligomer (CPO) and bone morphogenetic protein-2 (BMP-2) is developed based on natural biological events. The exceptional adjustability of GelMA facilitates the controlled release and on-demand application of biomolecules, and optimized delivery profiles of CPO and BMP-2 are explored. The sustained release of CPO during the initial healing stages contributes to early immunomodulation and promotes mineralization in the late stage. Meanwhile, the administration of BMP-2 at a relatively high concentration within the therapeutic range enhances the osteoinductive property. This delivery system, with fine-tuned release patterns, induces M2 macrophage polarization and creates a conducive immuno-microenvironment, which in turn facilitates effective bone regeneration in vivo. Collectively, this study proposes a bottom-up concept, aiming to develop a user-friendly and easily controlled delivery system targeting individual biological events, which may offer a new perspective on developing function-optimized biomaterials for clinical use.
Collapse
Affiliation(s)
- Yang Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jingyi Gu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Chun Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Rui Mi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhiwei Ke
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Mingjun Xie
- Plastic and Reconstructive Surgery Center, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Wenjing Jin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Changyu Shao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- The Second Affiliated Hospital of Zhejiang University and State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310027, China
| | - Jue Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
6
|
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
|
7
|
Ru X, Yang L, Shen G, Wang K, Xu Z, Bian W, Zhu W, Guo Y. Microelement strontium and human health: comprehensive analysis of the role in inflammation and non-communicable diseases (NCDs). Front Chem 2024; 12:1367395. [PMID: 38606081 PMCID: PMC11007224 DOI: 10.3389/fchem.2024.1367395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/08/2024] [Indexed: 04/13/2024] Open
Abstract
Strontium (Sr), a trace element with a long history and a significant presence in the Earth's crust, plays a critical yet often overlooked role in various biological processes affecting human health. This comprehensive review explores the multifaceted implications of Sr, especially in the context of non-communicable diseases (NCDs) such as cardiovascular diseases, osteoporosis, hypertension, and diabetes mellitus. Sr is predominantly acquired through diet and water and has shown promise as a clinical marker for calcium absorption studies. It contributes to the mitigation of several NCDs by inhibiting oxidative stress, showcasing antioxidant properties, and suppressing inflammatory cytokines. The review delves deep into the mechanisms through which Sr interacts with human physiology, emphasizing its uptake, metabolism, and potential to prevent chronic conditions. Despite its apparent benefits in managing bone fractures, hypertension, and diabetes, current research on Sr's role in human health is not exhaustive. The review underscores the need for more comprehensive studies to solidify Sr's beneficial associations and address the gaps in understanding Sr intake and its optimal levels for human health.
Collapse
Affiliation(s)
- Xin Ru
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Lida Yang
- College of Nursing, Mudanjiang Medical University, Mudanjiang, China
| | - Guohui Shen
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Kunzhen Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Zihan Xu
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Wenbo Bian
- Zibo Agricultural Science Research Institute, Shandong, China
- Digital Agriculture and Rural Research Institute of CAAS (Zibo), Shandong, China
| | - Wenqi Zhu
- Agricultural Information Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanzhi Guo
- Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
8
|
Zhang X, Zhou W, Xi W. Advancements in incorporating metal ions onto the surface of biomedical titanium and its alloys via micro-arc oxidation: a research review. Front Chem 2024; 12:1353950. [PMID: 38456182 PMCID: PMC10917964 DOI: 10.3389/fchem.2024.1353950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/31/2024] [Indexed: 03/09/2024] Open
Abstract
The incorporation of biologically active metallic elements into nano/micron-scale coatings through micro-arc oxidation (MAO) shows significant potential in enhancing the biological characteristics and functionality of titanium-based materials. By introducing diverse metal ions onto titanium implant surfaces, not only can their antibacterial, anti-inflammatory and corrosion resistance properties be heightened, but it also promotes vascular growth and facilitates the formation of new bone tissue. This review provides a thorough examination of recent advancements in this field, covering the characteristics of commonly used metal ions and their associated preparation parameters. It also highlights the diverse applications of specific metal ions in enhancing osteogenesis, angiogenesis, antibacterial efficacy, anti-inflammatory and corrosion resistance properties of titanium implants. Furthermore, the review discusses challenges faced and future prospects in this promising area of research. In conclusion, the synergistic approach of micro-arc oxidation and metal ion doping demonstrates substantial promise in advancing the effectiveness of biomedical titanium and its alloys, promising improved outcomes in medical implant applications.
Collapse
Affiliation(s)
- Xue’e Zhang
- Jiangxi Province Key Laboratory of Oral Biomedicine, School of Stomatology, Jiangxi Medical College, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, China
| | - Wuchao Zhou
- Jiangxi Province Key Laboratory of Oral Biomedicine, The Affiliated Stomatological Hospital, Jiangxi Medical College, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, China
| | - Weihong Xi
- Jiangxi Province Key Laboratory of Oral Biomedicine, The Affiliated Stomatological Hospital, Jiangxi Medical College, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Zhao Q, Ni Y, Wei H, Duan Y, Chen J, Xiao Q, Gao J, Yu Y, Cui Y, Ouyang S, Miron RJ, Zhang Y, Wu C. Ion incorporation into bone grafting materials. Periodontol 2000 2024; 94:213-230. [PMID: 37823468 DOI: 10.1111/prd.12533] [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: 06/30/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023]
Abstract
The use of biomaterials in regenerative medicine has expanded to treat various disorders caused by trauma or disease in orthopedics and dentistry. However, the treatment of large and complex bone defects presents a challenge, leading to a pressing need for optimized biomaterials for bone repair. Recent advances in chemical sciences have enabled the incorporation of therapeutic ions into bone grafts to enhance their performance. These ions, such as strontium (for bone regeneration/osteoporosis), copper (for angiogenesis), boron (for bone growth), iron (for chemotaxis), cobalt (for B12 synthesis), lithium (for osteogenesis/cementogenesis), silver (for antibacterial resistance), and magnesium (for bone and cartilage regeneration), among others (e.g., zinc, sodium, and silica), have been studied extensively. This review aims to provide a comprehensive overview of current knowledge and recent developments in ion incorporation into biomaterials for bone and periodontal tissue repair. It also discusses recently developed biomaterials from a basic design and clinical application perspective. Additionally, the review highlights the importance of precise ion introduction into biomaterials to address existing limitations and challenges in combination therapies. Future prospects and opportunities for the development and optimization of biomaterials for bone tissue engineering are emphasized.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yueqi Ni
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Hongjiang Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yiling Duan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Jingqiu Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Qi Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Jie Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yiqian Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yu Cui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Simin Ouyang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
10
|
Wu J, Cheng X, Wu J, Chen J, Pei X. The development of magnesium-based biomaterials in bone tissue engineering: A review. J Biomed Mater Res B Appl Biomater 2024; 112:e35326. [PMID: 37861271 DOI: 10.1002/jbm.b.35326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/15/2023] [Accepted: 08/23/2023] [Indexed: 10/21/2023]
Abstract
Bone regeneration is a vital clinical challenge in massive or complicated bone defects. Recently, bone tissue engineering has come to the fore to meet the demand for bone repair with various innovative materials. However, the reported materials usually cannot satisfy the requirements, such as ideal mechanical and osteogenic properties, as well as biocompatibility at the same time. Mg-based biomaterials have considerable potential in bone tissue engineering owing to their excellent mechanical strength and biosafety. Moreover, the biocompatibility and osteogenic activity of Mg-based biomaterials have been the research focuses in recent years. The main limitation faced in the applications of Mg-based biomaterials is rapid degradation, which can produce excessive Mg2+ and hydrogen, affecting the healing of the bone defect. In order to overcome the limitations, researchers have explored several ways to improve the properties of Mg-based biomaterials, including alloying, surface modification with coatings, and synthesizing other composite materials to control the degradation rate upon implantation. This article reviewed the osteogenic mechanism and requirement for appropriate degradation rate and focused on current progress in the biomedical use of Mg-based biomaterials to inspire more clinical applications of Mg in bone regeneration in the future.
Collapse
Affiliation(s)
- Jiaxin Wu
- 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, Sichuan, China
| | - Xinting Cheng
- 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, Sichuan, China
| | - Jicenyuan Wu
- 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, Sichuan, China
| | - Junyu Chen
- 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, Sichuan, China
| | - Xibo Pei
- 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, Sichuan, China
| |
Collapse
|
11
|
Yang X, Wang Q, Yan C, Huang D, Zhang Y, He H, Xiong S, Li C, Chen P, Ye T, Hu D, Wang L. A dual-functional strontium-decorated titanium implants that guides the immune response for osseointegration of osteoporotic rats. Colloids Surf B Biointerfaces 2024; 233:113643. [PMID: 37995629 DOI: 10.1016/j.colsurfb.2023.113643] [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: 09/13/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023]
Abstract
Due to the dynamic imbalance between osteogenesis and osteoclasis and the abnormal inflammatory microenvironment in situ, osteoporosis hampers the early osseointegration between implants and bones. To improve osseointegration with the osteoporosis, we first coated the titanium implants (Ti) with polydopamine (PDA) coating (Ti-PDA), followed by modification with strontium (Sr) to prepare the Ti-PDA-Sr implants. An osteoporotic rat model with femoral bone defect was verified to estimate the osseointegration of the implants. The Ti-PDA-Sr implants exhibited good biocompatibility with continuous release of Sr ions for up to 21 days. Ti-PDA-Sr implants promoted the osteogenesis of BMSCs and the polarization of BMMs to M2 phenotype compared to that of Ti and Ti-PDA implants, revealing the double-regulated effects in bone induction and immune regulation. According to the Micro-CT and histopathology results, Ti-PDA-Sr implants exhibited the most stable osseointegration between bone tissues and implants. According to the immunohistochemistry results, the Ti-PDA-Sr implants differentiated the BMMs to M2 phenotype, alleviating the abnormal inflammation in osteoporosis and preventing the consistent bone destruction between the implants and bone tissues. This study provides a practical and effective strategy in preparing bi-functional implants that can promote osseointegration with osteoporosis.
