1
|
Hou X, Zhang L, Chen Y, Liu Z, Zhao X, Lu B, Luo Y, Qu X, Musskaya O, Glazov I, Kulak AI, Chen F, Zhao J, Zhou Z, Zheng L. Photothermal switch by gallic acid-calcium grafts synthesized by coordination chemistry for sequential treatment of bone tumor and regeneration. Biomaterials 2025; 312:122724. [PMID: 39106818 DOI: 10.1016/j.biomaterials.2024.122724] [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: 02/07/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/09/2024]
Abstract
The residual bone tumor and defects which is caused by surgical therapy of bone tumor is a major and important problem in clinicals. And the sequential treatment for irradiating residual tumor and repairing bone defects has wildly prospects. In this study, we developed a general modification strategy by gallic acid (GA)-assisted coordination chemistry to prepare black calcium-based materials, which combines the sequential photothermal therapy of bone tumor and bone defects. The GA modification endows the materials remarkable photothermal properties. Under the near-infrared (NIR) irradiation with different power densities, the black GA-modified bone matrix (GBM) did not merely display an excellent performance in eliminating bone tumor with high temperature, but showed a facile effect of the mild-heat stimulation to accelerate bone regeneration. GBM can efficiently regulate the microenvironments of bone regeneration in a spatial-temporal manner, including inflammation/immune response, vascularization and osteogenic differentiation. Meanwhile, the integrin/PI3K/Akt signaling pathway of bone marrow mesenchymal stem cells (BMSCs) was revealed to be involved in the effect of osteogenesis induced by the mild-heat stimulation. The outcome of this study not only provides a serial of new multifunctional biomaterials, but also demonstrates a general strategy for designing novel blacked calcium-based biomaterials with great potential for clinical use.
Collapse
Affiliation(s)
- Xiaodong Hou
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China; Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Lei Zhang
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Yixing Chen
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Zhiqing Liu
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xinyu Zhao
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Bingqiang Lu
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Yiping Luo
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xinyu Qu
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Olga Musskaya
- Institute of General and Inorganic Chemistry, National Academy of Sciences of Belarus, Surganova Str. 9, 220072, Minsk, Belarus
| | - Ilya Glazov
- Institute of General and Inorganic Chemistry, National Academy of Sciences of Belarus, Surganova Str. 9, 220072, Minsk, Belarus
| | - Anatoly I Kulak
- Institute of General and Inorganic Chemistry, National Academy of Sciences of Belarus, Surganova Str. 9, 220072, Minsk, Belarus
| | - Feng Chen
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Zifei Zhou
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Longpo Zheng
- Center for Orthopedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China; Shanghai Trauma Emergency Center, Orthopedic Intelligent Minimally Invasive Diagnosis & Treatment Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
2
|
Yang R, Yan L, Xu T, Zhang K, Lu X, Xie C, Fu W. Injectable bioadhesive hydrogel as a local nanomedicine depot for targeted regulation of inflammation and ferroptosis in rheumatoid arthritis. Biomaterials 2024; 311:122706. [PMID: 39032219 DOI: 10.1016/j.biomaterials.2024.122706] [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: 01/12/2024] [Revised: 05/23/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Medicine intervention is the major clinical treatment used to relieve the symptoms and delay the progression of rheumatoid arthritis (RA), but is limited by its poor targeted delivery and short therapeutic duration. Herein, we developed an injectable and bioadhesive gelatin-based (Gel) hydrogel as a local depot of leonurine (Leon)-loaded and folate-functionalized polydopamine (FA-PDA@Leon) nanoparticles for anti-inflammation and chondroprotection in RA. The nanoparticles could protect Leon and facilitate its entry into the M1 phenotype macrophage for intracellular delivery of Leon, while the hydrogel tightly adhered to the tissues in the joint cavity and prolonged the retention of FA-PDA@Leon nanoparticles, thus achieving higher availability and therapeutic efficiency of Leon. In vitro and in vivo experiments demonstrated that the Gel/FA-PDA@Leon hydrogel could strongly suppress the inflammatory response by down-regulating the JAK2/STAT3 signaling pathway in macrophages and protect the chondrocytes from ferritinophagy/ferroptosis. This contributed to maintaining the structural integrity of articular cartilage and accelerating the joint functional recovery. This work provides an effective and convenient strategy to achieve higher bioavailability and long-lasting therapeutic duration of medicine intervention in arthritis diseases.
Collapse
Affiliation(s)
- Runze Yang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China
| | - Liwei Yan
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China
| | - Tianhao Xu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China
| | - Kaibo Zhang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China
| | - Xiong Lu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Chaoming Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China.
| | - Weili Fu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China.
| |
Collapse
|
3
|
Orozco-Osorio YA, Gaita-Anturi AV, Ossa-Orozco CP, Arias-Acevedo M, Uribe D, Paucar C, Vasquez AF, Saldarriaga W, Ramirez JG, Lopera A, García C. Utilization of Additive Manufacturing Techniques for the Development of a Novel Scaffolds with Magnetic Properties for Potential Application in Enhanced Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402419. [PMID: 39004887 DOI: 10.1002/smll.202402419] [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: 03/26/2024] [Revised: 06/24/2024] [Indexed: 07/16/2024]
Abstract
This study focuses on designing and evaluating scaffolds with essential properties for bone regeneration, such as biocompatibility, macroporous geometry, mechanical strength, and magnetic responsiveness. The scaffolds are made using 3D printing with acrylic resin and iron oxides synthesized through solution combustion. Utilizing triply periodic minimal surfaces (TPMS) geometry and mask stereolithography (MSLA) printing, the scaffolds achieve precise geometrical features. The mechanical properties are enhanced through resin curing, and magnetite particles from synthesized nanoparticles and alluvial magnetite are added for magnetic properties. The scaffolds show a balance between stiffness, porosity, and magnetic responsiveness, with maximum compression strength between 4.8 and 9.2 MPa and Young's modulus between 58 and 174 MPa. Magnetic properties such as magnetic coercivity, remanence, and saturation are measured, with the best results from scaffolds containing synthetic iron oxides at 1% weight. The viscosity of the mixtures used for printing is between 350 and 380 mPas, and contact angles between 90° and 110° are achieved. Biocompatibility tests indicate the potential for clinical trials, though further research is needed to understand the impact of magnetic properties on cellular interactions and optimize scaffold design for specific applications. This integrated approach offers a promising avenue for the development of advanced materials capable of promoting enhanced bone regeneration.
Collapse
Affiliation(s)
| | | | | | - María Arias-Acevedo
- Instituto Tecnológico Metropolitano, Calle 73 #76A-354, Campus Robledo, Medellín, Antioquia, 50034, Colombia
| | - Diego Uribe
- Instituto Tecnológico Metropolitano, Calle 73 #76A-354, Campus Robledo, Medellín, Antioquia, 50034, Colombia
| | - Carlos Paucar
- Universidad Nacional de Colombia sede Medellín, Carrera 65 # 59A-100, Medellin, Antioquia, 050034, Colombia
| | | | - Wilmer Saldarriaga
- Universidad Nacional de Colombia sede Medellín, Carrera 65 # 59A-100, Medellin, Antioquia, 050034, Colombia
| | | | - Alex Lopera
- Grupo de Nanoestructuras y Física Aplicada (NANOUPAR), Universidad Nacional de Colombia, La Paz, 202017, Colombia
| | - Claudia García
- Universidad Nacional de Colombia sede Medellín, Carrera 65 # 59A-100, Medellin, Antioquia, 050034, Colombia
| |
Collapse
|
4
|
Jiang K, Luo C, Li YM, Wang K, Huang S, You XH, Liu Y, Luo E, Xu JZ, Zhang L, Li ZM. An immunomodulatory and osteogenic bacterial cellulose scaffold for bone regeneration via regulating the immune microenvironment. Int J Biol Macromol 2024; 281:136375. [PMID: 39383912 DOI: 10.1016/j.ijbiomac.2024.136375] [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: 05/11/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Creating a bone homeostasis microenvironment that balances osteogenesis and immunity is a substantial challenge for bone regeneration. Here, we prepared an immunomodulatory and osteogenic bacterial cellulose scaffold (FOBS) via a facile one-pot approach. The aldehyde groups were generated via selective oxidation of the hydroxyl groups of bacterial cellulose, offering the bonding sites for dopamine through a Schiff base reaction. At the same time, the deposition of Ca2+ and PO43- was promoted on the aldehyde cellulose scaffold because of the high affinity of the catechol moiety for Ca2+. Compared with that of the unmodified scaffold, the hydroxyapatite content of FOBS increased by 47.1 % according to the ICP results. Interestingly, FOBS regulated the immune microenvironment to accelerate the conversion of M1 to M2 macrophages. The expressions of ARG-1 and Dectin-1 (M2) in the FOBS group increased by >100 %. The expression of osteogenic differentiation of BMSCs was also upregulated. In a rat cranial defect model, the BV/TV of FOBS was significantly increased. Further immunohistochemical analysis revealed that an improved immune microenvironment promoted the osteogenic differentiation of stem cells in vivo. This work provides an effective and easy-to-operate strategy for the development of the bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Kai Jiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Chuan Luo
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yuan-Min Li
- Key Laboratory of Transplant Engineering and Immunology, NHC, Chengdu 610065, China; Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Kai Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Shishu Huang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Xuan-He You
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Li Zhang
- Department of Rehabilitation Medicine, West China Second Hospital, Sichuan University, Chengdu 610065, China.
| | - Zhong-Ming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
5
|
Vu BT, Tran TH, Ly KL, Trinh KPN, Nguyen MNH, Doan HN, Duong TT, Hua HTN, Le HT, Le TD, Dang NNT, Nguyen HT. Polycaprolactone Hybrid Scaffold Loaded With N,O-Carboxymethyl Chitosan/Aldehyde Hyaluronic Acid/Hydroxyapatite Hydrogel for Bone Regeneration. J Biomed Mater Res B Appl Biomater 2024; 112:e35486. [PMID: 39295151 DOI: 10.1002/jbm.b.35486] [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: 01/11/2024] [Revised: 05/16/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024]
Abstract
Hydrogels have emerged as potential materials for bone grafting, thanks to their biocompatibility, biodegradation, and flexibility in filling irregular bone defects. In this study, we fabricated a novel NAH hydrogel system, composed of N,O-carboxymethyl chitosan (NOCC), aldehyde hyaluronic acid (AHA), and hydroxyapatite (HAp). To improve the mechanical strength of the fabricated hydrogel, a porous polycaprolactone (PCL) matrix was synthesized and used as a three-dimensional (3D) support template for NAH hydrogel loading, forming a novel PCL/NAH hybrid scaffold. A mixture of monosodium glutamate (M) and sucrose (S) at varied weight ratios (5M:5S, 7M:3S, and 9M:1S) was used for the fabrication of 3D PCL matrices. The morphology, interconnectivity, and water resistance of the porous PCL scaffolds were investigated for optimal hydrogel loading efficiency. The results demonstrated that PCL scaffolds with porogen ratios of 7M:3S and 9M:1S possessed better interconnectivity than 5M:5S ratio. The compressive strength of the PCL/NAH hybrid scaffolds with 9M:1S (561.6 ± 6.1 kPa) and 7M:3S (623.8 ± 6.8 kPa) ratios are similar to cancellous bone and all hybrid scaffolds were biocompatible. Rabbit models with tibial defects were implanted with the PCL/NAH scaffolds to assess the wound healing capability. The results suggest that the PCL/NAH hybrid scaffolds, specifically those with porogen ratio of 7M:3S, exhibit promising bone healing effects.
Collapse
Affiliation(s)
- Binh Thanh Vu
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Thai Hoang Tran
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Khanh Loan Ly
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Khanh Phan-Ngoc Trinh
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - My Ngoc-Hoang Nguyen
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Hoan Ngoc Doan
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Thanh-Tu Duong
- Department of Pathology, University of Medicine and Pharmacy at Ho chi Minh City, Ho Chi Minh City, Vietnam
| | - Ha Thi-Ngoc Hua
- Department of Pathology, University of Medicine and Pharmacy at Ho chi Minh City, Ho Chi Minh City, Vietnam
| | - Hung Thanh Le
- Medi Mech Research and Development Company, Ho Chi Minh City, Vietnam
| | | | - Nhi Ngoc-Thao Dang
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Hiep Thi Nguyen
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| |
Collapse
|
6
|
Mao J, Sun J, Wang L, Liu X, Bi J. Flexible and high-strength bioactive glass fiber membrane for bone regeneration with the aid of alkoxysilane sol spinnability. Mater Today Bio 2024; 28:101224. [PMID: 39290465 PMCID: PMC11407074 DOI: 10.1016/j.mtbio.2024.101224] [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: 06/01/2024] [Revised: 08/25/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024] Open
Abstract
In this research, the spinnability of bioactive glass (BG) precursor solution was supplied by alkoxysilane sol with appropriate molar ratio of H2O/silicon (R) to prepare bioactive glass fiber membrane (BFM) using electrospinning (ES) technique. Alkoxysilane could form a linear or chain-like colloidal aggregation in hydrolysis-polycondensation with R = 2 or so, thereby exhibiting good spinnability. Therefore, the role of polymer binders could be largely replaced. Due to the significant decrease of polymer binder, the defects within the fibers are largely reduced and degree of fiber densification was improved after calcination, leading to BFM drastically enhanced strength and flexibility. The effect of R and calcination temperature on mechanical performance were investigated in detail. The tensile strength could reach the highest value 2.31 MPa with R = 2 and calcination at 700 °C. In addition, under this preparation condition, the BFM also possessed good flexibility with bending rigidity 37.7 mN. Furthermore, the great performance of promoting cell proliferation and osteogenesis could be observed from in vitro cellular experiment. The BFM calcined at 750 °C exhibited the best promoting osteogenic differentiation ability. The rat skull defect model revealed BFM could perform well in osteogenesis in vivo.
Collapse
Affiliation(s)
- Junjie Mao
- Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, PR China
| | - Junyuan Sun
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China
| | - Lu Wang
- Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, PR China
| | - Xinyu Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China
| | - Jianqiang Bi
- Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, PR China
| |
Collapse
|
7
|
Xue JD, Gao J, Tang AF, Feng C. Shaping the immune landscape: Multidimensional environmental stimuli refine macrophage polarization and foster revolutionary approaches in tissue regeneration. Heliyon 2024; 10:e37192. [PMID: 39296009 PMCID: PMC11408064 DOI: 10.1016/j.heliyon.2024.e37192] [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: 05/10/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
In immunology, the role of macrophages extends far beyond their traditional classification as mere phagocytes; they emerge as pivotal architects of the immune response, with their function being significantly influenced by multidimensional environmental stimuli. This review investigates the nuanced mechanisms by which diverse external signals ranging from chemical cues to physical stress orchestrate macrophage polarization, a process that is crucial for the modulation of immune responses. By transitioning between pro-inflammatory (M1) and anti-inflammatory (M2) states, macrophages exhibit remarkable plasticity, enabling them to adapt to and influence their surroundings effectively. The exploration of macrophage polarization provides a compelling narrative on how these cells can be manipulated to foster an immune environment conducive to tissue repair and regeneration. Highlighting cutting-edge research, this review presents innovative strategies that leverage the dynamic interplay between macrophages and their environment, proposing novel therapeutic avenues that harness the potential of macrophages in regenerative medicine. Moreover, this review critically evaluates the current challenges and future prospects of translating macrophage-centered strategies from the laboratory to clinical applications.
Collapse
Affiliation(s)
- Jing-Dong Xue
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Gao
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ai-Fang Tang
- Department of Geratology, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Chao Feng
- Department of Reproductive Medicine, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China
| |
Collapse
|
8
|
Chen Z, Xiao N, Luo L, Zhang L, Yin F, Hu W, Wu Z, Chen Y, Luo K, Xu X. Nanosilicates facilitate periodontal regeneration potential by activating the PI3K-AKT signaling pathway in periodontal ligament cells. J Nanobiotechnology 2024; 22:532. [PMID: 39223550 PMCID: PMC11370094 DOI: 10.1186/s12951-024-02798-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The recent development of nanobiomaterials has shed some light on the field of periodontal tissue regeneration. Laponite (LAP), an artificially synthesized two-dimensional (2D) disk-shaped nanosilicate, has garnered substantial attention in regenerative biomedical applications owing to its distinctive structure, exceptional biocompatibility and bioactivity. This study endeavors to comprehensively evaluate the influence of LAP on periodontal regeneration. The effects of LAP on periodontal ligament cells (PDLCs) on osteogenesis, cementogenesis and angiogenesis were systematically assessed, and the potential mechanism was explored through RNA sequencing. The results indicated that LAP improved osteogenic and cementogenic differentiation of PDLCs, the regulatory effects of LAP on PDLCs were closely correlated with activation of PI3K-AKT signaling pathway. Moreover, LAP enhanced angiogenesis indirectly via manipulating paracrine of PDLCs. Then, LAP was implanted into rat periodontal defect to confirm its regenerative potential. Both micro-CT and histological analysis indicated that LAP could facilitate periodontal tissue regeneration in vivo. These findings provide insights into the bioactivity and underlying mechanism of LAP on PDLCs, highlighting it might be a potential therapeutic option in periodontal therapy.
Collapse
Affiliation(s)
- Ziqin Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Nianqi Xiao
- Gannan Health Vocational College, Ganzhou, Jiangxi, 341000, P.R. China
| | - Lan Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Fan Yin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Weiqiang Hu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Yuling Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| |
Collapse
|
9
|
Xue P, Xi H, Tan X, Chen H, Peng C, Sun G, Ye Y, Jiang X, Liu X, Du B. Near-Infrared Responsive Properties of Bone Repair Scaffolds Facilitated by Specific Osteoinductive Photothermal Converters for Highly Efficient Bone Repair. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37581-37595. [PMID: 38985579 DOI: 10.1021/acsami.4c06346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
The effective repair of bone defects has long been a major challenge in clinical practice. Currently, research efforts mostly focus on achieving sufficiently good bone repair, with little attention paid to achieving both good and fast repair. However, achieving highly efficient (H-efficient) bone repair, which is both good and fast, can shorten the treatment cycle and facilitate rapid patient recovery. Therefore, the development of a H-efficient bone repair material is of significant importance. This study incorporated the previously developed osteoinductive photothermal agent (PTA) BPICT into printing paste to prepare a near-infrared (NIR)-responsive BPICT scaffold. Subsequently, the effects of photothermal therapy (PTT) on bone repair and drug release were assessed in vitro. To further validate the H-efficient bone repair properties of the BPICT scaffold, the scaffold was implanted into bone defects and its ability to promote bone repair in vivo was evaluated through radiology and histopathological analysis. The results indicated that compared to scaffolds containing only Icaritin (ICT), the BPICT scaffold can achieve PTT to promote bone repair through NIR irradiation, while also enabling the controlled release of ICT from the scaffold to enhance bone repair. Within the same observation period, the BPICT scaffold achieves more efficient bone repair than the ICT scaffold, significantly shortening the bone repair cycle while ensuring the effectiveness of bone repair. Therefore, the NIR-responsive scaffold based on PTT-mediated controlled release of bone growth factors represents a feasible solution for promoting H-efficient bone repair in the area of bone defects.
Collapse
Affiliation(s)
- Peng Xue
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Hongzhong Xi
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Xiaoxue Tan
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Chen
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Chenjian Peng
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Guangquan Sun
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Youqing Ye
- International Chinese-Belorussian Scientific Laboratory on Vacuum-Plasma Technology, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Xiaohong Jiang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Liu
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Bin Du
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| |
Collapse
|
10
|
Mao J, Bi J, Sun Z, Wang L. MgSiO 3 Fiber Membrane Scaffold with Triggered Drug Delivery for Osteosarcoma Synergetic Therapy and Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34669-34683. [PMID: 38946103 DOI: 10.1021/acsami.4c05744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
In this research, a novel MgSiO3 fiber membrane (MSFM) loaded with indocyanine green (ICG) and doxorubicin (DOX) was prepared. Because of MgSiO3's unique lamellar structure composed of a silicon-oxygen tetrahedron, magnesium ion (Mg2+) moves easily and can be further replaced with other cations. Therefore, because of the positively charged functional group of ICG, MSFM has a rather high drug loading for ICG. In addition, there is electrostatic attraction between DOX (a cationic drug) and ICG (an anionic drug). Hence, after loading ICG, more DOX can be adsorbed into MSFM because of electrostatic interaction. The ICG endows the MSFM outstanding photothermal therapy (PTT) performance, and DOX as a chemotherapeutic drug can restrain tumor growth. On the one hand, H+ exchanged with the positively charged DOX based on the MgSiO3 special lamellar structure. On the other hand, the thermal effect could break the electrostatic interaction between ICG and DOX. Based on the above two points, both tumor acidic microenvironment and photothermal effect can trigger DOX release. What's more, in vitro and in vivo antiosteosarcoma therapy evaluations displayed a superior synergetic PTT-chemotherapy anticancer treatment and excellent biocompatibility of DOX&ICG-MSFM. Finally, the MSFM was proven to greatly promote cell proliferation, differentiation, and bone regeneration performance in vitro and in vivo. Therefore, MSFM provides a creative perspective in the design of multifunctional scaffolds and shows promising applications in controlled drug delivery, antitumor performance, and osteogenesis.
Collapse
Affiliation(s)
- Junjie Mao
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), Shandong University, Jinan 250061, P. R. China
- Schools of Materials Science and Engineering, Shandong University, Jinan 250061, P. R. China
| | - Jianqiang Bi
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), Shandong University, Jinan 250061, P. R. China
- Schools of Materials Science and Engineering, Shandong University, Jinan 250061, P. R. China
| | - Zhenqian Sun
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P. R. China
- The First Clinical Medical School, Shandong University, Jinan, Shandong 250012, P. R. China
| | - Lu Wang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), Shandong University, Jinan 250061, P. R. China
- Schools of Materials Science and Engineering, Shandong University, Jinan 250061, P. R. China
| |
Collapse
|
11
|
Du Q, Dickinson A, Nakuleswaran P, Maghami S, Alagoda S, Hook AL, Ghaemmaghami AM. Targeting Macrophage Polarization for Reinstating Homeostasis following Tissue Damage. Int J Mol Sci 2024; 25:7278. [PMID: 39000385 PMCID: PMC11242417 DOI: 10.3390/ijms25137278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Tissue regeneration and remodeling involve many complex stages. Macrophages are critical in maintaining micro-environmental homeostasis by regulating inflammation and orchestrating wound healing. They display high plasticity in response to various stimuli, showing a spectrum of functional phenotypes that vary from M1 (pro-inflammatory) to M2 (anti-inflammatory) macrophages. While transient inflammation is an essential trigger for tissue healing following an injury, sustained inflammation (e.g., in foreign body response to implants, diabetes or inflammatory diseases) can hinder tissue healing and cause tissue damage. Modulating macrophage polarization has emerged as an effective strategy for enhancing immune-mediated tissue regeneration and promoting better integration of implantable materials in the host. This article provides an overview of macrophages' functional properties followed by discussing different strategies for modulating macrophage polarization. Advances in the use of synthetic and natural biomaterials to fabricate immune-modulatory materials are highlighted. This reveals that the development and clinical application of more effective immunomodulatory systems targeting macrophage polarization under pathological conditions will be driven by a detailed understanding of the factors that regulate macrophage polarization and biological function in order to optimize existing methods and generate novel strategies to control cell phenotype.