Collapse
Affiliation(s)
- Xin Yang
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Qiang Wang
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Chaoxi Yan
- Department of Orthopedics, Renmin Hospital of Zhijiang, Yichang 443200, Hubei, China
| | - Degang Huang
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Yinchang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Huazheng He
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Shouliang Xiong
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Congming Li
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Pingbo Chen
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Tingjun Ye
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China.
| | - Dan Hu
- Department of Orthopedics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215500, Jiangsu, China.
| | - Lei Wang
- Department of Orthopedics, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China.
| |
Collapse
|
12
|
Byun H, Jang GN, Jeong H, Lee J, Huh SJ, Lee S, Kim E, Shin H. Development of a composite hydrogel incorporating anti-inflammatory and osteoinductive nanoparticles for effective bone regeneration. Biomater Res 2023; 27:132. [PMID: 38087321 PMCID: PMC10717596 DOI: 10.1186/s40824-023-00473-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/29/2023] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Bone tissue regeneration is regulated by complex events, including inflammation, osteoinduction, and remodeling. Therefore, to induce the complete restoration of defective bone tissue, biomaterials with the ability to regulate the collective bone regenerative system are beneficial. Although some studies conclude that reducing reactive oxygen species created a favorable environment for bone regeneration by controlling inflammation, biomaterials that can simultaneously promote osteogenesis and regulate inflammation have not been developed. Herein, we describe the development of a multi-functional nanoparticle and its hydrogel composite with osteoinductive, anti-inflammatory, and osteoclast-maturation regulatory functions for enhanced bone regeneration. METHODS Tannic acid-mineral nanoparticles (TMP) were prepared by self-assembly of tannic acid in an ion-rich simulated body fluid containing Ca2+ and PO43-. Particles with a diameter of 443 ± 91 nm were selected for their stable spherical morphology and minimal tendency to aggregate. The particles were homogeneously embedded within a gelatin-based cryogel (TMP/Gel) to be used in further experiments. The osteoinductive properties, anti-inflammatory and osteoclast-maturation regulatory functions in vitro were tested by culturing corresponding cells on either TMP/Gel or a gelatin-based cryogel without the particles (Gel). For in vivo analyses, a murine calvarial defect model was used. Statistical analyses were carried out using a Graphpad Prism 7 software (San Diego, CA, USA) to perform one-way analysis of variance ANOVA with Tukey's honest significant difference test and a Student's t-test (for two variables) (P < 0.05). RESULTS Excellent biocompatibility and radical scavenging abilities were exhibited by the TMP/Gel. The expression of osteogenic mRNA is significantly increased in human adipose-derived stem cells seeded on the TMP/Gel compared to those without the particles. Furthermore, RAW264.7 cells seeded on the TMP/Gel displayed significantly lower-than-normal levels of pro-inflammatory and osteoclastogenic genes. Finally, the in vivo results indicated that, compared with the cryogel with no anti-inflammatory effect, the TMP/Gel significantly enhanced both the quality and quantity of newly formed bone, demonstrating the importance of combining anti-inflammation with osteoinduction. CONCLUSION Collectively, these findings suggest our nanoparticle-hydrogel composite could be an effective tool to regulate complex events within the bone healing process.
Collapse
Affiliation(s)
- Hayeon Byun
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Gyu Nam Jang
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Hyewoo Jeong
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Jinkyu Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Seung Jae Huh
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Eunhyung Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
- Department of Bioengineering, BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
- Institute of Nano Science and Technology, Hanyang University, 222 Wangsimri-ro, Seongdong- gu, Seoul, 04763, Republic of Korea.
| |
Collapse
|
13
|
Yang S, Sun Y, Kapilevich L, Zhang X, Huang Y. Protective effects of curcumin against osteoporosis and its molecular mechanisms: a recent review in preclinical trials. Front Pharmacol 2023; 14:1249418. [PMID: 37790808 PMCID: PMC10544586 DOI: 10.3389/fphar.2023.1249418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Osteoporosis (OP) is one of the most common metabolic skeletal disorders and is commonly seen in the elderly population and postmenopausal women. It is mainly associated with progressive loss of bone mineral density, persistent deterioration of bone microarchitecture, and increased fracture risk. To date, drug therapy is the primary method used to prevent and treat osteoporosis. However, long-term drug therapy inevitably leads to drug resistance and specific side effects. Therefore, researchers are constantly searching for new monomer compounds from natural plants. As a candidate for the treatment of osteoporosis, curcumin (CUR) is a natural phenolic compound with various pharmacological and biological activities, including antioxidant, anti-apoptotic, and anti-inflammatory. This compound has gained research attention for maintaining bone health in various osteoporosis models. We reviewed preclinical and clinical studies of curcumin in preventing and alleviating osteoporosis. These results suggest that if subjected to rigorous pharmacological and clinical trials, naturally-derived curcumin could be used as a complementary and alternative medicine for the treatment of osteoporosis by targeting osteoporosis-related mechanistic pathways. This review summarizes the mechanisms of action and potential therapeutic applications of curcumin in the prevention and mitigation of osteoporosis and provides reference for further research and development of curcumin.
Collapse
Affiliation(s)
- Shenglei Yang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yuying Sun
- School of Stomatology, Binzhou Medical College, Yantai, China
| | - Leonid Kapilevich
- Faculty of Physical Education, Nаtionаl Reseаrch Tomsk Stаte University, Tomsk, Russiа
| | - Xin’an Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yue Huang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
14
|
Bosch-Rué È, Díez-Tercero L, Buitrago JO, Castro E, Pérez RA. Angiogenic and immunomodulation role of ions for initial stages of bone tissue regeneration. Acta Biomater 2023; 166:14-41. [PMID: 37302735 DOI: 10.1016/j.actbio.2023.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/10/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
It is widely known that bone has intrinsic capacity to self-regenerate after injury. However, the physiological regeneration process can be impaired when there is an extensive damage. One of the main reasons is due to the inability to establish a new vascular network that ensures oxygen and nutrient diffusion, leading to a necrotic core and non-junction of bone. Initially, bone tissue engineering (BTE) emerged to use inert biomaterials to just fill bone defects, but it eventually evolved to mimic bone extracellular matrix and even stimulate bone physiological regeneration process. In this regard, the stimulation of osteogenesis has gained a lot of attention especially in the proper stimulation of angiogenesis, being critical to achieve a successful osteogenesis for bone regeneration. Besides, the immunomodulation of a pro-inflammatory environment towards an anti-inflammatory one upon scaffold implantation has been considered another key process for a proper tissue restoration. To stimulate these phases, growth factors and cytokines have been extensively used. Nonetheless, they present some drawbacks such as low stability and safety concerns. Alternatively, the use of inorganic ions has attracted higher attention due to their higher stability and therapeutic effects with low side effects. This review will first focus in giving fundamental aspects of initial bone regeneration phases, focusing mainly on inflammatory and angiogenic ones. Then, it will describe the role of different inorganic ions in modulating the immune response upon biomaterial implantation towards a restorative environment and their ability to stimulate angiogenic response for a proper scaffold vascularization and successful bone tissue restoration. STATEMENT OF SIGNIFICANCE: The impairment of bone tissue regeneration when there is excessive damage has led to different tissue engineered strategies to promote bone healing. Significant importance has been given in the immunomodulation towards an anti-inflammatory environment together with proper angiogenesis stimulation in order to achieve successful bone regeneration rather than stimulating only the osteogenic differentiation. Ions have been considered potential candidates to stimulate these events due to their high stability and therapeutic effects with low side effects compared to growth factors. However, up to now, no review has been published assembling all this information together, describing individual effects of ions on immunomodulation and angiogenic stimulation, as well as their multifunctionality or synergistic effects when combined together.
Collapse
Affiliation(s)
- Èlia Bosch-Rué
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Josep Trueta, s/n, Sant Cugat del Vallès, Barcelona 08195, Spain; Basic Sciences Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Leire Díez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Josep Trueta, s/n, Sant Cugat del Vallès, Barcelona 08195, Spain; Basic Sciences Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Jenifer Olmos Buitrago
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Josep Trueta, s/n, Sant Cugat del Vallès, Barcelona 08195, Spain; Basic Sciences Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Emilio Castro
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Josep Trueta, s/n, Sant Cugat del Vallès, Barcelona 08195, Spain; Basic Sciences Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Roman A Pérez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Josep Trueta, s/n, Sant Cugat del Vallès, Barcelona 08195, Spain; Basic Sciences Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Barcelona 08195, Spain.
| |
Collapse
|
15
|
Frase D, Lee C, Nachiappan C, Gupta R, Akkouch A. The Inflammatory Contribution of B-Lymphocytes and Neutrophils in Progression to Osteoporosis. Cells 2023; 12:1744. [PMID: 37443778 PMCID: PMC10340451 DOI: 10.3390/cells12131744] [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: 06/04/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Osteoporosis is a bone disease characterized by structural deterioration and low bone mass, leading to fractures and significant health complications. In this review, we summarize the mechanisms by which B-lymphocytes and neutrophils contribute to the development of osteoporosis and potential therapeutics targeting these immune mediators to reduce the proinflammatory milieu. B-lymphocytes-typically appreciated for their canonical role in adaptive, humoral immunity-have emerged as critical regulators of bone remodeling. B-lymphocytes communicate with osteoclasts and osteoblasts through various cytokines, including IL-7, RANK, and OPG. In inflammatory conditions, B-lymphocytes promote osteoclast activation and differentiation. However, B-lymphocytes also possess immunomodulatory properties, with regulatory B-lymphocytes (Bregs) secreting TGF-β1 to restrain pathogenic osteoclastogenesis. Neutrophils, the body's most prevalent leukocyte, also contribute to the proinflammatory environment that leads to osteoporotic bone remodeling. In aged individuals, neutrophils display reduced chemotaxis, phagocytosis, and apoptosis. Understanding the delicate interplay between B-lymphocytes and neutrophils in the context of impaired bone metabolism is crucial for targeted therapies for osteoporosis.
Collapse
Affiliation(s)
- Drew Frase
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Chi Lee
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Chidambaram Nachiappan
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Richa Gupta
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
| | - Adil Akkouch
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA; (D.F.)
- Department of Orthopaedic Surgery and Medical Engineering Program, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| |
Collapse
|
16
|
Ren L, Gong P, Gao X, Wang Q, Xie L, Tang W, Long J, Liu C, Tian W, He M. Metal-phenolic networks acted as a novel bio-filler of a barrier membrane to improve guided bone regeneration via manipulating osteoimmunomodulation. J Mater Chem B 2022; 10:10128-10138. [PMID: 36468640 DOI: 10.1039/d2tb01804g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A guided bone tissue regeneration membrane (GBRM) is traditionally viewed as an inert physical barrier to isolate soft tissue from the bone defect area. However, as a "foreign body", the implantation of a GBRM would inevitably modulate immune response and subsequently affect bone dynamics. Herein, we developed strontium ion (Sr2+)-based metal-phenolic network complexes (MPNs) as a novel type of bio-filler to manipulate the osteoimmunomodulation of the advanced GBRM. For controllable delivery of Sr2+ depending on the difference in affinity between phenolic ligands and Sr2+, tannic acid (TA), epigallocatechin gallate (EGCG), and epigallocatechin (EGC) were selected to chelate with Sr2+. The formed MPNs were incorporated into PCL nanofibrous membranes by blending electrospinning. Among them, TA/Sr based MPN particles displayed the most sustainable release profile of phenolic ligands and Sr2+. Further investigations demonstrated that Sr2+ could not only directly promote osteogenic differentiation of BMSCs, but also manipulate an anti-inflammatory osteoimmune microenvironment in a synergistic manner with TA, thus enhancing osteogenesis and inhibiting bone resorption. The rat alveolar bone defect model also confirmed that the TA/Sr nanoparticle modified membrane displayed better bone regeneration performance than the pure PCL membrane via inhibiting bone resorption. This work provides a new platform for controllable delivery of bioactive nutrient elements, and holds great promise for advancing multi-functional biocomposites.
Collapse
Affiliation(s)
- Lulu Ren
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Pei Gong
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinghui Gao
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Wang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Li Xie
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wei Tang
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jie Long
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Can Liu
- Beijing Jimafei Technology Development Co., LTD, Beijing, China
| | - Weidong Tian
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Min He
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
17
|
Aguilera-Correa JJ, Gisbert-Garzarán M, Mediero A, Fernández-Aceñero MJ, de-Pablo-Velasco D, Lozano D, Esteban J, Vallet-Regí M. Antibiotic delivery from bone-targeted mesoporous silica nanoparticles for the treatment of osteomyelitis caused by methicillin-resistant Staphylococcus aureus. Acta Biomater 2022; 154:608-625. [PMID: 36341887 DOI: 10.1016/j.actbio.2022.10.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
Osteomyelitis is a hard-to-treat infection of the bone and bone marrow that is mainly caused by Staphylococcus aureus, with an increasing incidence of methicillin-resistant S. aureus (MRSA). Owing to the aggressiveness of these bacteria in colonizing and destroying the bone, systemic antibiotic treatments fail to eradicate the infection. Instead, it normally entails surgery to remove the dead or infected bone. In this work, we report bone-targeted mesoporous silica nanoparticles for the treatment of osteomyelitis. The nanoparticles have been engineered with a functional gelatine/colistin coating able to hamper premature release from the mesopores while effectively disaggregating the bacterial biofilm. Because antibiotic resistance is a global emergency, we have designed two sets of identical nanoparticles, carrying each of them a clinically relevant antibiotic, that have demonstrated to have synergistic effect. The bone-targeted nanoparticles have been thoroughly evaluated in vitro and in vivo, obtaining a notable reduction of the amount of bacteria in the bone in just 24 h after only one dose, and paving the way for localized, nanoparticle-mediated treatment of MRSA-caused osteomyelitis. STATEMENT OF SIGNIFICANCE: In this work, we propose the use of bone-targeted mesoporous silica nanoparticles to address S. aureus-caused osteomyelitis that render synergistic therapeutic effect via multidrug delivery. Because the bacterial biofilm is responsible for an aggressive surgical approach and prolonged antibiotic treatment, the nanoparticles have been functionalized with a functional coating able to both disaggregate the biofilm, hamper premature antibiotic release and protect the intact bone. These engineered nanoparticles are able to effectively target bone tissue both in vitro and in vivo, showing high biocompatibility and elevated antibacterial effect.
Collapse
Affiliation(s)
- J J Aguilera-Correa
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - M Gisbert-Garzarán
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - A Mediero
- Bone and Joint Unit, IIS- Fundación Jimenez Diaz, UAM, Avenida Reyes Católicos, 2 28037 Madrid, Spain
| | | | | | - D Lozano
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - J Esteban
- CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain; Clinical Microbiology Department, IIS-Fundación Jiménez Diaz, UAM, Avenida Reyes Católicos, 2 28037 Madrid, Spain.
| | - M Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| |
Collapse
|
18
|
Chai H, Wang W, Yuan X, Zhu C. Bio-Activated PEEK: Promising Platforms for Improving Osteogenesis through Modulating Macrophage Polarization. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120747. [PMID: 36550953 PMCID: PMC9774947 DOI: 10.3390/bioengineering9120747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/30/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022]
Abstract
The attention on orthopedic biomaterials has shifted from their direct osteogenic properties to their osteoimmunomodulation, especially the modulation of macrophage polarization. Presently, advanced technologies endow polyetheretherketone (PEEK) with good osteoimmunomodulation by modifying PEEK surface characteristics or incorporating bioactive substances with regulating macrophage polarization. Recent studies have demonstrated that the fabrication of a hydrophilic surface and the incorporation of bioactive substances into PEEK (e.g., zinc, calcium, and phosphate) are good strategies to promote osteogenesis by enhancing the polarization of M2 macrophages. Furthermore, the modification by other osteoimmunomodulatory composites (e.g., lncRNA-MM2P, IL-4, IL-10, and chitosan) and their controlled and desired release may make PEEK an optimal bio-activated implant for regulating and balancing the osteogenic system and immune system. The purpose of this review is to comprehensively evaluate the potential of bio-activated PEEK in polarizing macrophages into M2 phenotype to improve osteogenesis. For this objective, we retrieved and discussed different kinds of bio-activated PEEK regarding improving osteogenesis through modulating macrophage polarization. Meanwhile, the relevant challenges and outlook were presented. We hope that this review can shed light on the development of bio-activated PEEK with more favorable osteoimmunomodulation.
Collapse
Affiliation(s)
- Haobu Chai
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei 230001, China
| | - Wenzhi Wang
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei 230001, China
| | - Xiangwei Yuan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
- Correspondence: (X.Y.); (C.Z.)
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei 230001, China
- Correspondence: (X.Y.); (C.Z.)
| |
Collapse
|
19
|
Cerqueira A, García-Arnáez I, Muriach M, Azkargorta M, Elortza F, Izquierdo R, Romero-Gavilán F, Gurruchaga M, Suay J, Goñi I. The effect of calcium-magnesium mixtures in sol-gel coatings on bone tissue regeneration. Biomater Sci 2022; 10:5634-5647. [PMID: 35993129 DOI: 10.1039/d2bm00742h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Calcium and magnesium are two elements essential for bone structure and metabolism. However, their synergistic or competitive effects on bone regeneration are often overlooked during biomaterial development. We examined the interactions between Ca and Mg in sol-gel coatings doped with mixtures of CaCl2 (0.5%) and MgCl2 (0.5, 1, and 1.5%). After physicochemical characterisation, the materials were incubated in vitro with MC3T3-E1 osteoblastic cells and RAW264.7 macrophages, and the protein adsorption was analysed using nLC-MS/MS. The incorporation of the ions did not lead to the formation of crystalline structures and did not affect the sol-gel network cross-linking. The release of the ions did not cause cytotoxic effects at any tested concentration. The proteomic analysis showed that adding the Ca and Mg ions elevated the adsorption of proteins associated with inflammatory response regulation (e.g., ALBU, CLUS, HPT, HPTR, A1AG1 and A1AG2) but decreased the adsorption of immunoglobulins. The CaMg coatings had reduced affinity to proteins associated with coagulation (e.g., FA9, FA10, FA11, FA12) but increased the adsorption of proteins involved in cell adhesion (DSG1, DESP, FBLN1, ZA2G). In vitro assays revealed that the cellular response was affected by changing the concentration of Mg. Moreover, our results show that these differences reflect the changes in the concentrations of both ions in the mix but are not a simple additive effect.
Collapse
Affiliation(s)
- Andreia Cerqueira
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain.
| | - Iñaki García-Arnáez
- Departament of Science and Technology of Polymers, Universidad del País Vasco, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - María Muriach
- Deparment of Medicine, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - Félix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - Raúl Izquierdo
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain.
| | - Francisco Romero-Gavilán
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain.
| | - Mariló Gurruchaga
- Departament of Science and Technology of Polymers, Universidad del País Vasco, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - Julio Suay
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain.
| | - Isabel Goñi
- Departament of Science and Technology of Polymers, Universidad del País Vasco, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| |
Collapse
|
20
|
You J, Zhang Y, Zhou Y. Strontium Functionalized in Biomaterials for Bone Tissue Engineering: A Prominent Role in Osteoimmunomodulation. Front Bioeng Biotechnol 2022; 10:928799. [PMID: 35875505 PMCID: PMC9298737 DOI: 10.3389/fbioe.2022.928799] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
With the development of bone tissue engineering bio-scaffold materials by adding metallic ions to improve bone healing have been extensively explored in the past decades. Strontium a non-radioactive element, as an essential osteophilic trace element for the human body, has received widespread attention in the medical field due to its superior biological properties of inhibiting bone resorption and promoting osteogenesis. As the concept of osteoimmunology developed, the design of orthopedic biomaterials has gradually shifted from “immune-friendly” to “immunomodulatory” with the aim of promoting bone healing by modulating the immune microenvironment through implanted biomaterials. The process of bone healing can be regarded as an immune-induced procedure in which immune cells can target the effector cells such as macrophages, neutrophils, osteocytes, and osteoprogenitor cells through paracrine mechanisms, affecting pathological alveolar bone resorption and physiological bone regeneration. As a kind of crucial immune cell, macrophages play a critical role in the early period of wound repair and host defense after biomaterial implantation. Despite Sr-doped biomaterials being increasingly investigated, how extracellular Sr2+ guides the organism toward favorable osteogenesis by modulating macrophages in the bone tissue microenvironment has rarely been studied. This review focuses on recent knowledge that the trace element Sr regulates bone regeneration mechanisms through the regulation of macrophage polarization, which is significant for the future development of Sr-doped bone repair materials. We will also summarize the primary mechanism of Sr2+ in bone, including calcium-sensing receptor (CaSR) and osteogenesis-related signaling pathways.
Collapse
Affiliation(s)
- Jiaqian You
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yidi Zhang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanmin Zhou
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
21
|
Yu D, Guo S, Yu M, Liu W, Li X, Chen D, Li B, Guo Z, Han Y. Immunomodulation and osseointegration activities of Na 2TiO 3 nanorods-arrayed coatings doped with different Sr content. Bioact Mater 2022; 10:323-334. [PMID: 34901549 PMCID: PMC8636710 DOI: 10.1016/j.bioactmat.2021.08.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 01/17/2023] Open
Abstract
To endow Ti-based orthopedic implants immunomodulatory capability and thus enhanced osseointegration, different amounts of Sr are doped in Na2TiO3 nanorods in the arrays with identical nanotopographic parameters (rod diameter, length and inter-rod spacing) by substitution of Na+ using hydrothermal treatment. The obtained arrays are denoted as STSr2, STSr4, and STSr7, where the arabic numbers indicate the incorporating amounts of Sr in Na2TiO3. The modulation effects of the Sr-doped nanorods arrays on macrophage polarization and osteogenetic functions of osteoblasts are investigated, together with the array without Sr (ST). Moreover, osseointegration of these arrays are also assayed in rat femoral condyles. Sr-doped nanorods arrays accelerate M1 (pro-inflammatory phenotype)-to-M2 (anti-inflammatory phenotype) transformation of the adhered macrophages, enhancing secretion of pro-osteogenetic cytokines and growth factors (TGF-β1 and BMP2), moreover, the Sr doped arrays directly enhance osteogenetic functions of osteoblasts. The enhancement of paracrine of M2 macrophages and osteogenetic function of osteoblasts is promoted with the increase of Sr incorporating amounts. Consequently, Sr doped arrays show significantly enhanced osseointegration in vivo compared to ST, and STSr7 exhibits the best performance. Our work sheds a new light on the design of surface chemical components and structures for orthopedic implants to enhance their osseointegration.