Collapse
Affiliation(s)
- Qiran Du
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Anna Dickinson
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Pruthvi Nakuleswaran
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK;
| | - Savindu Alagoda
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Andrew L. Hook
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Amir M. Ghaemmaghami
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| |
Collapse
|
12
|
Wang H, Zhang Y, Zhang Y, Li C, Zhang M, Wang J, Zhang Y, Du Y, Cui W, Chen W. Activating Macrophage Continual Efferocytosis via Microenvironment Biomimetic Short Fibers for Reversing Inflammation in Bone Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402968. [PMID: 38706203 DOI: 10.1002/adma.202402968] [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: 02/27/2024] [Revised: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Efferocytosis-mediated inflammatory reversal plays a crucial role in bone repairing process. However, in refractory bone defects, the macrophage continual efferocytosis may be suppressed due to the disrupted microenvironment homeostasis, particularly the loss of apoptotic signals and overactivation of intracellular oxidative stress. In this study, a polydopamine-coated short fiber matrix containing biomimetic "apoptotic signals" to reconstruct the microenvironment and reactivate macrophage continual efferocytosis for inflammatory reversal and bone defect repair is presented. The "apoptotic signals" (AM/CeO2) are prepared using CeO2 nanoenzymes with apoptotic neutrophil membrane coating for macrophage recognition and oxidative stress regulation. Additionally, a short fiber "biomimetic matrix" is utilized for loading AM/CeO2 signals via abundant adhesion sites involving π-π stacking and hydrogen bonding interactions. Ultimately, the implantable apoptosis-mimetic nanoenzyme/short-fiber matrixes (PFS@AM/CeO2), integrating apoptotic signals and biomimetic matrixes, are constructed to facilitate inflammatory reversal and reestablish the pro-efferocytosis microenvironment. In vitro and in vivo data indicate that the microenvironment biomimetic short fibers can activate macrophage continual efferocytosis, leading to the suppression of overactivated inflammation. The enhanced repair of rat femoral defect further demonstrates the osteogenic potential of the pro-efferocytosis strategy. It is believed that the regulation of macrophage efferocytosis through microenvironment biomimetic materials can provide a new perspective for tissue repair.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedic Surgery, the Hebei Medical University Third Hospital, Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, No.139 Ziqiang Road, Shijiazhuang, 050051, P. R. China
- 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, P. R. China
| | - Yu Zhang
- 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, P. R. China
| | - Yipu Zhang
- 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, P. R. China
- Department of Orthopaedics, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, P. R. China
| | - Chao Li
- Department of Orthopaedic Surgery, the Hebei Medical University Third Hospital, Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, No.139 Ziqiang Road, Shijiazhuang, 050051, P. R. China
- 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, P. R. China
| | - Mo Zhang
- Department of Orthopaedic Surgery, the Hebei Medical University Third Hospital, Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, No.139 Ziqiang Road, Shijiazhuang, 050051, P. R. China
| | - Juan Wang
- 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, P. R. China
| | - Yingze Zhang
- Department of Orthopaedic Surgery, the Hebei Medical University Third Hospital, Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, No.139 Ziqiang Road, Shijiazhuang, 050051, P. R. China
| | - Yawei Du
- 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, P. R. China
| | - Wenguo Cui
- 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, P. R. China
| | - Wei Chen
- Department of Orthopaedic Surgery, the Hebei Medical University Third Hospital, Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, No.139 Ziqiang Road, Shijiazhuang, 050051, P. R. China
| |
Collapse
|
13
|
Xiao L, Liu H, Huang H, Wu S, Xue L, Geng Z, Cai L, Yan F. 3D nanofiber scaffolds from 2D electrospun membranes boost cell penetration and positive host response for regenerative medicine. J Nanobiotechnology 2024; 22:322. [PMID: 38849858 PMCID: PMC11162076 DOI: 10.1186/s12951-024-02578-2] [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: 11/10/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
The ideal tissue engineering scaffold should facilitate rapid cell infiltration and provide an optimal immune microenvironment during interactions with the host. Electrospinning can produce two-dimensional (2D) membranes mimicking the extracellular matrix. However, their dense structure hinders cell penetration, and their thin form restricts scaffold utility. In this study, latticed hydrogels were three-dimensional (3D) printed onto electrospun membranes. This technique allowed for layer-by-layer assembly of the membranes into 3D scaffolds, which maintained their resilience impressively under both dry and wet conditions. We assessed the cellular and host responses of these 3D nanofiber scaffolds by comparing random membranes and mesh-like membranes with three different mesh sizes (250, 500, and 750 μm). It was found that scaffolds with a mesh size of 500 μm were superior for M2 macrophage phenotype polarization, vascularization, and matrix deposition. Furthermore, it was confirmed by subsequent experiments such as RNA sequencing that the mesh-like topology may promote polarization to the M2 phenotype by affecting the PI3K/AKT pathway. In conclusion, our work offers a novel method for transforming 2D nanofiber membranes into 3D scaffolds. This method boasts flexibility, allowing for the use of varied electrospun membranes and hydrogels in terms of structure and composition. It has vast potential in tissue repair and regeneration.
Collapse
Affiliation(s)
- Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Huifan Liu
- Department of Anesthesiology, Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Huayi Huang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shujuan Wu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Longjian Xue
- The Institute of Technological Science, School of Power and Mechanical Engineering, Wuhan University, Wuhan, 430072, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Feifei Yan
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
14
|
Özcolak B, Erenay B, Odabaş S, Jandt KD, Garipcan B. Effects of bone surface topography and chemistry on macrophage polarization. Sci Rep 2024; 14:12721. [PMID: 38830871 PMCID: PMC11148019 DOI: 10.1038/s41598-024-62484-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
Surface structure plays a crucial role in determining cell behavior on biomaterials, influencing cell adhesion, proliferation, differentiation, as well as immune cells and macrophage polarization. While grooves and ridges stimulate M2 polarization and pits and bumps promote M1 polarization, these structures do not accurately mimic the real bone surface. Consequently, the impact of mimicking bone surface topography on macrophage polarization remains unknown. Understanding the synergistic sequential roles of M1 and M2 macrophages in osteoimmunomodulation is crucial for effective bone tissue engineering. Thus, exploring the impact of bone surface microstructure mimicking biomaterials on macrophage polarization is critical. In this study, we aimed to sequentially activate M1 and M2 macrophages using Poly-L-Lactic acid (PLA) membranes with bone surface topographical features mimicked through the soft lithography technique. To mimic the bone surface topography, a bovine femur was used as a model surface, and the membranes were further modified with collagen type-I and hydroxyapatite to mimic the bone surface microenvironment. To determine the effect of these biomaterials on macrophage polarization, we conducted experimental analysis that contained estimating cytokine release profiles and characterizing cell morphology. Our results demonstrated the potential of the hydroxyapatite-deposited bone surface-mimicked PLA membranes to trigger sequential and synergistic M1 and M2 macrophage polarizations, suggesting their ability to achieve osteoimmunomodulatory macrophage polarization for bone tissue engineering applications. Although further experimental studies are required to completely investigate the osteoimmunomodulatory effects of these biomaterials, our results provide valuable insights into the potential advantages of biomaterials that mimic the complex microenvironment of bone surfaces.
Collapse
Affiliation(s)
- Birgün Özcolak
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Berkay Erenay
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
| | - Sedat Odabaş
- Biomaterials and Tissue Engineering Laboratory (bteLAB), Department of Chemistry, Faculty of Science, Ankara University, 06560, Ankara, Turkey
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, 06560, Ankara, Turkey
| | - Klaus D Jandt
- Chair of Materials Science (CMS), Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
| | - Bora Garipcan
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey.
| |
Collapse
|
15
|
Lv N, Hou M, Deng L, Hua X, Zhou X, Liu H, Zhu X, Xu Y, Qian Z, Li Q, Liu M, He F. A sponge-like nanofiber melatonin-loaded scaffold accelerates vascularized bone regeneration via improving mitochondrial energy metabolism. Mater Today Bio 2024; 26:101078. [PMID: 38765244 PMCID: PMC11101953 DOI: 10.1016/j.mtbio.2024.101078] [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: 03/02/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/21/2024] Open
Abstract
Electrospun nanofibers have been widely employed in bone tissue engineering for their ability to mimic the micro to nanometer scale network of the native bone extracellular matrix. However, the dense fibrous structure and limited mechanical support of these nanofibers pose challenges for the treatment of critical size bone defects. In this study, we propose a facile approach for creating a three-dimensional scaffold using interconnected electrospun nanofibers containing melatonin (Scaffold@MT). The hypothesis posited that the sponge-like Scaffold@MT could potentially enhance bone regeneration and angiogenesis by modulating mitochondrial energy metabolism. Melatonin-loaded gelatin and poly-lactic-acid nanofibers were fabricated using electrospinning, then fragmented into shorter fibers. The sponge-like Scaffold@MT was created through a process involving homogenization, low-temperature lyophilization, and chemical cross-linking, while maintaining the microstructure of the continuous nanofibers. The incorporation of short nanofibers led to a low release of melatonin and increased Young's modulus of the scaffold. Scaffold@MT demonstrated positive biocompatibility by promoting a 14.2 % increase in cell proliferation. In comparison to the control group, Scaffold@MT significantly enhanced matrix mineralization by 3.2-fold and upregulated the gene expression of osteoblast-specific markers, thereby facilitating osteogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs). Significantly, Scaffold@MT led to a marked enhancement in the mitochondrial energy function of BMMSCs, evidenced by elevated adenosine triphosphate (ATP) production, mitochondrial membrane potential, and protein expression of respiratory chain factors. Furthermore, Scaffold@MT promoted the migration of human umbilical vein endothelial cells (HUVECs) and increased tube formation by 1.3 times compared to the control group, accompanied by an increase in vascular endothelial growth factor (VEGFA) expression. The results of in vivo experiments indicate that the implantation of Scaffold@MT significantly improved vascularized bone regeneration in a distal femur defect in rats. Micro-computed tomography analysis conducted 8 weeks post-surgery revealed that Scaffold@MT led to optimal development of new bone microarchitecture. Histological and immunohistochemical analyses demonstrated that Scaffold@MT facilitated bone matrix deposition and new blood vessel formation at the defect site. Overall, the utilization of melatonin-loaded nanofiber sponges exhibits significant promise as a scaffold that promotes bone growth and angiogenesis, making it a viable option for the repair of critical-sized bone defects.
Collapse
Affiliation(s)
- Nanning Lv
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Department of Orthopaedics, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, 222003, China
- Department of Orthopaedics, Lianyungang Second People's Hospital Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222003, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Lei Deng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xi Hua
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xinfeng Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Hao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yong Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Zhonglai Qian
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Qing Li
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Mingming Liu
- Department of Orthopaedics, The Affiliated Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, 222003, China
- Department of Orthopaedics, Lianyungang Second People's Hospital Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222003, China
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215000, China
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| |
Collapse
|
16
|
Luo C, Li YM, Jiang K, Wang K, Kuzmanović M, You XH, Zhang Y, Lei J, Huang SS, Xu JZ. ECM-inspired calcium/zinc laden cellulose scaffold for enhanced bone regeneration. Carbohydr Polym 2024; 331:121823. [PMID: 38388030 DOI: 10.1016/j.carbpol.2024.121823] [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: 10/18/2023] [Revised: 01/01/2024] [Accepted: 01/11/2024] [Indexed: 02/24/2024]
Abstract
Cellulose-based polymer scaffolds are highly diverse for designing and fabricating artificial bone substitutes. However, realizing the multi-biological functions of cellulose-based scaffolds has long been challenging. In this work, inspired by the structure and function of the extracellular matrix (ECM) of bone, we developed a novel yet feasible strategy to prepare ECM-like scaffolds with hybrid calcium/zinc mineralization. The 3D porous structure was formed via selective oxidation and freeze drying of bacterial cellulose. Following the principle of electrostatic interaction, calcium/zinc hybrid hydroxyapatite nucleated, crystallized, and precipitated on the 3D scaffold in simulated physiological conditions, which was well confirmed by morphology and composition analysis. Compared with alternative scaffold cohorts, this hybrid ion-loaded cellulose scaffold exhibited a pronounced elevation in alkaline phosphatase (ALP) activity, osteogenic gene expression, and cranial defect regeneration. Notably, the hybrid ion-loaded cellulose scaffold effectively fostered an M2 macrophage milieu and had a strong immune effect in vivo. In summary, this study developed a hybrid multifunctional cellulose-based scaffold that appropriately simulates the ECM to regulate immunomodulatory and osteogenic differentiation, setting a measure for artificial bone substitutes.
Collapse
Affiliation(s)
- Chuan Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yuan-Min Li
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Kai Jiang
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Kai Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Maja Kuzmanović
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xuan-He You
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yao Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Jun Lei
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Shi-Shu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China.
| | - Jia-Zhuang Xu
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
17
|
Huang L, Song Z, Wang J, Bian M, Zou J, Zou Y, Ge J, Lu S. Absorbable calcium and phosphorus bioactive membranes promote bone marrow mesenchymal stem cells osteogenic differentiation for bone regeneration. Open Life Sci 2024; 19:20220854. [PMID: 38633414 PMCID: PMC11022123 DOI: 10.1515/biol-2022-0854] [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: 11/09/2023] [Revised: 01/29/2024] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Large segmental bone defects are commonly operated with autologous bone grafting, which has limited bone sources and poses additional surgical risks. In this study, we fabricated poly(lactide-co-glycolic acid) (PLGA)/β-tricalcium phosphate (β-TCP) composite membranes by electrostatic spinning and further promoted osteogenesis by regulating the release of β-TCP in the hope of replacing autologous bone grafts in the clinical practice. The addition of β-TCP improved the mechanical strength of PLGA by 2.55 times. Moreover, β-TCP could accelerate the degradation of PLGA and neutralize the negative effects of acidification of the microenvironment caused by PLGA degradation. In vitro experiments revealed that PLGA/TCP10 membranes are biocompatible and the released β-TCP can modulate the activity of osteoblasts by enhancing the calcium ions concentration in the damaged area and regulating the pH of the local microenvironment. Simultaneously, an increase in β-TCP can moderate the lactate content of the local microenvironment, synergistically enhancing osteogenesis by promoting the tube-forming effect of human umbilical vein endothelial cells. Therefore, it is potential to utilize PLGA/TCP bioactive membranes to modulate the microenvironment at the site of bone defects to promote bone regeneration.