Collapse
Affiliation(s)
- Dongmei Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Shuo Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Meng Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Wenwen Liu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Xiaokang Li
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Bo Li
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Zheng Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Yong Han
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| |
Collapse
|
22
|
Huang L, Chen W, Wei L, Su Y, Liang J, Lian H, Wang H, Long F, Yang F, Gao S, Tan Z, Xu J, Zhao J, Liu Q. Lonafarnib Inhibits Farnesyltransferase via Suppressing ERK Signaling Pathway to Prevent Osteoclastogenesis in Titanium Particle-Induced Osteolysis. Front Pharmacol 2022; 13:848152. [PMID: 35300293 PMCID: PMC8921770 DOI: 10.3389/fphar.2022.848152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
Wear debris after total joint arthroplasty can attract the recruitment of macrophages, which release pro-inflammatory substances, triggering the activation of osteoclasts, thereby leading to periprosthetic osteolysis (PPOL) and aseptic loosening. However, the development of pharmacological strategies targeting osteoclasts to prevent periprosthetic osteolysis has not been fruitful. In this study, we worked toward researching the effects and mechanisms of a farnesyltransferase (FTase) inhibitor Lonafarnib (Lon) on receptor activator of nuclear factor κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis and bone resorption, as well as the impacts of Lon on titanium particle-induced osteolysis. To investigate the impacts of Lon on bone resorption and osteoclastogenesis in vitro, bone marrow macrophages were incubated and stimulated with RANKL and macrophage colony-stimulating factor (M-CSF). The influence of Lon on osteolysis prevention in vivo was examined utilizing a titanium particle-induced mouse calvarial osteolysis model. The osteoclast-relevant genes expression was explored by real-time quantitative PCR. Immunofluorescence was used to detect intracellular localization of nuclear factor of activated T cells 1 (NFATc1). SiRNA silence assay was applied to examine the influence of FTase on osteoclasts activation. Related signaling pathways, including NFATc1 signaling, NF-κB, mitogen-activated protein kinases pathways were identified by western blot assay. Lon was illustrated to suppress bone resorptive function and osteoclastogenesis in vitro, and it also reduced the production of pro-inflammatory substances and prevented titanium particle-induced osteolysis in vivo. Lon decreased the expression of osteoclast-relevant genes and suppressed NFATc1 nuclear translocation and auto-amplification. Mechanistically, Lon dampened FTase, and inhibition of FTase reduced osteoclast formation by suppressing ERK signaling. Lon is a promising treatment option for osteoclast-related osteolysis diseases including periprosthetic osteolysis by targeted inhibition of FTase through suppressing ERK signaling.
Collapse
Affiliation(s)
- Linke Huang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Weiwei Chen
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Linhua Wei
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China.,The Affiliated Nanning Infectious Disease Hospital of Guangxi Medical University, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yuangang Su
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Jiamin Liang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Haoyu Lian
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Hui Wang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Feng Long
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Fan Yang
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Shiyao Gao
- Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhen Tan
- Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Qian Liu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
23
|
Yu D, Li B, Yu M, Guo S, Guo Z, Han Y. Cubic multi-ions-doped Na2TiO3 nanorod-like coatings: Structure-stable, highly efficient platform for ions-exchanged release to immunomodulatory promotion on vascularized bone apposition. Bioact Mater 2022; 18:72-90. [PMID: 35387170 PMCID: PMC8961311 DOI: 10.1016/j.bioactmat.2022.01.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/10/2022] [Accepted: 01/22/2022] [Indexed: 12/11/2022] Open
Abstract
The dissolution-derived release of bioactive ions from ceramic coatings on metallic implants, despite improving osseointegration, renders a concern on the interfacial breakdown of the metal/coating/bone system during long-term service. Consequently, persistent efforts to seek alternative strategies instead of dissolution-derived activation are pressingly carrying out. Inspired by bone mineral containing ions as Ca2+, Mg2+, Sr2+ and Zn2+, here we hydrothermally grew the quadruple ions co-doped Na2TiO3 nanorod-like coatings. The co-doped ions partially substitute Na+ in Na2TiO3, and can be efficiently released from cubic lattice via exchange with Na+ in fluid rather than dissolution, endowing the coatings superior long-term stability of structure and bond strength. Regulated by the coatings-conditioned extracellular ions, TLR4-NFκB signalling is enhanced to act primarily in macrophages (MΦs) at 6 h while CaSR-PI3K-Akt1 signalling is potentiated to act predominately since 24 h, triggering MΦs in a M1 response early and then in a M2 response to sequentially secrete diverse cytokines. Acting on endothelial and mesenchymal stem cells with the released ions and cytokines, the immunomodulatory coatings greatly promote Type-H (CD31hiEmcnhi) angiogenesis and osteogenesis in vitro and in vivo, providing new insights into orchestrating insoluble ceramics-coated implants for early vascularized osseointegration in combination with long-term fixation to bone. Co-doped Ca2+, Mg2+, Sr2+ and Zn2+ in Na2TiO3 efficiently release via ion exchange. QID elevates extracellular concentrations of the ions and MΦ intracellular [Ca2+]. Co-doped Na2TiO3 coatings promote immunomodulatory apposition of vascularized bone.
Collapse
Affiliation(s)
- Dongmei Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Bo Li
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Meng Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Shuo Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Zheng Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
- Corresponding author.
| | - Yong Han
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
- Corresponding author.
| |
Collapse
|
24
|
Vafaei S, Wu X, Tu J, Nematollahi-mahani SN. The Effects of Crocin on Bone and Cartilage Diseases. Front Pharmacol 2022; 12:830331. [PMID: 35126154 PMCID: PMC8807478 DOI: 10.3389/fphar.2021.830331] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Crocin, the main biologically active carotenoid of saffron, generally is derived from the dried trifid stigma of Crocus sativus L. Many studies have demonstrated that crocin has several therapeutic effects on biological systems through its anti-oxidant and anti-inflammatory properties. The wide range of crocin activities is believed to be because of its ability to anchor to many proteins, triggering some cellular pathways responsible for cell proliferation and differentiation. It also has therapeutic potentials in arthritis, osteoarthritis, rheumatoid arthritis, and articular pain probably due to its anti-inflammatory properties. Anti-apoptotic effects, as well as osteoclast inhibition effects of crocin, have suggested it as a natural substance to treat osteoporosis and degenerative disease of bone and cartilage. Different mechanisms underlying crocin effects on bone and cartilage repair have been investigated, but remain to be fully elucidated. The present review aims to undertake current knowledge on the effects of crocin on bone and cartilage degenerative diseases with an emphasis on its proliferative and differentiative properties in mesenchymal stem cells.
Collapse
Affiliation(s)
- Shayan Vafaei
- Department of Anatomical Science, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Xuming Wu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Jiajie Tu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
- *Correspondence: Jiajie Tu, ; Seyed Noureddin Nematollahi-mahani,
| | - Seyed Noureddin Nematollahi-mahani
- Department of Anatomical Science, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- *Correspondence: Jiajie Tu, ; Seyed Noureddin Nematollahi-mahani,
| |
Collapse
|
25
|
Aguilera-Correa J, Gisbert-Garzarán M, Mediero A, Carias-Cálix R, Jiménez-Jiménez C, Esteban J, Vallet-Regí M. Arabic gum plus colistin coated moxifloxacin-loaded nanoparticles for the treatment of bone infection caused by Escherichia coli. Acta Biomater 2022; 137:218-237. [PMID: 34653694 DOI: 10.1016/j.actbio.2021.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/20/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022]
Abstract
Osteomyelitis is an inflammatory process of bone and bone marrow that may even lead to patient death. Even though this disease is mainly caused by Gram-positive organisms, the proportion of bone infections caused by Gram-negative bacteria, such as Escherichia coli, has significantly increased in recent years. In this work, mesoporous silica nanoparticles have been employed as platform to engineer a nanomedicine able to eradicate E. coli- related bone infections. For that purpose, the nanoparticles have been loaded with moxifloxacin and further functionalized with Arabic gum and colistin (AG+CO-coated MX-loaded MSNs). The nanosystem demonstrated high affinity toward E. coli biofilm matrix, thanks to AG coating, and marked antibacterial effect because of the bactericidal effect of moxifloxacin and the disaggregating effect of colistin. AG+CO-coated MX-loaded MSNs were able to eradicate the infection developed on a trabecular bone in vitro and showed pronounced antibacterial efficacy in vivo against an osteomyelitis provoked by E. coli. Furthermore, AG+CO-coated MX-loaded MSNs were shown to be essentially non-cytotoxic with only slight effect on cell proliferation and mild hepatotoxicity, which might be attributed to the nature of both antibiotics. In view of these results, these nanoparticles may be considered as a promising treatment for bone infections caused by enterobacteria, such as E. coli, and introduce a general strategy against bone infections based on the implementation of antibiotics with different but complementary activity into a single nanocarrier. STATEMENT OF SIGNIFICANCE: In this work, we propose a methodology to address E.coli bone infections by using moxifloxacin-loaded mesoporous silica nanoparticles coated with Arabic gum containing colistin (AG+CO-coated MX-loaded MSNs). The in vitro evaluation of this nanosystem demonstrated high affinity toward E. coli biofilm matrix thanks to the Arabic gum coating, a disaggregating and antibacterial effect of colistin, and a remarkable antibiofilm action because of the bactericidal ability of moxifloxacin and colistin. This anti-E. coli capacity of AG+CO-coated MX-loaded MSNs was brought out in an in vivo rabbit model of osteomyelitis where the nanosystem was able to eradicate more than 90% of the bacterial load within the infected bone.
Collapse
|
26
|
Chai H, Sang S, Luo Y, He R, Yuan X, Zhang X. Icariin-loaded Sulfonated Polyetheretherketone with Osteogenesis Promotion and Osteoclastogenesis Inhibition Properties via Immunomodulation for Advanced Osseointegration. J Mater Chem B 2022; 10:3531-3540. [PMID: 35416810 DOI: 10.1039/d1tb02802b] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Preventing prosthesis loosening due to insufficient osseointegration is critical for patients with osteoporosis. Endowing implants with immunomodulatory function can effectively enhance osseointegration. In this work, we loaded icariin (ICA) onto...
Collapse
Affiliation(s)
- Haobu Chai
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| | - Shang Sang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| | - Yao Luo
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| | - Renke He
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| | - Xiangwei Yuan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| |
Collapse
|
27
|
Chung JT, Lau CML, Chau Y. The effect of polysaccharide-based hydrogels on the response of antigen-presenting cell lines to immunomodulators. Biomater Sci 2021; 9:6542-6554. [PMID: 34582528 DOI: 10.1039/d1bm00854d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hydrogel presents as foreign material to the host and participates in immune responses, which skew the biofunctions of immunologic loads (antigen and adjuvants) during in situ DC priming. This study aims to investigate the effect of the hydrogel made from different polysaccharides on macrophage (RAW264.7) activation and DC (JAWSII) modulation. We adopted polysaccharides of different sugar chemistry to fabricate hydrogels. Hyaluronate (HA), glycol chitosan (GC) and dextran (DX) were functionalized with vinyl sulfone and chemically cross-linked with dithiothreitol via thiol-click chemistry. We found that HA reduced macrophage adhesion and activation on the hydrogel surface. GC and DX promoted M1 polarization in terms of higher CCR7 expression and TNF-α, IL-6 production. In terms of DC engagement, GC promoted antigen uptake by JAWSII and all hydrogels promoted antigen presentation on MHC-I molecules. GC and DX favoured the generation of immunogenic DC while accommodating immunostimulatory functions of IFN-γ and polyI:C or LPS during co-incubation. Particularly, the co-incubation of IP with GC promoted CCR7 expression on JAWSII. Conversely, HA was more appropriate for the construction of a tolerogenic DC priming platform. We observed that HA did not induce co-stimulatory markers expression on DC but suppressed the action of LPS in inducing TNF-α generation. Moreover, when immunosuppressive cytokines, IL-10 and TGF-β were added, cytokines' immunosuppressive action was amplified by hydrogel bedding, HA, GC and to a less extent DX in suppressing LPS-induced IL-6 generation from JAWSII. We concluded that HA is preferable for tolerogenic DC development while minimizing the macrophage response in conferring foreign body response, whereas DX and GC are more appropriate for immunogenic DC development. This study demonstrates the potential of polysaccharides in conferring in situ DC priming together with antigen and adjuvant loads while addressing the tradeoff between the foreign body responses and DC engagement by selecting appropriate polysaccharides for the hydrogel platform construction.
Collapse
Affiliation(s)
- Jin Teng Chung
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Chi Ming Laurence Lau
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Ying Chau
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
28
|
Liu W, Yu M, Chen F, Wang L, Ye C, Chen Q, Zhu Q, Xie D, Shao M, Yang L. A novel delivery nanobiotechnology: engineered miR-181b exosomes improved osteointegration by regulating macrophage polarization. J Nanobiotechnology 2021; 19:269. [PMID: 34493305 PMCID: PMC8424816 DOI: 10.1186/s12951-021-01015-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/25/2021] [Indexed: 12/18/2022] Open
Abstract
Background Many patients suffer from implant loosening after the implantation of titanium alloy caused by immune response to the foreign bodies and this could inhibit the following osteogenesis, which could possibly give rise to aseptic loosening and poor osteointegration while there is currently no appropriate solution in clinical practice. Exosome (Exo) carrying miRNA has been proven to be a suitable nanocarrier for solving this problem. In this study, we explored whether exosomes overexpressing miR-181b (Exo-181b) could exert beneficial effect on promoting M2 macrophage polarization, thus inhibiting inflammation as well as promoting osteogenesis and elaborated the underlying mechanism in vitro. Furthermore, we aimed to find whether Exo-181b could enhance osteointegration. Results In vitro, we firstly verified that Exo-181b significantly enhanced M2 polarization and inhibited inflammation by suppressing PRKCD and activating p-AKT. Then, in vivo, we verified that Exo-181b enhanced M2 polarization, reduced the inflammatory response and enhanced osteointegration. Also, we verified that the enhanced M2 polarization could indirectly promote the migration and osteogenic differentiation by secreting VEGF and BMP-2 in vitro. Conclusions Exo-181b could suppress inflammatory response by promoting M2 polarization via activating PRKCD/AKT signaling pathway, which further promoting osteogenesis in vitro and promote osteointegration in vivo. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01015-y.
Collapse
Affiliation(s)
- Wei Liu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Muyu Yu
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Medical Centre of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Key Clinic Centre of Metabolism Disease, Shanghai Institute for Diabetes, Shanghai, China
| | - Feng Chen
- Department of Orthopaedics, Shanghai Fengxian Central Hospital, Branch of the Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 201400, People's Republic of China.,College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Longqing Wang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Cheng Ye
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Qing Chen
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Qi Zhu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Dong Xie
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Mingzhe Shao
- Department of Vascular Surgery, Multidisciplinary Collaboration Group of Diabetic Foot, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Lili Yang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China.
| |
Collapse
|
29
|
Wang D, Tan J, Zhu H, Mei Y, Liu X. Biomedical Implants with Charge-Transfer Monitoring and Regulating Abilities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004393. [PMID: 34166584 PMCID: PMC8373130 DOI: 10.1002/advs.202004393] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/12/2021] [Indexed: 05/06/2023]
Abstract
Transmembrane charge (ion/electron) transfer is essential for maintaining cellular homeostasis and is involved in many biological processes, from protein synthesis to embryonic development in organisms. Designing implant devices that can detect or regulate cellular transmembrane charge transfer is expected to sense and modulate the behaviors of host cells and tissues. Thus, charge transfer can be regarded as a bridge connecting living systems and human-made implantable devices. This review describes the mode and mechanism of charge transfer between organisms and nonliving materials, and summarizes the strategies to endow implants with charge-transfer regulating or monitoring abilities. Furthermore, three major charge-transfer controlling systems, including wired, self-activated, and stimuli-responsive biomedical implants, as well as the design principles and pivotal materials are systematically elaborated. The clinical challenges and the prospects for future development of these implant devices are also discussed.
Collapse
Affiliation(s)
- Donghui Wang
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- School of Materials Science and EngineeringHebei University of TechnologyTianjin300130China
| | - Ji Tan
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
| | - Hongqin Zhu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- Department of Materials ScienceFudan UniversityShanghai200433China
| | - Yongfeng Mei
- Department of Materials ScienceFudan UniversityShanghai200433China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- School of Chemistry and Materials ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| |
Collapse
|
30
|
Li T, He H, Yang Z, Wang J, Zhang Y, He G, Huang J, Song D, Ni J, Zhou X, Zhu J, Ding M. Strontium-doped gelatin scaffolds promote M2 macrophage switch and angiogenesis through modulating the polarization of neutrophils. Biomater Sci 2021; 9:2931-2946. [PMID: 33621297 DOI: 10.1039/d0bm02126a] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The immune system mediates inflammation, vascularization and the first response to injuries or implanted biomaterials. Although the function of neutrophils in tissue repair has been extensively studied, its complete role in the tissue regeneration of biomaterials, specifically the resolution of inflammation and promotion of angiogenesis, is unclear. Here, we fabricate nanofibrous gelatin scaffolds containing 10% (w/w) strontium-hydroxyapatite (SrHA) via phase-separation methods to investigate Sr-mediated regulation of neutrophil polarization and, subsequently, the effects on angiogenesis and macrophage polarization. Compared with neutrophils cultured on pure gelatin or HA-incorporated gelatin scaffolds, neutrophils on SrHA-incorporated gelatin scaffolds show more N2 polarization in vitro and in vivo and significantly greater production of immunomodulatory and angiogenic factors. The Sr-induced immunomodulatory and proangiogenic functions of neutrophils are mediated through NF-κB pathway downregulation and increased STAT3 phosphorylation. Thus, neutrophils play a vital role in tissue engineering, and Sr-incorporated scaffolds efficiently promote neutrophil polarization to the N2 phenotype, enhancing resolution of inflammation and ultimately promoting angiogenesis and tissue regeneration. Thus, incorporation of neutrophils in analyses of the immune characteristics of scaffolds and the development of immunomodulatory biomaterials that can regulate neutrophils are novel and promising strategies in tissue engineering.
Collapse
Affiliation(s)
- Tao Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China. and Department of Orthopaedics, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China.
| | - Hongtao He
- The Third Ward of Department of Orthopedics, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Shahekou District, Dalian, Liaoning Province 116000, P. R. China
| | - Zezheng Yang
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Minhang District, Shanghai 200240, P. R. China
| | - Junjie Wang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China.
| | - Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Huangpu District, Shanghai 200011, China
| | - Guangxu He
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China.
| | - Jun Huang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China.
| | - Deye Song
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China.
| | - Jiangdong Ni
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China.
| | - Xiaojun Zhou
- College of Chemistry, Chemical Engineering and Biotechnology; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai 201620, P. R. China.
| | - Junfeng Zhu
- Department of Orthopaedics, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China.
| | - Muliang Ding
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China.
| |
Collapse
|
31
|
Sadowska JM, Ginebra MP. Inflammation and biomaterials: role of the immune response in bone regeneration by inorganic scaffolds. J Mater Chem B 2021; 8:9404-9427. [PMID: 32970087 DOI: 10.1039/d0tb01379j] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The regulatory role of the immune system in maintaining bone homeostasis and restoring its functionality, when disturbed due to trauma or injury, has become evident in recent years. The polarization of macrophages, one of the main constituents of the immune system, into the pro-inflammatory or anti-inflammatory phenotype has great repercussions for cellular crosstalk and the subsequent processes needed for proper bone regeneration such as angiogenesis and osteogenesis. In certain scenarios, the damaged osseous tissue requires the placement of synthetic bone grafts to facilitate the healing process. Inorganic biomaterials such as bioceramics or bioactive glasses are the most widely used due to their resemblance to the mineral phase of bone and superior osteogenic properties. The immune response of the host to the inorganic biomaterial, which is of an exogenous nature, might determine its fate, leading either to active bone regeneration or its failure. Therefore, various strategies have been employed, like the modification of structural/chemical features or the incorporation of bioactive molecules, to tune the interplay with the immune cells. Understanding how these particular modifications impact the polarization of macrophages and further osteogenic and osteoclastogenic events is of great interest in view of designing a new generation of osteoimmunomodulatory materials that support the regeneration of osseous tissue during all stages of bone healing.