Collapse
Affiliation(s)
- Lei Huang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhuorun Song
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jiayi Wang
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiapeng Zou
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yanpei Zou
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Ge
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| |
Collapse
|
18
|
Lu F, Verleg SMNE, Groven RVM, Poeze M, van Griensven M, Blokhuis TJ. Is there a role for N1-N2 neutrophil phenotypes in bone regeneration? A systematic review. Bone 2024; 181:117021. [PMID: 38253189 DOI: 10.1016/j.bone.2024.117021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/02/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024]
Abstract
PURPOSE This review aims to provide an overview of the multiple functions of neutrophils, with the recognition of the inflammatory (N1) and regenerative (N2) phenotypes, in relation to fracture healing. METHODS A literature search was performed using the PubMed database. The quality of the articles was evaluated using critical appraisal checklists. RESULTS Thirty one studies were included in this review. These studies consistently support that neutrophils exert both beneficial and detrimental effects on bone regeneration, influenced by Tumor Necrosis Factor-α (TNF-α), Interleukin 8 (IL-8), mast cells, and macrophages. The N2 phenotype has recently emerged as one promoter of bone healing. The N1 phenotype has progressively been connected with inflammatory neutrophils during fracture healing. CONCLUSIONS This review has pinpointed various aspects and mechanisms of neutrophil influence on bone healing. The recognition of N1 and N2 neutrophil phenotypes potentially shed new light on the dynamic shifts taking place within the Fracture Hematoma (FH).
Collapse
Affiliation(s)
- Fangzhou Lu
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands; Division of Trauma Surgery, Department of Surgery, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands.
| | - Samai M N E Verleg
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands; Division of Trauma Surgery, Department of Surgery, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands.
| | - Rald V M Groven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands; Division of Trauma Surgery, Department of Surgery, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands.
| | - Martijn Poeze
- Division of Trauma Surgery, Department of Surgery, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands.
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands.
| | - Taco J Blokhuis
- Division of Trauma Surgery, Department of Surgery, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands.
| |
Collapse
|
19
|
Huang XY, Zhou XX, Yang H, Xu T, Dao JW, Bian L, Wei DX. Directed osteogenic differentiation of human bone marrow mesenchymal stem cells via sustained release of BMP4 from PBVHx-based nanoparticles. Int J Biol Macromol 2024; 265:130649. [PMID: 38453121 DOI: 10.1016/j.ijbiomac.2024.130649] [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: 01/21/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Bone Morphogenetic Protein 4 (BMP4) is crucial for bone and cartilage tissue regeneration, essential in medical tissue engineering, cosmetology, and aerospace. However, its cost and degradation susceptibility pose significant clinical challenges. To enhance its osteogenic activity while reducing dosage and administration frequency, we developed a novel long-acting BMP4 delivery system using poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-hydroxyhexanoate) (PBVHx) nanoparticles with soybean lecithin-modified BMP4 (sBP-NPs). These nanoparticles promote directed osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) through sustained BMP4 release. sBP-NPs exhibited uniform size (100-200 nm) and surface charges, with higher BMP4 entrapment efficiency (82.63 %) compared to controls. After an initial burst release within 24 h, sBP-NPs achieved 80 % cumulative BMP4 release within 20 days, maintaining levels better than control BP-NPs with unmodified BMP4. Co-incubation and nanoparticle uptake experiments confirmed excellent biocompatibility of sBP-NPs, promoting hBMSC differentiation towards osteogenic lineage with increased expression of type I collagen, calcium deposition, and ALP activity (> 20,000 U/g protein) compared to controls. Moreover, hBMSCs treated with sBP-NPs exhibited heightened expression of osteogenic genetic markers, surpassing control groups. Hence, this innovative strategy of sustained BMP4 release from sBP-NPs holds potential to revolutionize bone regeneration in minimally invasive surgery, medical cosmetology or space environments.
Collapse
Affiliation(s)
- Xiao-Yun Huang
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China; Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Xiao-Xiang Zhou
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China
| | - Hui Yang
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China
| | - Tao Xu
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China
| | - Jin-Wei Dao
- Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong 643002, China
| | - Li Bian
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Dai-Xu Wei
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China; School of Clinical Medicine, Chengdu University, Chengdu, China; Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong 643002, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| |
Collapse
|
20
|
Hessel E, Ghanta P, Winschel T, Melnyk L, Oyewumi MO. Fabrication of 3D-printed scaffolds loaded with gallium acetylacetonate for potential application in osteoclastic bone resorption. Pharm Dev Technol 2024; 29:339-352. [PMID: 38502579 DOI: 10.1080/10837450.2024.2332459] [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: 11/09/2023] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
We recently reported the potential of a new gallium compound, gallium acetylacetonate (GaAcAc) in combating osteoclastic bone resorption through inhibition of osteoclast differentiation and function. Herein, we focused on 3D-printed polylactic acid scaffolds that were loaded with GaAcAc and investigated the impact of scaffold pretreatment with polydopamine (PDA) or sodium hydroxide (NaOH). We observed a remarkable increase in scaffold hydrophilicity with PDA or NaOH pretreatment while biocompatibility and in vitro degradation were not affected. NaOH-pretreated scaffolds showed the highest amount of GaAcAc loading when compared to other scaffolds (p < 0.05). NaOH-pretreated scaffolds with GaAcAc loading showed effective reduction of osteoclast counts and size. The trend was supported by suppression of key osteoclast differentiation markers such as NFAT2, c-Fos, TRAF6, & TRAP. All GaAcAc-loaded scaffolds, regardless of surface pretreatment, were effective in inhibiting osteoclast function as evidenced by reduction in the number of resorptive pits in bovine cortical bone slices (p < 0.01). The suppression of osteoclast function according to the type of scaffold followed the ranking: GaAcAc loading without surface pretreatment > GaAcAc loading with NaOH pretreatment > GaAcAc loading with PDA pretreatment. Additional studies will be needed to fully elucidate the impact of surface pretreatment on the efficacy and safety of GaAcAc-loaded 3D-printed scaffolds.
Collapse
Affiliation(s)
- Evin Hessel
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Pratyusha Ghanta
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Timothy Winschel
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Larissa Melnyk
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Moses O Oyewumi
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
21
|
Jin S, Wen J, Zhang Y, Mou P, Luo Z, Cai Y, Chen A, Fu X, Meng W, Zhou Z, Li J, Zeng W. M2 macrophage-derived exosome-functionalized topological scaffolds regulate the foreign body response and the coupling of angio/osteoclasto/osteogenesis. Acta Biomater 2024; 177:91-106. [PMID: 38311198 DOI: 10.1016/j.actbio.2024.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Abstract
Designing scaffolds that can regulate the innate immune response and promote vascularized bone regeneration holds promise for bone tissue engineering. Herein, electrospun scaffolds that combined physical and biological cues were fabricated by anchoring reparative M2 macrophage-derived exosomes onto topological pore structured nanofibrous scaffolds. The topological pore structure of the fiber and the immobilization of exosomes increased the nanoscale roughness and hydrophilicity of the fibrous scaffold. In vitro cell experiments showed that exosomes could be internalized by target cells to promote cell migration, tube formation, osteogenic differentiation, and anti-inflammatory macrophage polarization. The activation of fibrosis, angiogenesis, and macrophage was elucidated during the exosome-functionalized fibrous scaffold-mediated foreign body response (FBR) in subcutaneous implantation in mice. The exosome-functionalized nanofibrous scaffolds also enhanced vascularized bone formation in a critical-sized rat cranial bone defect model. Importantly, histological analysis revealed that the biofunctional scaffolds regulated the coupling effect of angiogenesis, osteoclastogenesis, and osteogenesis by stimulating type H vessel formation. This study elaborated on the complex processes within the cell microenvironment niche during fibrous scaffold-mediated FBR and vascularized bone regeneration to guide the design of implants or devices used in orthopedics and maxillofacial surgery. STATEMENT OF SIGNIFICANCE: How to design scaffold materials that can regulate the local immune niche and truly achieve functional vascularized bone regeneration still remain an open question. Here, combining physical and biological cues, we proposed new insight to cell-free and growth factor-free therapy, anchoring reparative M2 macrophage-derived exosomes onto topological pore structured nanofibrous scaffolds. The exosomes functionalized-scaffold system mitigated foreign body response, including excessive fibrosis, tumor-like vascularization, and macrophage activation. Importantly, the biofunctional scaffolds regulated the coupling effect of angiogenesis, osteoclastogenesis, and osteogenesis by stimulating type H vessel formation.
Collapse
Affiliation(s)
- Shue Jin
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Wen
- Analytical & Testing Center, Sichuan University, Chengdu 610065, China
| | - Yao Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Mou
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zeyu Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongrui Cai
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Anjin Chen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoxue Fu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weikun Meng
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zongke Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jidong Li
- Analytical & Testing Center, Sichuan University, Chengdu 610065, China
| | - Weinan Zeng
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
22
|
Wang X, Fu X, Luo D, Hou R, Li P, Chen Y, Zhang X, Meng X, Yue Y, Liu J. 3D printed high-precision porous scaffolds prepared by fused deposition modeling induce macrophage polarization to promote bone regeneration. Biomed Mater 2024; 19:035006. [PMID: 38422525 DOI: 10.1088/1748-605x/ad2ed0] [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: 10/31/2023] [Accepted: 02/29/2024] [Indexed: 03/02/2024]
Abstract
Macrophage-mediated bone immune responses significantly influence the repair of bone defects when utilizing tissue-engineered scaffolds. Notably, the scaffolds' physical structure critically impacts macrophage polarization. The optimal pore size for facilitating bone repair remains a topic of debate due to the imprecision of traditional methods in controlling scaffold pore dimensions and spatial architecture. In this investigation, we utilized fused deposition modeling (FDM) technology to fabricate high-precision porous polycaprolactone (PCL) scaffolds, aiming to elucidate the impact of pore size on macrophage polarization. We assessed the scaffolds' mechanical attributes and biocompatibility. Real-time quantitative reverse transcription polymerase chain reaction was used to detect the expression levels of macrophage-related genes, and enzyme linked immunosorbent assay for cytokine secretion levels.In vitroosteogenic capacity was determined through alkaline phosphatase and alizarin red staining. Our findings indicated that macroporous scaffolds enhanced macrophage adhesion and drove their differentiation towards the M2 phenotype. This led to the increased production of anti-inflammatory factors and a reduction in pro-inflammatory agents, highlighting the scaffolds' immunomodulatory capabilities. Moreover, conditioned media from macrophages cultured on these macroporous scaffolds bolstered the osteogenic differentiation of bone marrow mesenchymal stem cells, exhibiting superior osteogenic differentiation potential. Consequently, FDM-fabricated PCL scaffolds, with precision-controlled pore sizes, present promising prospects as superior materials for bone tissue engineering, leveraging the regulation of macrophage polarization.
Collapse
Affiliation(s)
- Xiangyu Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Xinyu Fu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Dongmei Luo
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Ruxia Hou
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Peiwen Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Yurou Chen
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Xinyao Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Xiangjie Meng
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Yingge Yue
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| | - Junyu Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, People's Republic of China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, People's Republic of China
| |
Collapse
|
23
|
Yuan Z, Jiang D, Yang M, Tao J, Hu X, Yang X, Zeng Y. Emerging Roles of Macrophage Polarization in Osteoarthritis: Mechanisms and Therapeutic Strategies. Orthop Surg 2024; 16:532-550. [PMID: 38296798 PMCID: PMC10925521 DOI: 10.1111/os.13993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 02/02/2024] Open
Abstract
Osteoarthritis (OA) is the most common chronic degenerative joint disease in middle-aged and elderly people, characterized by joint pain and dysfunction. Macrophages are key players in OA pathology, and their activation state has been studied extensively. Various studies have suggested that macrophages might respond to stimuli in their microenvironment by changing their phenotypes to pro-inflammatory or anti-inflammatory phenotypes, which is called macrophage polarization. Macrophages accumulate and become polarized (M1 or M2) in many tissues, such as synovium, adipose tissue, bone marrow, and bone mesenchymal tissues in joints, while resident macrophages as well as other stromal cells, including fibroblasts, chondrocytes, and osteoblasts, form the joint and function as an integrated unit. In this study, we focus exclusively on synovial macrophages, adipose tissue macrophages, and osteoclasts, to investigate their roles in the development of OA. We review recent key findings related to macrophage polarization and OA, including pathogenesis, molecular pathways, and therapeutics. We summarize several signaling pathways in macrophage reprogramming related to OA, including NF-κB, MAPK, TGF-β, JAK/STAT, PI3K/Akt/mTOR, and NLRP3. Of note, despite the increasing availability of treatments for osteoarthritis, like intra-articular injections, surgery, and cellular therapy, the demand for more effective clinical therapies has remained steady. Therefore, we also describe the current prospective therapeutic methods that deem macrophage polarization to be a therapeutic target, including physical stimulus, chemical compounds, and biological molecules, to enhance cartilage repair and alleviate the progression of OA.
Collapse
Affiliation(s)
- Zimu Yuan
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Decheng Jiang
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Mengzhu Yang
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Jie Tao
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Xin Hu
- Orthopedic Research Institute, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengduChina
| | - Xiao Yang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduChina
| | - Yi Zeng
- Orthopedic Research Institute, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
24
|
Yang SY, Zhou YN, Yu XG, Fu ZY, Zhao CC, Hu Y, Lin KL, Xu YJ. A xonotlite nanofiber bioactive 3D-printed hydrogel scaffold based on osteo-/angiogenesis and osteoimmune microenvironment remodeling accelerates vascularized bone regeneration. J Nanobiotechnology 2024; 22:59. [PMID: 38347563 PMCID: PMC10863132 DOI: 10.1186/s12951-024-02323-9] [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: 08/22/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Coordination between osteo-/angiogenesis and the osteoimmune microenvironment is essential for effective bone repair with biomaterials. As a highly personalized and precise biomaterial suitable for repairing complex bone defects in clinical practice, it is essential to endow 3D-printed scaffold the above key capabilities. RESULTS Herein, by introducing xonotlite nanofiber (Ca6(Si6O17) (OH)2, CS) into the 3D-printed silk fibroin/gelatin basal scaffold, a novel bone repair system named SGC was fabricated. It was noted that the incorporation of CS could greatly enhance the chemical and mechanical properties of the scaffold to match the needs of bone regeneration. Besides, benefiting from the addition of CS, SGC scaffolds could accelerate osteo-/angiogenic differentiation of bone mesenchymal stem cells (BMSCs) and meanwhile reprogram macrophages to establish a favorable osteoimmune microenvironment. In vivo experiments further demonstrated that SGC scaffolds could efficiently stimulate bone repair and create a regeneration-friendly osteoimmune microenvironment. Mechanistically, we discovered that SGC scaffolds may achieve immune reprogramming in macrophages through a decrease in the expression of Smad6 and Smad7, both of which participate in the transforming growth factor-β (TGF-β) signaling pathway. CONCLUSION Overall, this study demonstrated the clinical potential of the SGC scaffold due to its favorable pro-osteo-/angiogenic and osteoimmunomodulatory properties. In addition, it is a promising strategy to develop novel bone repair biomaterials by taking osteoinduction and osteoimmune microenvironment remodeling functions into account.
Collapse
Affiliation(s)
- Shi-Yuan Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Ning Zhou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Xing-Ge Yu
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Yu Fu
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Can-Can Zhao
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Hu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Kai-Li Lin
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuan-Jin Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
- College of Stomatology, National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
25
|
Xu Y, Saiding Q, Zhou X, Wang J, Cui W, Chen X. Electrospun fiber-based immune engineering in regenerative medicine. SMART MEDICINE 2024; 3:e20230034. [PMID: 39188511 PMCID: PMC11235953 DOI: 10.1002/smmd.20230034] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/26/2024] [Indexed: 08/28/2024]
Abstract
Immune engineering, a burgeoning field within regenerative medicine, involves a spectrum of strategies to optimize the intricate interplay between tissue regenerative biomaterials and the host tissue. These strategies are applied across different types of biomaterials and various disease models, which encompasses finely modulating the immune response at the levels of immune cells and factors, aiming to mitigate adverse effects like fibrosis and persistent inflammation that may arise at the injury site and consequently promote tissue regeneration. With the continuous progress in electrospinning technology, the immunoregulatory capabilities of electrospun fibers have gained substantial attention over the years. Electrospun fibers, with their extracellular matrix-like characteristics, high surface-area-to-volume ratio, and reliable pharmaceutical compound capacity, have emerged as key players among tissue engineering materials. This review specifically focuses on the role of electrospun fiber-based immune engineering, emphasizing their unique design strategies. Notably, electrospinning actively engages in immune engineering by modulating immune responses through four essential strategies: (i) surface modification, (ii) drug loading, (iii) physicochemical parameters, and (iv) biological grafting. This review presents a comprehensive overview of the intricate mechanisms of the immune system in injured tissues while unveiling the key strategies adopted by electrospun fibers to orchestrate immune regulation. Furthermore, the review explores the current developmental trends and limitations concerning the immunoregulatory function of electrospun fibers, aiming to drive the advancements in electrospun fiber-based immune engineering to its full potential.
Collapse
Affiliation(s)
- Yiru Xu
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Qimanguli Saiding
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue Zhou
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xinliang Chen
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| |
Collapse
|
26
|
Yuan Y, Xu Y, Mao Y, Liu H, Ou M, Lin Z, Zhao R, Long H, Cheng L, Sun B, Zhao S, Zeng M, Lu B, Lu H, Zhu Y, Chen C. Three Birds, One Stone: An Osteo-Microenvironment Stage-Regulative Scaffold for Bone Defect Repair through Modulating Early Osteo-Immunomodulation, Middle Neovascularization, and Later Osteogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306428. [PMID: 38060833 PMCID: PMC10853759 DOI: 10.1002/advs.202306428] [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: 09/06/2023] [Revised: 10/28/2023] [Indexed: 02/10/2024]
Abstract
In order to repair critical-sized bone defects, various polylactic acid-glycolic acid (PLGA)-based hybrid scaffolds are successfully developed as bone substitutes. However, the byproducts of these PLGA-based scaffolds are known to acidify the implanted site, inducing tiresome acidic inflammation. Moreover, these degradation productions cannot offer an osteo-friendly microenvironment at the implanted site, matching natural bone healing. Herein, inspired by bone microenvironment atlas of natural bone-healing process, an osteo-microenvironment stage-regulative scaffold (P80/D10/M10) is fabricated by incorporating self-developed decellularized bone matrix microparticles (DBM-MPs) and multifunctional magnesium hydroxide nanoparticles (MH-NPs) into PLGA with an optimized proportion using low-temperature rapid prototyping (LT-RP) 3D-printing technology. The cell experiments show that this P80/D10/M10 exhibits excellent properties in mechanics, biocompatibility, and biodegradability, meanwhile superior stimulations in osteo-immunomodulation, angiogenesis, and osteogenesis. Additionally, the animal experiments determined that this P80/D10/M10 can offer an osteo-friendly microenvironment in a stage-matched pattern for enhanced bone regeneration, namely, optimization of early inflammation, middle neovascularization, and later bone formation. Furthermore, transcriptomic analysis suggested that the in vivo performance of P80/D10/M10 on bone defect repair is mostly attributed to regulating artery development, bone development, and bone remodeling. Overall, this study reveals that the osteo-microenvironment stage-regulative scaffold provides a promising treatment for bone defect repair.
Collapse
Affiliation(s)
- Yuhao Yuan
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yan Xu
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
- Department of Sports MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yiyang Mao
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
| | - Hongbin Liu
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Minning Ou
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Zhangyuan Lin
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Ruibo Zhao
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Haitao Long
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Liang Cheng
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Buhua Sun
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Shushan Zhao
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Ming Zeng
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Bangbao Lu
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hongbin Lu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
- Department of Sports MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yong Zhu
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Can Chen
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaHunan410008China
| |
Collapse
|
27
|
Shi Y, Tao W, Yang W, Wang L, Qiu Z, Qu X, Dang J, He J, Fan H. Calcium phosphate coating enhances osteointegration of melt electrowritten scaffold by regulating macrophage polarization. J Nanobiotechnology 2024; 22:47. [PMID: 38297240 PMCID: PMC10829397 DOI: 10.1186/s12951-024-02310-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/26/2024] [Indexed: 02/02/2024] Open
Abstract
The osteoimmune microenvironment induced by implants plays a significant role in bone regeneration. It is essential to efficiently and timely switch the macrophage phenotype from M1 to M2 for optimal bone healing. This study examined the impact of a calcium phosphate (CaP) coating on the physiochemical properties of highly ordered polycaprolactone (PCL) scaffolds fabricated using melt electrowritten (MEW). Additionally, it investigated the influence of these scaffolds on macrophage polarization and their immunomodulation on osteogenesis. The results revealed that the CaP coated PCL scaffold exhibited a rougher surface topography and higher hydrophilicity in comparison to the PCL scaffold without coating. Besides, the surface morphology of the coating and the release of Ca2+ from the CaP coating were crucial in regulating the transition of macrophages from M1 to M2 phenotypes. They might activate the PI3K/AKT and cAMP-PKA pathways, respectively, to facilitate M2 polarization. In addition, the osteoimmune microenvironment induced by CaP coated PCL could not only enhance the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) in vitro but also promote the bone regeneration in vivo. Taken together, the CaP coating can be employed to control the phenotypic switching of macrophages, thereby creating a beneficial immunomodulatory microenvironment that promotes bone regeneration.
Collapse
Affiliation(s)
- Yubo Shi
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weidong Tao
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenjing Yang
- Xijing 986 Hospital Department, The Fourth Military Medical University, Xi'an, China
| | - Lei Wang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhennan Qiu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Dang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Hongbin Fan
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
28
|
Lv X, Zhang C, Liu X, Li P, Yang Y. 3D bioprinting technology to construct bone reconstruction research model and its feasibility evaluation. Front Bioeng Biotechnol 2024; 12:1328078. [PMID: 38314351 PMCID: PMC10834755 DOI: 10.3389/fbioe.2024.1328078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
Objective: To explore and construct a 3D bone remodeling research model displaying stability, repeatability, and precise simulation of the physiological and biochemical environment in vivo. Methods: In this study, 3D bioprinting was used to construct a bone reconstruction model. Sodium alginate (SA), hydroxyapatite (HA) and gelatin (Gel) were mixed into hydrogel as scaffold material. The osteoblast precursor cells MC3T3-E1 and osteoclast precursor cells RAW264.7 were used as seed cells, which may or may not be separated by polycarbonate membrane. The cytokines osteoprotegerin (OPG) and receptor activator of NF-κB ligand (RANKL) were used to induce cell differentiation. The function of scaffolds in the process of bone remodeling was analyzed by detecting the related markers of osteoblasts (alkaline phosphatase, ALP) and osteoclasts (tartrate resistant acid phosphatase, TRAP). Results: The scaffold showed good biocompatibility and low toxicity. The surface morphology aided cell adhesion and growth. The scaffold had optimum degradability, water absorption capacity and porosity, which are in line with the conditions of biological experiments. The effect of induced differentiation of cells was the best when cultured alone. After direct contact between the two types of cells at 2D or 3D level, the induced differentiation of cells was inhibited to varying degrees, although they still showed osteogenesis and osteoclast. After the cells were induced by indirect contact culture, the effect of induced differentiation improved when compared with direct contact culture, although it was still not as good as that of single culture. On the whole, the effect of inducing differentiation at 3D level was the same as that at 2D level, and its relative gene expression and enzyme activity were higher than that in the control group. Hence the scaffold used in this study could induce osteogenesis as well as osteoclast, thereby rendering it more effective in inducing new bone formation. Conclusion: This method can be used to construct the model of 3D bone remodeling mechanism.