Collapse
Affiliation(s)
- Joanna M Sadowska
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Ireland
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya, Av. Eduard Maristany 16, 08019 Barcelona, Spain. and Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| |
Collapse
|
32
|
Huang J, Li R, Yang J, Cai M, Lee Y, Wang A, Cheng B, Wang Y. Bioadaptation of implants to In vitro and In vivo oxidative stress pathological conditions via nanotopography-induced FoxO1 signaling pathways to enhance Osteoimmunal regeneration. Bioact Mater 2021; 6:3164-3176. [PMID: 33778196 PMCID: PMC7970012 DOI: 10.1016/j.bioactmat.2021.02.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Varieties of pathological conditions, including diabetes, are closely related to oxidative stress (OS), but the osseointegration or bioadaptation of implants to OS and the related mechanism remain poorly explored. In this study, the antioxidation and osteoimmune regeneration of titanium implants with micro/nanotopographies were evaluated under H2O2-, lipopolysaccharide (LPS)- and hyperglycemia-mediated cellular OS models and in diabetic rats as a representative animal model of OS. TiO2 nanotube (TNT) coating on titanium implants directly induced superior osteogenic differentiation of bone mesenchymal stem cells (MSCs) and osseointegration compared with microscale sand blasted-acid etched topography (SLA) under OS, attributed to higher superoxide dismutase 2 activity, the neutralization of intracellular reactive oxygen species (ROS), and less apoptosis. Mechanistically, the oxidation resistance on TNT is driven by upregulated forkhead box transcription factor O1 (FoxO1), which is abolished after knockdown of FoxO1 via shRNA in MSCs. Indirectly, TNT also alleviates OS in macrophages, therefore inducing a higher portion of the M2 phenotype under OS with increased secretion of the anti-inflammatory cytokine IL-10, further promoting the osseoimmunity capacity compared with SLA. The current study not only suggests the potential application of TiO2 nanotube-coated titanium implants in compromised conditions but also provides a systematic evaluation strategy for the future development of bone biomaterials. H2O2, lipopolysaccharide and hyperglycemia induced cellular oxidative stress models. TiO2 nanotubes promote oxidation resistance and osteogenesis under oxidative stress. TiO2 nanotubes activate forkhead box transcription factor O1 to enhance osteogenesis. TiO2-nanotube-coated implants promote osseointegration in diabetic rats. TiO2 nanotubes induce anti-inflammatory osteoimmunity under oxidative stress.
Collapse
Affiliation(s)
- Jingyan Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, China
| | - Ruoqi Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, China
| | - Jinghong Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, China
| | - Min Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, China
| | - Yichen Lee
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, China
| | - Anxun Wang
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, China
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, China
| |
Collapse
|
33
|
Chen L, Wang D, Qiu J, Zhang X, Liu X, Qiao Y, Liu X. Synergistic effects of immunoregulation and osteoinduction of ds-block elements on titanium surface. Bioact Mater 2021; 6:191-207. [PMID: 32913928 PMCID: PMC7452063 DOI: 10.1016/j.bioactmat.2020.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/01/2020] [Accepted: 08/01/2020] [Indexed: 12/18/2022] Open
Abstract
Ds-block elements have been gaining increasing attention in the field of biomaterials modification, owing to their excellent biological properties, such as antibiosis, osteogenesis, etc. However, their function mechanisms are not well understood and conflicting conclusions were drawn by previous studies on this issue, which are mainly resulted from the inconsistent experimental conditions. In this work, three most widely used ds-block elements, copper, zinc, and silver were introduced on titanium substrate by plasma immersion ion implantation method to investigate the rule of ds-block elements in the immune responses. Results showed that the implanted samples could decrease the inflammatory responses compared with Ti sample. The trend of anti-inflammatory effects of macrophages on samples was in correlation with cellular ROS levels, which was induced by the implanted biomaterials and positively correlated with the number of valence electrons of ds-block elements. The co-culture experiments of macrophages and bone marrow mesenchymal stem cells showed that these two kinds of cells could enhance the anti-inflammation and osteogenesis of samples by the paracrine manner of PGE2. In general, in their steady states on titanium substrate (Cu2+, Zn2+, Ag), the ds-block elements with more valence electrons exhibit better anti-inflammatory and osteogenic effects. Moreover, molecular biology experiments indicate that the PGE2-related signaling pathway may contribute to the desired immunoregulation and osteoinduction capability of ds-block elements. These findings suggest a correlation between the number of valence electrons of ds-block elements and the relevant biological responses, which provides new insight into the selection of implanted ions and surface design of biomaterials.
Collapse
Affiliation(s)
- Lan Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Donghui Wang
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Jiajun Qiu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Xianming Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Xingdan Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqin Qiao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| |
Collapse
|
34
|
van Hengel I, Gelderman F, Athanasiadis S, Minneboo M, Weinans H, Fluit A, van der Eerden B, Fratila-Apachitei L, Apachitei I, Zadpoor A. Functionality-packed additively manufactured porous titanium implants. Mater Today Bio 2020; 7:100060. [PMID: 32577614 PMCID: PMC7305382 DOI: 10.1016/j.mtbio.2020.100060] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/30/2022] Open
Abstract
The holy grail of orthopedic implant design is to ward off both aseptic and septic loosening for long enough that the implant outlives the patient. Questing this holy grail is feasible only if orthopedic biomaterials possess a long list of functionalities that enable them to discharge the onerous task of permanently replacing the native bone tissue. Here, we present a rationally designed and additive manufacturing (AM) topologically ordered porous metallic biomaterial that is made from Ti-6Al-4V using selective laser melting and packs most (if not all) of the required functionalities into a single implant. In addition to presenting a fully interconnected porous structure and form-freedom that enables realization of patient-specific implants, the biomaterials developed here were biofunctionalized using plasma electrolytic oxidation to locally release both osteogenic (i.e. strontium) and antibacterial (i.e. silver ions) agents. The same single-step biofunctionalization process also incorporated hydroxyapatite into the surface of the implants. Our measurements verified the continued release of both types of active agents up to 28 days. Assessment of the antibacterial activity in vitro and in an ex vivo murine model demonstrated extraordinarily high levels of bactericidal effects against a highly virulent and multidrug-resistant Staphylococcus aureus strain (i.e. USA300) with total eradication of both planktonic and adherent bacteria. This strong antibacterial behavior was combined with a significantly enhanced osteogenic behavior, as evidenced by significantly higher levels of alkaline phosphatase (ALP) activity compared with non-biofunctionalized implants. Finally, we discovered synergistic antibacterial behavior between strontium and silver ions, meaning that 4-32 folds lower concentrations of silver ions were required to achieve growth inhibition and total killing of bacteria. The functionality-packed biomaterial presented here demonstrates a unique combination of functionalities that make it an advanced prototype of future orthopedic biomaterials where implants will outlive patients.
Collapse
Affiliation(s)
- I.A.J. van Hengel
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| | - F.S.A. Gelderman
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| | - S. Athanasiadis
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| | - M. Minneboo
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| | - H. Weinans
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - A.C. Fluit
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - L.E. Fratila-Apachitei
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| | - I. Apachitei
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| | - A.A. Zadpoor
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, the Netherlands
| |
Collapse
|
35
|
Ion R, Necula MG, Mazare A, Mitran V, Neacsu P, Schmuki P, Cimpean A. Drug Delivery Systems Based on Titania Nanotubes and Active Agents for Enhanced Osseointegration of Bone Implants. Curr Med Chem 2020; 27:854-902. [PMID: 31362646 DOI: 10.2174/0929867326666190726123229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 01/16/2019] [Accepted: 05/04/2019] [Indexed: 12/31/2022]
Abstract
TiO2 nanotubes (TNTs) are attractive nanostructures for localized drug delivery. Owing to their excellent biocompatibility and physicochemical properties, numerous functionalizations of TNTs have been attempted for their use as therapeutic agent delivery platforms. In this review, we discuss the current advances in the applications of TNT-based delivery systems with an emphasis on the various functionalizations of TNTs for enhancing osteogenesis at the bone-implant interface and for preventing implant-related infection. Innovation of therapies for enhancing osteogenesis still represents a critical challenge in regeneration of bone defects. The overall concept focuses on the use of osteoconductive materials in combination with the use of osteoinductive or osteopromotive factors. In this context, we highlight the strategies for improving the functionality of TNTs, using five classes of bioactive agents: growth factors (GFs), statins, plant derived molecules, inorganic therapeutic ions/nanoparticles (NPs) and antimicrobial compounds.
Collapse
Affiliation(s)
- Raluca Ion
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Anca Mazare
- University of Erlangen-Nuremberg, Department of Materials Science, Erlangen, Germany
| | - Valentina Mitran
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Patricia Neacsu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Patrik Schmuki
- University of Erlangen-Nuremberg, Department of Materials Science, Erlangen, Germany
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
36
|
Figueiredo RDA, Ortega AC, González Maldonado LA, Castro RDD, Ávila-Campos MJ, Rossa C, Aquino SGD. Perillyl alcohol has antibacterial effects and reduces ROS production in macrophages. J Appl Oral Sci 2020; 28:e20190519. [PMID: 32348444 PMCID: PMC7185983 DOI: 10.1590/1678-7757-2019-0519] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/12/2019] [Indexed: 11/22/2022] Open
Abstract
Natural products have emerged as a rich source of bioactive compounds for adjunctive treatments of many infectious and inflammatory conditions, including periodontitis. Among the monoterpenes with significant biological properties, there is the perillyl alcohol (POH), which can be found in several essential oils and has shown immunomodulatory properties in recent studies, which may be interesting in the treatment of non-neoplastic inflammatory disorders.
Collapse
Affiliation(s)
| | - Adriana Cabrera Ortega
- Departamento de Diagnóstico e Cirurgia, Faculdade de Odontologia de Araraquara, Universidade Estadual Paulista, Araraquara, São Paulo, Brasil
| | - Laura Andrea González Maldonado
- Departamento de Diagnóstico e Cirurgia, Faculdade de Odontologia de Araraquara, Universidade Estadual Paulista, Araraquara, São Paulo, Brasil
| | - Ricardo Dias de Castro
- Programa de Pós-Graduação em Odontologia, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brasil
| | - Mario Julio Ávila-Campos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade de São Paulo, São Paulo, Brasil
| | - Carlos Rossa
- Departamento de Diagnóstico e Cirurgia, Faculdade de Odontologia de Araraquara, Universidade Estadual Paulista, Araraquara, São Paulo, Brasil
| | - Sabrina Garcia de Aquino
- Programa de Pós-Graduação em Odontologia, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brasil
| |
Collapse
|
37
|
Shimizu Y, Fujibayashi S, Yamaguchi S, Mori S, Kitagaki H, Shimizu T, Okuzu Y, Masamoto K, Goto K, Otsuki B, Kawai T, Morizane K, Kawata T, Matsuda S. Bioactive effects of strontium loading on micro/nano surface Ti6Al4V components fabricated by selective laser melting. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 109:110519. [PMID: 32228917 DOI: 10.1016/j.msec.2019.110519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022]
Abstract
Selective laser melting (SLM) titanium alloys require surface modification to achieve early bone-bonding. This study investigated the effects of solution and heat treatment to induce the sustained release of strontium (Sr) ions from SLM Ti6Al4V implants (Sr-S64). The results were compared with a control group comprising an untreated surface [SLM pure titanium (STi) and SLM Ti6Al4V (S64)] and a treated surface to induce the release of calcium (Ca) ions from SLM Ti6Al4V (Ca-S64). The surface-treated materials showed homogenous nanoscale network formation on the original micro-topographical surface and formed bone-like apatite on the surface in a simulated body fluid within 3 days. In vitro evaluation using MC3T3-E1 cells showed that the cells were viable on Sr-S64 surface, and Sr-S64 enhanced cell adhesion-related and osteogenic differentiation-related genes expression. In vivo rabbit tibia model, Sr-S64 provided significantly greater bone-bonding strength and bone-implant contact area than those in controls (STi and S64) in the early phase (2-4 weeks) after implantation; however, there was no statistical difference between Ca-S64 and controls. In conclusion, Sr solution and heat treatment was a safe and effective method to enhance early bone-bonding ability of S-64 by improving the surface characteristics and sustained delivery for Sr.