Collapse
Affiliation(s)
- Xiao Lv
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Chenyang Zhang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Xingzhu Liu
- West China Hospital, Sichuan University, Hangzhou, China
| | - Ping Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yadong Yang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
29
|
Liu Y, Wang S, Quan C, Luan S, Shi H, Wang L. Metal-organic framework-based platforms for implantation applications: recent advances and challenges. J Mater Chem B 2024; 12:637-649. [PMID: 38165820 DOI: 10.1039/d3tb02620e] [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: 01/04/2024]
Abstract
The development of minimally invasive technology has promoted the widespread use of implant interventional materials, which play an important role in alleviating patients' pain during and after surgery. Metal-organic frameworks (MOFs) and their related hybrids formed by bridging ligands and metal nodes via covalent bonds represent one of the smart platforms in implant interventional fields due to their large surface area, adjustable compositions and structures, biodegradability, etc. Significant progresses in the implantation application of MOF-based materials have been achieved recently, but these studies are still in the initial stage. This review highlights the recent advances of MOFs and their related hybrids in orthopedic implantation, cardio-vascular implantation, neural tissue engineering, and biochemical sensing. Each correction between the structural features of MOFs and their corresponding implanted works is highlighted. Finally, the confronting challenges and future perspectives in the implant interventional field are discussed.
Collapse
Affiliation(s)
- Yifan Liu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Shuteng Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Chunhua Quan
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002, P. R. China.
| | - Shifang Luan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Hengchong Shi
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Lei Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
| |
Collapse
|
30
|
Hu Y, Tang L, Wang Z, Yan H, Yi X, Wang H, Ma L, Yang C, Ran J, Yu A. Inducing in situ M2 macrophage polarization to promote the repair of bone defects via scaffold-mediated sustained delivery of luteolin. J Control Release 2024; 365:889-904. [PMID: 37952829 DOI: 10.1016/j.jconrel.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
Immunoregulation mediated bone tissue engineering (BTE) has demonstrated huge potential in promoting repair of critical-size bone defects (CSBDs). The trade-off between stable immunoregulation function and extended immunoregulation period has posed a great challenge to this strategy. Here, we reported a 3D porous biodegradable Poly(HEMA-co-3APBA)/LUT scaffold, in which reversible boronic acid ester bond was formed between the 3APBA moiety and the catechol moiety of luteolin (LUT). The boronic acid ester bond not only protected the bioactivity of LUT but also extended the release period of LUT. The rationale behind the phenomenon of sustained LUT release was explained using a classical transition state theory. In vitro/in vivo assays proved the immunoregulation function of the scaffold in inducing M2 polarization of both M0 and M1 Mφ. The crosstalk between the scaffold treated Raw 264.7 and BMSCs were also investigated through the in vitro co-culture assay. The results demonstrated that the scaffold could induce immunoregulation mediated osteogenic differentiation of BMSCs. In addition, CSBDs model of SD rats was also applied, and the corresponding data proved that the scaffold could accelerate new bone formation, therefore promoting repair of CSBDs. The as-prepared scaffold might be a promising candidate for repair of CSBDs in the future.
Collapse
Affiliation(s)
- Yan Hu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Lixi Tang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Honghan Yan
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Xinzeyu Yi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Huimin Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Liya Ma
- Core Facility of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Changying Yang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jiabing Ran
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China.
| |
Collapse
|
31
|
Wu M, Liu H, Li D, Zhu Y, Wu P, Chen Z, Chen F, Chen Y, Deng Z, Cai L. Smart-Responsive Multifunctional Therapeutic System for Improved Regenerative Microenvironment and Accelerated Bone Regeneration via Mild Photothermal Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304641. [PMID: 37933988 PMCID: PMC10787108 DOI: 10.1002/advs.202304641] [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: 08/08/2023] [Indexed: 11/08/2023]
Abstract
The treatment of bone defects remains a substantial clinical challenge due to the lack of spatiotemporal management of the immune microenvironment, revascularization, and osteogenic differentiation. Herein, deferoxamine (DFO)-loaded black phosphorus nanosheets decorated by polydopamine layer are prepared (BPPD) and compounded into gelatin methacrylate/sodium alginate methacrylate (GA) hybrid hydrogel as a smart-responsive therapeutic system (GA/BPPD) for accelerated bone regeneration. The BPPD nanocomposites served as bioactive components and near-infrared (NIR) photothermal agents, which conferred the hydrogel with excellent NIR/pH dual-responsive properties, realizing the stimuli-responsive release of DFO and PO4 3 - during bone regeneration. Under the action of NIR-triggered mild photothermal therapy, the GA/BPPD hydrogel exhibited a positive effect on promoting osteogenesis and angiogenesis, eliminating excessive reactive oxygen species, and inducing macrophage polarization to the M2 phenotype. More significantly, through macrophage M2 polarization-induced osteoimmune microenvironment, this hydrogel platform could also drive functional cytokine secretion for enhanced angiogenesis and osteogenesis. In vivo experiments further demonstrated that the GA/BPPD system could facilitate bone healing by attenuating the local inflammatory response, increasing the secretion of pro-healing factors, stimulating endogenous cell recruitment, and accelerating revascularization. Collectively, the proposed intelligent photothermal hydrogel platform provides a promising strategy to reshape the damaged tissue microenvironment for augmented bone regeneration.
Collapse
Affiliation(s)
- Minhao Wu
- Department of Spine Surgery and Musculoskeletal TumorZhongnan Hospital of Wuhan University168 Donghu Street, Wuchang DistrictWuhanHubei430071P. R. China
| | - Huifan Liu
- Department of AnesthesiologyResearch Centre of Anesthesiology and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhanHubeiP. R. China
| | - Dan Li
- Department of Neonatology, Xianning Central hospitalSchool of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and TechnologyXianningHubei437100P. R. China
| | - Yufan Zhu
- Department of Spine Surgery and Musculoskeletal TumorZhongnan Hospital of Wuhan University168 Donghu Street, Wuchang DistrictWuhanHubei430071P. R. China
| | - Ping Wu
- Research Units of Clinical Translation of Cell Growth Factors and Diseases ResearchChinese Academy of Medical ScienceZhejiang325000P. R. China
| | - Zhe Chen
- Department of Spine Surgery and Musculoskeletal TumorZhongnan Hospital of Wuhan University168 Donghu Street, Wuchang DistrictWuhanHubei430071P. R. China
| | - Feixiang Chen
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medicine Sciences)Wuhan UniversityWuhan430071P. R. China
| | - Yun Chen
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medicine Sciences)Wuhan UniversityWuhan430071P. R. China
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal TumorZhongnan Hospital of Wuhan University168 Donghu Street, Wuchang DistrictWuhanHubei430071P. R. China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal TumorZhongnan Hospital of Wuhan University168 Donghu Street, Wuchang DistrictWuhanHubei430071P. R. China
| |
Collapse
|
32
|
Qiu M, Tulufu N, Tang G, Ye W, Qi J, Deng L, Li C. Black Phosphorus Accelerates Bone Regeneration Based on Immunoregulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304824. [PMID: 37953457 PMCID: PMC10767454 DOI: 10.1002/advs.202304824] [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: 07/16/2023] [Revised: 09/25/2023] [Indexed: 11/14/2023]
Abstract
A fundamental understanding of inflammation and tissue healing suggests that the precise regulation of the inflammatory phase, both in terms of location and timing, is crucial for bone regeneration. However, achieving the activation of early inflammation without causing chronic inflammation while facilitating quick inflammation regression to promote bone regeneration continues to pose challenges. This study reveals that black phosphorus (BP) accelerates bone regeneration by building an osteogenic immunological microenvironment. BP amplifies the acute pro-inflammatory response and promotes the secretion of anti-inflammatory factors to accelerate inflammation regression and tissue regeneration. Mechanistically, BP creates an osteoimmune-friendly microenvironment by stimulating macrophages to express interleukin 33 (IL-33), amplifying the inflammatory response at an early stage, and promoting the regression of inflammation. In addition, BP-mediated IL-33 expression directly promotes osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), which further facilitates bone repair. To the knowledge, this is the first study to reveal the immunomodulatory potential of BP in bone regeneration through the regulation of both early-stage inflammatory responses and later-stage inflammation resolution, along with the associated molecular mechanisms. This discovery serves as a foundation for the clinical use of BP and is an efficient approach for managing the immune microenvironment during bone regeneration.
Collapse
Affiliation(s)
- Minglong Qiu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Nijiati Tulufu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Guoqing Tang
- Kunshan Hospital of Traditional Chinese MedicineAffiliated Hospital of Yangzhou University388 Zuchongzhi RoadKunshan CityJiangsu Province215300P. R. China
| | - Wenkai Ye
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Jin Qi
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Lianfu Deng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Changwei Li
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| |
Collapse
|
33
|
Timofticiuc IA, Călinescu O, Iftime A, Dragosloveanu S, Caruntu A, Scheau AE, Badarau IA, Didilescu AC, Caruntu C, Scheau C. Biomaterials Adapted to Vat Photopolymerization in 3D Printing: Characteristics and Medical Applications. J Funct Biomater 2023; 15:7. [PMID: 38248674 PMCID: PMC10816811 DOI: 10.3390/jfb15010007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
Along with the rapid and extensive advancements in the 3D printing field, a diverse range of uses for 3D printing have appeared in the spectrum of medical applications. Vat photopolymerization (VPP) stands out as one of the most extensively researched methods of 3D printing, with its main advantages being a high printing speed and the ability to produce high-resolution structures. A major challenge in using VPP 3D-printed materials in medicine is the general incompatibility of standard VPP resin mixtures with the requirements of biocompatibility and biofunctionality. Instead of developing completely new materials, an alternate approach to solving this problem involves adapting existing biomaterials. These materials are incompatible with VPP 3D printing in their pure form but can be adapted to the VPP chemistry and general process through the use of innovative mixtures and the addition of specific pre- and post-printing steps. This review's primary objective is to highlight biofunctional and biocompatible materials that have been adapted to VPP. We present and compare the suitability of these adapted materials to different medical applications and propose other biomaterials that could be further adapted to the VPP 3D printing process in order to fulfill patient-specific medical requirements.
Collapse
Affiliation(s)
- Iosif-Aliodor Timofticiuc
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
| | - Octavian Călinescu
- Department of Biophysics, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
| | - Adrian Iftime
- Department of Biophysics, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
| | - Andreea Cristiana Didilescu
- Department of Embryology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
34
|
Sun W, Ye B, Chen S, Zeng L, Lu H, Wan Y, Gao Q, Chen K, Qu Y, Wu B, Lv X, Guo X. Neuro-bone tissue engineering: emerging mechanisms, potential strategies, and current challenges. Bone Res 2023; 11:65. [PMID: 38123549 PMCID: PMC10733346 DOI: 10.1038/s41413-023-00302-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
The skeleton is a highly innervated organ in which nerve fibers interact with various skeletal cells. Peripheral nerve endings release neurogenic factors and sense skeletal signals, which mediate bone metabolism and skeletal pain. In recent years, bone tissue engineering has increasingly focused on the effects of the nervous system on bone regeneration. Simultaneous regeneration of bone and nerves through the use of materials or by the enhancement of endogenous neurogenic repair signals has been proven to promote functional bone regeneration. Additionally, emerging information on the mechanisms of skeletal interoception and the central nervous system regulation of bone homeostasis provide an opportunity for advancing biomaterials. However, comprehensive reviews of this topic are lacking. Therefore, this review provides an overview of the relationship between nerves and bone regeneration, focusing on tissue engineering applications. We discuss novel regulatory mechanisms and explore innovative approaches based on nerve-bone interactions for bone regeneration. Finally, the challenges and future prospects of this field are briefly discussed.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bing Ye
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Siyue Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lian Zeng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongwei Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yizhou Wan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qing Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaifang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bin Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
35
|
Qian M, Li S, Xi K, Tang J, Shen X, Liu Y, Guo R, Zhang N, Gu Y, Xu Y, Cui W, Chen L. ECM-engineered electrospun fibers with an immune cascade effect for inhibiting tissue fibrosis. Acta Biomater 2023; 171:308-326. [PMID: 37673231 DOI: 10.1016/j.actbio.2023.08.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
Tissue regeneration/fibrosis after injury is intricately regulated by the immune cascade reaction and extracellular matrix (ECM). Dysregulated cascade signal could jeopardize tissue homeostasis leading to fibrosis. Bioactive scaffolds mimicking natural ECM microstructure and chemistry could regulate the cascade reaction to achieve tissue regeneration. The current study constructed an ECM-engineered micro/nanofibrous scaffold using self-assembled nanofibrous collagen and decorin (DCN)-loaded microfibers to regulate the immune cascade reaction. The ECM-engineered scaffold promoted anti-inflammatory and pro-regenerative effects, M2 polarization of macrophages, by nanofibrous collagen. The ECM-engineered scaffold could release DCN to inhibit inflammation-associated fibrous angiogenesis. Yet, to prevent excessive M2 activity leading to tissue fibrosis, controlled release of DCN was expected to elicit M1 activity and achieve M1/M2 balance in the repair process. Regulated cascade reaction guided favorable crosstalk between macrophages, endothelial cells and fibroblasts by proximity. Additionally, decorin could also antagonize TGF-β1 via TGF-β/Smad3 pathway to suppress fibrotic activity of fibroblasts. Hence, ECM-engineered scaffolds could exert effective regulation of the immune cascade reaction by microstructure and DCN release and achieve the balance between tissue fibrosis and regeneration. STATEMENT OF SIGNIFICANCE: With the incidence of up to 74.6%, failed back surgery syndrome (FBSS) has been a lingering issue in spine surgery, which poses a heavy socio-economic burden to society. Epidural fibrosis is believed to be responsible for the onset of FBSS. Current biomaterial-based strategies treating epidural fibrosis mainly rely on physical barriers and unidirectional suppression of inflammation. Regulation of the immune cascade reaction for inhibiting fibrosis has not been widely studied. Based on the simultaneous regulation of M1/M2 polarization and intercellular crosstalk, the ECM-engineered micro/nanofibrous scaffolds constructed in the current study could exert an immune cascade effect to coordinate tissue regeneration and inhibit fibrosis. This finding makes a significant contribution in the development of a treatment for epidural fibrosis and FBSS.