Collapse
Affiliation(s)
- Yu Shimizu
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Shunsuke Fujibayashi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Seiji Yamaguchi
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Shigeo Mori
- Osaka Yakin Kogyo Co., Ltd., 4-4-28 Zuiko, Higashiyodogawa-ku, Osaka 533-0005, Japan
| | - Hisashi Kitagaki
- Osaka Yakin Kogyo Co., Ltd., 4-4-28 Zuiko, Higashiyodogawa-ku, Osaka 533-0005, Japan
| | - Takayoshi Shimizu
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yaichiro Okuzu
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazutaka Masamoto
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Koji Goto
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Bungo Otsuki
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshiyuki Kawai
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazuaki Morizane
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomotoshi Kawata
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
38
|
Zhu L, Luo D, Liu Y. Effect of the nano/microscale structure of biomaterial scaffolds on bone regeneration. Int J Oral Sci 2020; 12:6. [PMID: 32024822 PMCID: PMC7002518 DOI: 10.1038/s41368-020-0073-y] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/15/2019] [Accepted: 12/18/2019] [Indexed: 01/12/2023] Open
Abstract
Natural bone is a mineralized biological material, which serves a supportive and protective framework for the body, stores minerals for metabolism, and produces blood cells nourishing the body. Normally, bone has an innate capacity to heal from damage. However, massive bone defects due to traumatic injury, tumor resection, or congenital diseases pose a great challenge to reconstructive surgery. Scaffold-based tissue engineering (TE) is a promising strategy for bone regenerative medicine, because biomaterial scaffolds show advanced mechanical properties and a good degradation profile, as well as the feasibility of controlled release of growth and differentiation factors or immobilizing them on the material surface. Additionally, the defined structure of biomaterial scaffolds, as a kind of mechanical cue, can influence cell behaviors, modulate local microenvironment and control key features at the molecular and cellular levels. Recently, nano/micro-assisted regenerative medicine becomes a promising application of TE for the reconstruction of bone defects. For this reason, it is necessary for us to have in-depth knowledge of the development of novel nano/micro-based biomaterial scaffolds. Thus, we herein review the hierarchical structure of bone, and the potential application of nano/micro technologies to guide the design of novel biomaterial structures for bone repair and regeneration.
Collapse
Affiliation(s)
- Lisha Zhu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Dan Luo
- State Key Laboratory of Heavy Oil Processing, College of New Energy and Materials, Beijing Key Laboratory of Biogas Upgrading Utilization, China University of Petroleum (Beijing), Beijing, China
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| |
Collapse
|
39
|
Liu G, Wang X, Zhou X, Zhang L, Mi J, Shan Z, Huang B, Chen Z, Chen Z. Modulating the cobalt dose range to manipulate multisystem cooperation in bone environment: a strategy to resolve the controversies about cobalt use for orthopedic applications. Theranostics 2020; 10:1074-1089. [PMID: 31938052 PMCID: PMC6956813 DOI: 10.7150/thno.37931] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
The paradoxical effect of cobalt on biological processes has aroused controversy regarding the application of cobalt-based biomaterials in bone regeneration. Tuning the dose range of cobalt ions may be a valid strategy to resolve the controversies about cobalt use for orthopedic applications. Recent progress in bone biology has highlighted the effects of multisystem cooperation (especially of osteoimmune, skeletal, and vascular systems) on bone dynamics. Before the application of this dose-tuning strategy, a deeper understanding of its dose-dependent effect on the cooperation of osteoimmune, skeletal, and vascular systems is needed. However, due to the difficulties with investigating the interaction of multiple systems in vitro, the multimodal effects of cobalt on bone homeostasis were investigated here, in an in vivo scenario. Methods: In vitro CCK8 assay and cytoskeletal staining were preformed to detecte the cell cytotoxic reaction in response to 0.1-100 ppm cobalt stimulation. Blood clot containing 0.1 to 5 ppm of cobalt were implanted in the rat calvarium defect. The gene profile of osteoimmune, skeletal, and vascular system as well as the systemic toxicity were evaluated via RT-qPCR, histological analysis and inductively coupled plasma mass spectrometry. The bone regeneration, osteoclastogenesis and vascularization were assessed by micro-ct and histological analysis. Results: Cobalt concentration below 5 ppm did not cause cell toxicity in vitro. No systemic toxicity was observed in vivo at 0.1-5 ppm cobalt concentration. It was found that the early cytokine profiles of the multiple interacting systems were different in response to different cobalt doses. Most of the anti-inflammatory, osteogenic, and proangiogenic factors were upregulated in the 1 ppm cobalt group at the early stage. In the late stage, the 1ppm group was most superior in bone regenerative effect while the 5 ppm group displayed the strongest osteoclastogenesis activity. Conclusions: The 1 ppm concentration of cobalt yielded the most favorable cooperation of the osteoimmune, skeletal, and vascular systems and subsequently optimal bone regeneration outcomes. Tuning the cobalt dose range to manipulate the cooperation of osteoimmune, skeletal, and vascular systems could be a promising and valuable strategy to prevent paradoxical effects of cobalt while preserving its beneficial effects.
Collapse
Affiliation(s)
- Guanqi Liu
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xiaoshuang Wang
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xuan Zhou
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Linjun Zhang
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Jiaomei Mi
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zhengjie Shan
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Baoxin Huang
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zhuofan Chen
- Zhujiang New Town Clinic, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zetao Chen
- Institute of Stomatology and Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
40
|
Wang Q, Tang Y, Ke Q, Yin W, Zhang C, Guo Y, Guan J. Magnetic lanthanum-doped hydroxyapatite/chitosan scaffolds with endogenous stem cell-recruiting and immunomodulatory properties for bone regeneration. J Mater Chem B 2020; 8:5280-5292. [PMID: 32441294 DOI: 10.1039/d0tb00342e] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Magnetic lanthanum hydroxyapatite/chitosan scaffolds can better repair bone defects through stem cell recruitment and immunomodulation.
Collapse
Affiliation(s)
- Qiyang Wang
- Department of Orthopedic Surgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai Jiao Tong University
- Shanghai 200233
- China
| | - Yaqi Tang
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials
- Shanghai Normal University
- Shanghai 200234
- China
| | - Qinfei Ke
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials
- Shanghai Normal University
- Shanghai 200234
- China
- School of Materials Science and Engineering
| | - Wenjing Yin
- Department of Orthopedic Surgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai Jiao Tong University
- Shanghai 200233
- China
| | - Changqing Zhang
- Department of Orthopedic Surgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai Jiao Tong University
- Shanghai 200233
- China
| | - Yaping Guo
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials
- Shanghai Normal University
- Shanghai 200234
- China
| | - Junjie Guan
- Department of Orthopedic Surgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai Jiao Tong University
- Shanghai 200233
- China
| |
Collapse
|
41
|
Zhou W, Wang T, Gan Y, Yang J, Zhu H, Wang A, Wang Y, Xi W. Effect of micropore/microsphere topography and a silicon-incorporating modified titanium plate surface on the adhesion and osteogenic differentiation of BMSCs. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 48:230-241. [PMID: 31851839 DOI: 10.1080/21691401.2019.1699829] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Wuchao Zhou
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine of Jiangxi Province, Medical College of Nanchang University, Nanchang, China
| | - Tiesheng Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine of Jiangxi Province, Medical College of Nanchang University, Nanchang, China
- Department of Oral and Maxillofacial Surgery, Pingxiang People's Hospital, Pingxiang, China
| | - Yanzi Gan
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine of Jiangxi Province, Medical College of Nanchang University, Nanchang, China
| | - Jian Yang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine of Jiangxi Province, Medical College of Nanchang University, Nanchang, China
| | - Hongshui Zhu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine of Jiangxi Province, Medical College of Nanchang University, Nanchang, China
| | - Anxun Wang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yujiang Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine of Jiangxi Province, Medical College of Nanchang University, Nanchang, China
| | - Weihong Xi
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine of Jiangxi Province, Medical College of Nanchang University, Nanchang, China
| |
Collapse
|
42
|
Yang C, Zhu K, Yuan X, Zhang X, Qian Y, Cheng T. Curcumin has immunomodulatory effects on RANKL-stimulated osteoclastogenesis in vitro and titanium nanoparticle-induced bone loss in vivo. J Cell Mol Med 2019; 24:1553-1567. [PMID: 31845532 PMCID: PMC6991655 DOI: 10.1111/jcmm.14842] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/08/2019] [Accepted: 10/27/2019] [Indexed: 12/25/2022] Open
Abstract
Wear particle‐stimulated inflammatory bone destruction and the consequent aseptic loosening remain the primary causes of artificial prosthesis failure and revision. Previous studies have demonstrated that curcumin has a protective effect on bone disorders and inflammatory diseases and can ameliorate polymethylmethacrylate‐induced osteolysis in vivo. However, the effect on immunomodulation and the definitive mechanism by which curcumin reduces the receptor activators of nuclear factor‐kappa B ligand (RANKL)‐stimulated osteoclast formation and prevents the activation of osteoclastic signalling pathways are unclear. In this work, the immunomodulation effect and anti‐osteoclastogenesis capacities exerted by curcumin on titanium nanoparticle‐stimulated macrophage polarization and on RANKL‐mediated osteoclast activation and differentiation in osteoclastic precursor cells in vitro were investigated. As expected, curcumin inhibited RANKL‐stimulated osteoclast maturation and formation and had an immunomodulatory effect on macrophage polarization in vitro. Furthermore, studies aimed to identify the potential molecular and cellular mechanisms revealed that this protective effect of curcumin on osteoclastogenesis occurred through the amelioration of the activation of Akt/NF‐κB/NFATc1 pathways. Additionally, an in vivo mouse calvarial bone destruction model further confirmed that curcumin ameliorated the severity of titanium nanoparticle‐stimulated bone loss and destruction. Our results conclusively indicated that curcumin, a major biologic component of Curcuma longa with anti‐inflammatory and immunomodulatory properties, may serve as a potential therapeutic agent for osteoclastic diseases.