Collapse
Affiliation(s)
- Ming Qian
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006 PR China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China
| | - Kun Xi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006 PR China
| | - Jincheng Tang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006 PR China
| | - Xiaofeng Shen
- Department of Orthopaedic Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 889 Wuzhong West Road, Suzhou, Jiangsu 215006, PR China
| | - Yong Liu
- Department of Orthopaedic Surgery, Affiliated Jiangyin Hospital of Nantong University, Jiangyin, Jiangsu 215600, PR China
| | - Ran Guo
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China
| | - Nannan Zhang
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China
| | - Yong Gu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006 PR China.
| | - Yun Xu
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China.
| | - Wenguo Cui
- 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, PR China.
| | - Liang Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006 PR China.
| |
Collapse
|
36
|
Liu W, Zhang K, Nan J, Lei P, Sun Y, Hu Y. Nano artificial periosteum PCL/Ta/ZnO accelerates repair of periosteum via antibacterial, promoting vascularization and osteogenesis. BIOMATERIALS ADVANCES 2023; 154:213624. [PMID: 37716333 DOI: 10.1016/j.bioadv.2023.213624] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/31/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
The periosteum plays a critical role in bone development, shaping, remodeling, and fracture healing due to its abundance of osteoprogenitor cells, osteoblasts, and capillary network. However, the role of periosteum in bone injury healing has been underestimated, thus there is an urgent need to develop a multifunctional artificial periosteum that mimics the natural one. To tackle this issue, electrospinning technology was employed to fabricate an artificial periosteum composed of Poly-ε-caprolactone (PCL) doped with tantalum (Ta) and zinc oxide (ZnO) nanoparticles to enhance its antibacterial, osteogenic, and angiogenic properties. The in vitro cell experiments have demonstrated that the PCL/Ta/ZnO artificial periosteum exhibits excellent biocompatibility and can effectively facilitate osteogenic differentiation of BMSCs as well as angiogenic differentiation of EPCs. Antibacterial experiments have demonstrated the excellent bactericidal effects of PCL/Ta/ZnO artificial periosteum against both S. aureus and E. coli. The subcutaneous infection and critical-sized skull bone defect models have validated its in vivo properties of antibacterial activity, promotion of osteogenesis, and angiogenic potential. The PCL/Ta/ZnO artificial periosteum demonstrates remarkable efficacy in infection control and favorable immunomodulation, thereby achieving rapid vascularized bone repair. In conclusion, the utilization of PCL/Ta/ZnO tissue-engineered periosteum has been demonstrated to exhibit antibacterial properties, pro-vascularization effects, and promotion of osteogenesis at the site of bone defects. This promising approach could potentially offer effective treatment for bone defects.
Collapse
Affiliation(s)
- Wenbin Liu
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha 410008, China
| | - Kai Zhang
- Department of Orthopedic Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiangyu Nan
- Department of Orthopedic Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Lei
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China; Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China.
| | - Yan Sun
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China; Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China.
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China; Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China.
| |
Collapse
|
37
|
Liao X, Shen M, Li T, Feng L, Lin Z, Shi G, Pei G, Cai X. Combined Molybdenum Gelatine Methacrylate Injectable Nano-Hydrogel Effective Against Diabetic Bone Regeneration. Int J Nanomedicine 2023; 18:5925-5942. [PMID: 37881608 PMCID: PMC10596232 DOI: 10.2147/ijn.s428429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/08/2023] [Indexed: 10/27/2023] Open
Abstract
Introduction Bone defects in diabetes mellitus (DM) remain a major challenge for clinical treatment. Fluctuating glucose levels in DM patients lead to excessive production of reactive oxygen species (ROS), which disrupt bone repair homeostasis. Bone filler materials have been widely used in the clinical treatment of DM-related bone defects, but overall they lack efficacy in improving the bone microenvironment and inducing osteogenesis. We utilized a gelatine methacrylate (GelMA) hydrogel with excellent biological properties in combination with molybdenum (Mo)-based polyoxometalate nanoclusters (POM) to scavenge ROS and promote osteoblast proliferation and osteogenic differentiation through the slow-release effect of POM, providing a feasible strategy for the application of biologically useful bone fillers in bone regeneration. Methods We synthesized an injectable hydrogel by gelatine methacrylate (GelMA) and POM. The antioxidant capacity and biological properties of the synthesized GelMA/POM hydrogel were tested. Results In vitro, studies showed that hydrogels can inhibit excessive reactive oxygen species (ROS) and reduce oxidative stress in cells through the beneficial effects of pH-sensitive POM. Osteogenic differentiation assays showed that GelMA/POM had good osteogenic properties with upregulated expression of osteogenic genes (BMP2, RUNX2, Osterix, ALP). Furthermore, RNA-sequencing revealed that activation of the PI3K/Akt signalling pathway in MC3T3-E1 cells with GelMA/POM may be a potential mechanism to promote osteogenesis. In an in vivo study, radiological and histological analyses showed enhanced bone regeneration in diabetic mice, after the application of GelMA/POM. Conclusion In summary, GelMA/POM hydrogels can enhance bone regeneration by directly scavenging ROS and activating the PI3K/Akt signalling pathway.
Collapse
Affiliation(s)
- Xun Liao
- Department of Orthopedics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People’s Republic of China
| | - Mingkui Shen
- Henan Provincial Third People’s Hospital, Zhengzhou, Henan Province, 450000, People’s Republic of China
| | - Tengbo Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, 519000, People’s Republic of China
| | - Li Feng
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, 519000, People’s Republic of China
| | - Zhao Lin
- Department of Orthopedics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People’s Republic of China
| | - Guang Shi
- Department of Orthopedics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People’s Republic of China
| | - Guoxian Pei
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, 519000, People’s Republic of China
| | - Xiyu Cai
- Department of Orthopedics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People’s Republic of China
| |
Collapse
|
38
|
Zhou H, Ye S, Xu M, Hao L, Chen J, Fang Z, Guo K, Chen Y, Wang L. Dynamic surface adapts to multiple service stages by orchestrating responsive polymers and functional peptides. Biomaterials 2023; 301:122200. [PMID: 37423184 DOI: 10.1016/j.biomaterials.2023.122200] [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: 10/26/2022] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
Control over the implant surface functions is highly desirable to enhance tissue healing outcomes but has remained unexplored to adapt to the different service stages. In the present study, we develop a smart titanium surface by orchestrating thermoresponsive polymer and antimicrobial peptide to enable dynamic adaptation to the implantation stage, normal physiological stage and bacterial infection stage. The optimized surface inhibited bacterial adhesion and biofilm formation during surgical implantation, while promoted osteogenesis in the physiological stage. The further temperature increase driven by bacterial infection induced polymer chain collapse to expose antimicrobial peptides by rupturing bacterial membranes, as well as protect the adhered cells from the hostile environment of infection and abnormal temperature. The engineered surface could inhibit infection and promote tissue healing in rabbit subcutaneous and bone defect infection models. This strategy enables the possibility to create a versatile surface platform to balance bacteria/cell-biomaterial interactions at different service stages of implants that has not been achieved before.
Collapse
Affiliation(s)
- Haiyan Zhou
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Silin Ye
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Mingjian Xu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Lihui Hao
- Department of Stomatology, Xingtai Medical College, Xingtai 054000, China
| | - Junjian Chen
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China.
| | - Zhou Fang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Kunzhong Guo
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yunhua Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China.
| | - Lin Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
39
|
Long J, Yao Z, Zhang W, Liu B, Chen K, Li L, Teng B, Du X, Li C, Yu X, Qin L, Lai Y. Regulation of Osteoimmune Microenvironment and Osteogenesis by 3D-Printed PLAG/black Phosphorus Scaffolds for Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302539. [PMID: 37616380 PMCID: PMC10558667 DOI: 10.1002/advs.202302539] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/07/2023] [Indexed: 08/26/2023]
Abstract
The treatment of bone defects remains a significant challenge to be solved clinically. Immunomodulatory properties of orthopedic biomaterials have significance in regulating osteoimmune microenvironment for osteogenesis. A lactic acid-co-glycolic acid (PLGA) scaffold incorporates black phosphorus (BP) fabricated by 3D printing technology to investigate the effect of BP on osteoimmunomodulation and osteogenesis in site. The PLGA/BP scaffold exhibits suitable biocompatibility, biodegradability, and mechanical properties as an excellent microenvironment to support new bone formation. The studies' result also demonstrate that the PLGA/BP scaffolds are able to recruit and stimulate macrophages M2 polarization, inhibit inflammation, and promote human bone marrow mesenchymal stem cells (hBMSCs) proliferation and differentiation, which in turn promotes bone regeneration in the distal femoral defect region of steroid-associated osteonecrosis (SAON) rat model. Moreover, it is screened and demonstrated that PLGA/BP scaffolds can promote osteogenic differentiation by transcriptomic analysis, and PLGA/BP scaffolds promote osteogenic differentiation and mineralization by activating PI3K-AKT signaling pathway in hBMSC cells. In this study, it is shown that the innovative PLGA/BP scaffolds are extremely effective in stimulating bone regeneration by regulating macrophage M2 polarization and a new strategy for the development of biomaterials that can be used to repair bone defects is offered.