Collapse
Affiliation(s)
- Chao Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Kechao Zhu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiangwei Yuan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yebin Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tao Cheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
43
|
Yang C, Wang W, Zhu K, Liu W, Luo Y, Yuan X, Wang J, Cheng T, Zhang X. Lithium chloride with immunomodulatory function for regulating titanium nanoparticle-stimulated inflammatory response and accelerating osteogenesis through suppression of MAPK signaling pathway. Int J Nanomedicine 2019; 14:7475-7488. [PMID: 31571859 PMCID: PMC6750619 DOI: 10.2147/ijn.s210834] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/30/2019] [Indexed: 12/26/2022] Open
Abstract
Background Wear particle-induced inflammatory osteolysis and the consequent aseptic loosening constitute the leading reasons for prosthesis failure and revision surgery. Several studies have demonstrated that the macrophage polarization state and immune response play critical roles in periprosthetic osteolysis and tissue repair, but the immunomodulatory role of lithium chloride (LiCl), which has a protective effect on wear particle-induced osteolysis by suppressing osteoclasts and attenuating inflammatory responses, has never been investigated. Methods In this work, the immunomodulatory capability of LiCl on titanium (Ti) nanoparticle-stimulated transformation of macrophage phenotypes and the subsequent effect on osteogenic differentiation were investigated. We first speculated that LiCl attenuated Ti nanoparticle-stimulated inflammation responses by driving macrophage polarization and generating an immune micro-environment to improve osteogenesis. Furthermore, a metal nanoparticle-stimulated murine air pouch inflammatory model was applied to confirm this protective effect in vivo. Results The results revealed that metal nanoparticles significantly activate M1 phenotype (proinflammatory macrophage) expression and increase proinflammatory cytokines secretions in vitro and in vivo, whereas LiCl drives macrophages to the M2 phenotype (anti-inflammatory macrophage) and increases the release of anti-inflammatory and bone-related cytokines. This improved the osteogenic differentiation capability of rat bone marrow mesenchymal stem cells (rBMSCs). In addition, we also provided evidence that LiCl inhibits the phosphorylation of the p38 mitogen-activated protein kinase (p38) and extracellular signal-regulated kinase (ERK) pathways in wear particle-treated macrophages. Conclusion LiCl has the immunomodulatory effects to alleviate Ti nanoparticle-mediated inflammatory reactions and enhance the osteogenic differentiation of rBMSCs by driving macrophage polarization. Thus, LiCl may be an effective therapeutic alternative for preventing and treating wear debris-induced inflammatory osteolysis.
Collapse
Affiliation(s)
- Chao Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Wei Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Kechao Zhu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Wei Liu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Yao Luo
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Xiangwei Yuan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Jiaxing Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Tao Cheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| |
Collapse
|
44
|
Unagolla JM, Jayasuriya AC. Enhanced cell functions on graphene oxide incorporated 3D printed polycaprolactone scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:1-11. [PMID: 31146979 PMCID: PMC6546300 DOI: 10.1016/j.msec.2019.04.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 03/28/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023]
Abstract
For tissue engineering applications, a porous scaffold with an interconnected network is essential to facilitate the cell attachment and proliferation in a three dimensional (3D) structure. This study aimed to fabricate the scaffolds by an extrusion-based 3D printer using a blend of polycaprolactone (PCL), and graphene oxide (GO) as a favorable platform for bone tissue engineering. The mechanical properties, morphology, biocompatibility, and biological activities such as cell proliferation and differentiation were studied concerning the two different pore sizes; 400 μm, and 800 μm, and also with two different GO content; 0.1% (w/w) and 0.5% (w/w). The compressive strength of the scaffolds was not significantly changed due to the small amount of GO, but, as expected scaffolds with 400 μm pores showed a higher compressive modulus in comparison to the scaffolds with 800 μm pores. The data indicated that the cell attachment and proliferation were increased by adding a small amount of GO. According to the results, pore size did not play a significant role in cell proliferation and differentiation. Alkaline Phosphate (ALP) activity assay further confirmed that the GO increase the ALP activity and further Elemental analysis of Calcium and Phosphorous showed that the GO increased the mineralization compared to PCL only scaffolds. Western blot analysis showed the porous structure facilitate the secretion of bone morphogenic protein-2 (BMP-2) and osteopontin at both day 7 and 14 which galvanizes the osteogenic capability of PCL and PCL + GO scaffolds.
Collapse
Affiliation(s)
- Janitha M Unagolla
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43607, USA
| | - Ambalangodage C Jayasuriya
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43607, USA; Department of Orthopedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
45
|
Crocin inhibits titanium particle-induced inflammation and promotes osteogenesis by regulating macrophage polarization. Int Immunopharmacol 2019; 76:105865. [PMID: 31476694 DOI: 10.1016/j.intimp.2019.105865] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/16/2019] [Accepted: 08/23/2019] [Indexed: 12/24/2022]
Abstract
Wear particle-induced periprosthetic inflammatory osteolysis and resultant aseptic loosening are major causes of orthopedic implant failure, for which there are no effective treatments other than revision surgery. Crocin, a carotenoid compound derived from crocus flowers, has anti-inflammatory properties, but its immunomodulatory function and role in particle-induced osteolysis are not well characterized. Here we report the effect of crocin on titanium (Ti) particle-induced macrophage polarization and osteogenic differentiation. We found that crocin induced anti-inflammatory (M2) macrophage polarization and attenuated Ti particle-induced inflammation by promoting the expression of anti-inflammatory cytokines in vitro as well as in vivo in a mouse air-pouch model. Additionally, crocin pre-treated macrophages promoted osteogenic differentiation of co-cultured mouse bone mesenchymal stem cells (BMSCs). These effects were mediated via inhibition of p38 and c-Jun N-terminal kinase signaling. Our results indicate that crocin suppresses Ti particle-induced inflammation and enhances osteogenic differentiation of BMSCs by inducing M2 macrophage polarization, highlighting its therapeutic potential for preventing wear particle-induced osteolysis.
Collapse
|
46
|
Rajendran A, Kapoor U, Jothinarayanan N, Lenka N, Pattanayak DK. Effect of Silver-Containing Titania Layers for Bioactivity, Antibacterial Activity, and Osteogenic Differentiation of Human Mesenchymal Stem Cells on Ti Metal. ACS APPLIED BIO MATERIALS 2019; 2:3808-3819. [DOI: 10.1021/acsabm.9b00420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Archana Rajendran
- CSIR-Central Electrochemical Research Institute, Karaikudi, Tamil Nadu 630006, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Upasana Kapoor
- National Centre for Cell Science, Pune, Maharashtra 411007, India
| | | | - Nibedita Lenka
- National Centre for Cell Science, Pune, Maharashtra 411007, India
| | - Deepak K. Pattanayak
- CSIR-Central Electrochemical Research Institute, Karaikudi, Tamil Nadu 630006, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
47
|
Macrophage polarization following three-dimensional porous PEEK. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109948. [PMID: 31499957 DOI: 10.1016/j.msec.2019.109948] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 03/30/2019] [Accepted: 07/04/2019] [Indexed: 01/18/2023]
Abstract
Macrophages play an important role in foreign body reaction (FBR), and exhibit a detrimental or beneficial function in tissue repair while polarized into different phenotypes. The objective of this work is to evaluate the effect of three-dimensional (3D) porous polyetheretherketone (PEEK) on macrophage polarization through sulfonating PEEK and establishing a mouse air pouch model. The in vivo results show that the sulfonated PEEK induced higher levels of anti-inflammatory cytokine together with lower levels of pro-inflammatory cytokine. In addition, it was found that a relatively mild infiltration of inflammatory cells was caused and there were more M2 macrophages and less M1 ones when compared with PEEK. It indicates that 3D porous PEEK induces a shift to M2 macrophages and has large potential in regenerative medicine application.
Collapse
|
48
|
Yin X, Li Y, Yang C, Weng J, Wang J, Zhou J, Feng B. Alginate/chitosan multilayer films coated on IL-4-loaded TiO2 nanotubes for modulation of macrophage phenotype. Int J Biol Macromol 2019; 133:503-513. [DOI: 10.1016/j.ijbiomac.2019.04.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 01/21/2023]
|
49
|
Alginate/chitosan multilayer films coated on IL-4-loaded TiO2 nanotubes for modulation of macrophage phenotype. Int J Biol Macromol 2019; 132:495-505. [DOI: 10.1016/j.ijbiomac.2019.03.184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/08/2019] [Accepted: 03/25/2019] [Indexed: 11/24/2022]
|
50
|
Chen L, Wang D, Peng F, Qiu J, Ouyang L, Qiao Y, Liu X. Nanostructural Surfaces with Different Elastic Moduli Regulate the Immune Response by Stretching Macrophages. NANO LETTERS 2019; 19:3480-3489. [PMID: 31091110 DOI: 10.1021/acs.nanolett.9b00237] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A proper immune response is key for the successful implantation of biomaterials, and designing and fabricating biomaterials to regulate immune responses is the future trend. In this work, three different nanostructures were constructed on the surface of titanium using a hydrothermal method, and through a series of in vitro and in vivo experiments, we found that the aspect ratio of nanostructures can affect the elastic modulus of a material surface and further regulate immune cell behaviors. This work demonstrates that nanostructures with a higher aspect ratio can endow a material surface with a lower elastic modulus, which was confirmed by experiments and theoretical analyses. The deflection of nanostructures under the cell adsorption force is a substantial factor in stretching macrophages to enhance cell adhesion and spreading, further inducing macrophage polarization toward the M1 phenotype and leading to intense immune responses. In contrast, a nanostructure with a lower aspect ratio on a material surface leads to a higher surface elastic modulus, making deflection of the material difficult and creating a surface that is not conducive to macrophage adhesion and spreading, thus reducing the immune response. Moreover, molecular biology experiments indicated that regulation of the immune response by the elastic modulus is primarily related to the NF-κB signaling pathway. These findings suggest that the immune response can be regulated by constructing nanostructural surfaces with the proper elastic modulus through their influence on cell adhesion and spreading, which provides new insights into the surface design of biomaterials.
Collapse
Affiliation(s)
- Lan Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Donghui Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
| | - Feng Peng
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Jiajun Qiu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Liping Ouyang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yuqin Qiao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
- Center of Materials Science and Optoelectronics Engineering , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|