Collapse
Affiliation(s)
- Jing Long
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Zhenyu Yao
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Wei Zhang
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Ben Liu
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Kaiming Chen
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Long Li
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Bin Teng
- Center for Energy Metabolism and ReproductionShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Xiang‐Fu Du
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Cairong Li
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Xue‐Feng Yu
- Materials and Interfaces CenterShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Ling Qin
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- Musculoskeletal Research LaboratoryDepartment of Orthopaedics & TraumatologyThe Chinese University of Hong KongHKHong Kong SAR999077P. R. China
- CAS‐HK Joint Lab of BiomaterialsShenzhen518055P. R. China
| | - Yuxiao Lai
- Centre for Translational Medicine Research & DevelopmentShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- CAS‐HK Joint Lab of BiomaterialsShenzhen518055P. R. China
- Guangdong Engineering Laboratory of Biomaterials Additive ManufacturingShenzhen518055P. R. China
- Orthopaedics/Department of Spine Surgerythe First Affiliated Hospital, Shenzhen University, Shenzhen Second People’s HospitalShenzhen518035P. R. China
| |
Collapse
|
40
|
Tang H, Sun W, Liu X, Gao Q, Chen Y, Xie C, Lin W, Chen J, Wang L, Fan Z, Zhang L, Ren Y, She Y, He Y, Chen C. A bioengineered trachea-like structure improves survival in a rabbit tracheal defect model. Sci Transl Med 2023; 15:eabo4272. [PMID: 37729433 DOI: 10.1126/scitranslmed.abo4272] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
A practical strategy for engineering a trachea-like structure that could be used to repair or replace a damaged or injured trachea is an unmet need. Here, we fabricated bioengineered cartilage (BC) rings from three-dimensionally printed fibers of poly(ɛ-caprolactone) (PCL) and rabbit chondrocytes. The extracellular matrix (ECM) secreted by the chondrocytes combined with the PCL fibers formed a "concrete-rebar structure," with ECM deposited along the PCL fibers, forming a grid similar to that of native cartilage. PCL fiber-hydrogel rings were then fabricated and alternately stacked with BC rings on silicone tubes. This trachea-like structure underwent vascularization after heterotopic transplantation into rabbits for 4 weeks. The vascularized bioengineered trachea-like structure was then orthotopically transplanted by end-to-end anastomosis to native rabbit trachea after a segment of trachea had been resected. The bioengineered trachea-like structure displayed mechanical properties similar to native rabbit trachea and transmural angiogenesis between the rings. The 8-week survival rate in transplanted rabbits was 83.3%, and the respiratory rate of these animals was similar to preoperative levels. This bioengineered trachea-like structure may have potential for treating tracheal stenosis and other tracheal injuries.
Collapse
Affiliation(s)
- Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Qing Gao
- 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, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Chaoqi Xie
- 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, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Ziwen Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, 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, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| |
Collapse
|
41
|
Wang P, Gong Y, Zhou G, Ren W, Wang X. Biodegradable Implants for Internal Fixation of Fractures and Accelerated Bone Regeneration. ACS OMEGA 2023; 8:27920-27931. [PMID: 37576626 PMCID: PMC10413843 DOI: 10.1021/acsomega.3c02727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023]
Abstract
Bone fractures have always been a burden to patients due to their common occurrence and severe complications. Traditionally, operative treatments have been widely used in the clinic for implanting, despite the fact that they can only achieve bone fixation with limited stability and pose no effect on promoting tissue growth. In addition, the nondegradable implants usually need a secondary surgery for implant removal, otherwise they may block the regeneration of bones resulting in bone nonunion. To overcome the low degradability of implants and avoid multiple surgeries, tissue engineers have investigated various biodegradable materials for bone regeneration, whereas the significance of stability of long-term bone fixation tends to be neglected during this process. Combining the traditional orthopedic implantation surgeries and emerging tissue engineering, we believe that both bone fixation and bone regeneration are indispensable factors for a successful bone repair. Herein, we define such a novel idea as bone regenerative fixation (BRF), which should be the main future development trend of biodegradable materials.
Collapse
Affiliation(s)
- Pei Wang
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yan Gong
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Guangdong Zhou
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Institute
of Regenerative Medicine and Orthopedics, Institutes of Health Central
Plain, Xinxiang Medical University, Henan 453003, China
| | - Wenjie Ren
- Institute
of Regenerative Medicine and Orthopedics, Institutes of Health Central
Plain, Xinxiang Medical University, Henan 453003, China
| | - Xiansong Wang
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Institute
of Regenerative Medicine and Orthopedics, Institutes of Health Central
Plain, Xinxiang Medical University, Henan 453003, China
| |
Collapse
|
42
|
Ghosh A, Orasugh JT, Ray SS, Chattopadhyay D. Integration of 3D Printing-Coelectrospinning: Concept Shifting in Biomedical Applications. ACS OMEGA 2023; 8:28002-28025. [PMID: 37576662 PMCID: PMC10413848 DOI: 10.1021/acsomega.3c03920] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 07/06/2023] [Indexed: 08/15/2023]
Abstract
Porous structures with sizes between the submicrometer and nanometer scales can be produced using efficient and adaptable electrospinning technology. However, to approximate desirable structures, the construction lacks mechanical sophistication and conformance and requires three-dimensional solitary or multifunctional structures. The diversity of high-performance polymers and blends has enabled the creation of several porous structural conformations for applications in advanced materials science, particularly in biomedicine. Two promising technologies can be combined, such as electrospinning with 3D printing or additive manufacturing, thereby providing a straightforward yet flexible technique for digitally controlled shape-morphing fabrication. The hierarchical integration of configurations is used to imprint complex shapes and patterns onto mesostructured, stimulus-responsive electrospun fabrics. This technique controls the internal stresses caused by the swelling/contraction mismatch in the in-plane and interlayer regions, which, in turn, controls the morphological characteristics of the electrospun membranes. Major innovations in 3D printing, along with additive manufacturing, have led to the production of materials and scaffold systems for tactile and wearable sensors, filtration structures, sensors for structural health monitoring, tissue engineering, biomedical scaffolds, and optical patterning. This review discusses the synergy between 3D printing and electrospinning as a constituent of specific microfabrication methods for quick structural prototypes that are expected to advance into next-generation constructs. Furthermore, individual techniques, their process parameters, and how the fabricated novel structures are applied holistically in the biomedical field have never been discussed in the literature. In summary, this review offers novel insights into the use of electrospinning and 3D printing as well as their integration for cutting-edge applications in the biomedical field.
Collapse
Affiliation(s)
- Adrija Ghosh
- Department
of Polymer Science and Technology, University
of Calcutta, Kolkata 700009, India
| | - Jonathan Tersur Orasugh
- Centre
for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology
Innovation Centre, Council for Scientific
and Industrial Research, Pretoria 0001, South Africa
- Department
of Chemical Sciences, University of Johannesburg, Doorfontein, Johannesburg 2028, South Africa
| | - Suprakas Sinha Ray
- Centre
for Nanostructures and Advanced Materials, DSI-CSIR Nanotechnology
Innovation Centre, Council for Scientific
and Industrial Research, Pretoria 0001, South Africa
- Department
of Chemical Sciences, University of Johannesburg, Doorfontein, Johannesburg 2028, South Africa
| | - Dipankar Chattopadhyay
- Department
of Polymer Science and Technology, University
of Calcutta, Kolkata 700009, India
- Center
for Research in Nanoscience and Nanotechnology, Acharya Prafulla Chandra
Roy Sikhsha Prangan, University of Calcutta, JD-2, Sector-III, Saltlake City, Kolkata 700098, India
| |
Collapse
|
43
|
Wu P, Shen L, Liu HF, Zou XH, Zhao J, Huang Y, Zhu YF, Li ZY, Xu C, Luo LH, Luo ZQ, Wu MH, Cai L, Li XK, Wang ZG. The marriage of immunomodulatory, angiogenic, and osteogenic capabilities in a piezoelectric hydrogel tissue engineering scaffold for military medicine. Mil Med Res 2023; 10:35. [PMID: 37525300 PMCID: PMC10388535 DOI: 10.1186/s40779-023-00469-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Most bone-related injuries to grassroots troops are caused by training or accidental injuries. To establish preventive measures to reduce all kinds of trauma and improve the combat effectiveness of grassroots troops, it is imperative to develop new strategies and scaffolds to promote bone regeneration. METHODS In this study, a porous piezoelectric hydrogel bone scaffold was fabricated by incorporating polydopamine (PDA)-modified ceramic hydroxyapatite (PDA-hydroxyapatite, PHA) and PDA-modified barium titanate (PDA-BaTiO3, PBT) nanoparticles into a chitosan/gelatin (Cs/Gel) matrix. The physical and chemical properties of the Cs/Gel/PHA scaffold with 0-10 wt% PBT were analyzed. Cell and animal experiments were performed to characterize the immunomodulatory, angiogenic, and osteogenic capabilities of the piezoelectric hydrogel scaffold in vitro and in vivo. RESULTS The incorporation of BaTiO3 into the scaffold improved its mechanical properties and increased self-generated electricity. Due to their endogenous piezoelectric stimulation and bioactive constituents, the as-prepared Cs/Gel/PHA/PBT hydrogels exhibited cytocompatibility as well as immunomodulatory, angiogenic, and osteogenic capabilities; they not only effectively induced macrophage polarization to M2 phenotype but also promoted the migration, tube formation, and angiogenic differentiation of human umbilical vein endothelial cells (HUVECs) and facilitated the migration, osteo-differentiation, and extracellular matrix (ECM) mineralization of MC3T3-E1 cells. The in vivo evaluations showed that these piezoelectric hydrogels with versatile capabilities significantly facilitated new bone formation in a rat large-sized cranial injury model. The underlying molecular mechanism can be partly attributed to the immunomodulation of the Cs/Gel/PHA/PBT hydrogels as shown via transcriptome sequencing analysis, and the PI3K/Akt signaling axis plays an important role in regulating macrophage M2 polarization. CONCLUSION The piezoelectric Cs/Gel/PHA/PBT hydrogels developed here with favorable immunomodulation, angiogenesis, and osteogenesis functions may be used as a substitute in periosteum injuries, thereby offering the novel strategy of applying piezoelectric stimulation in bone tissue engineering for the enhancement of combat effectiveness in grassroots troops.
Collapse
Affiliation(s)
- Ping Wu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Hui-Fan Liu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiang-Hui Zou
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Juan Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Yu Huang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Yu-Fan Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhao-Yu Li
- Department of Overseas Education College, Jimei University, Xiamen, 361021, Fujian, China
| | - Chao Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Li-Hua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhi-Qiang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Min-Hao Wu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Xiao-Kun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Zhou-Guang Wang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
44
|
Shokrani H, Shokrani A, Seidi F, Mashayekhi M, Kar S, Nedeljkovic D, Kuang T, Saeb MR, Mozafari M. Polysaccharide-based biomaterials in a journey from 3D to 4D printing. Bioeng Transl Med 2023; 8:e10503. [PMID: 37476065 PMCID: PMC10354780 DOI: 10.1002/btm2.10503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/31/2023] [Accepted: 02/18/2023] [Indexed: 07/22/2023] Open
Abstract
3D printing is a state-of-the-art technology for the fabrication of biomaterials with myriad applications in translational medicine. After stimuli-responsive properties were introduced to 3D printing (known as 4D printing), intelligent biomaterials with shape configuration time-dependent character have been developed. Polysaccharides are biodegradable polymers sensitive to several physical, chemical, and biological stimuli, suited for 3D and 4D printing. On the other hand, engineering of mechanical strength and printability of polysaccharide-based scaffolds along with their aneural, avascular, and poor metabolic characteristics need to be optimized varying printing parameters. Multiple disciplines such as biomedicine, chemistry, materials, and computer sciences should be integrated to achieve multipurpose printable biomaterials. In this work, 3D and 4D printing technologies are briefly compared, summarizing the literature on biomaterials engineering though printing techniques, and highlighting different challenges associated with 3D/4D printing, as well as the role of polysaccharides in the technological shift from 3D to 4D printing for translational medicine.
Collapse
Affiliation(s)
- Hanieh Shokrani
- Jiangsu Co‐Innovation Center for Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and MaterialsNanjing Forestry UniversityNanjingChina
- Department of Chemical EngineeringSharif University of TechnologyTehranIran
| | | | - Farzad Seidi
- Jiangsu Co‐Innovation Center for Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and MaterialsNanjing Forestry UniversityNanjingChina
| | | | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle EastKuwait
| | - Dragutin Nedeljkovic
- College of Engineering and Technology, American University of the Middle EastKuwait
| | - Tairong Kuang
- College of Material Science and Engineering, Zhejiang University of TechnologyHangzhouChina
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
45
|
Elahpour N, Niesner I, Bossard C, Abdellaoui N, Montouillout V, Fayon F, Taviot-Guého C, Frankenbach T, Crispin A, Khosravani P, Holzapfel BM, Jallot E, Mayer-Wagner S, Lao J. Zinc-Doped Bioactive Glass/Polycaprolactone Hybrid Scaffolds Manufactured by Direct and Indirect 3D Printing Methods for Bone Regeneration. Cells 2023; 12:1759. [PMID: 37443794 PMCID: PMC10341101 DOI: 10.3390/cells12131759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
A novel organic-inorganic hybrid, based on SiO2-CaO-ZnO bioactive glass (BG) and polycaprolactone (PCL), associating the highly bioactive and versatile bioactive glass with clinically established PCL was examined. The BG-PCL hybrid is obtained by acid-catalyzed silica sol-gel process inside PCL solution either by direct or indirect printing. Apatite-formation tests in simulated body fluid (SBF) confirm the ion release along with the hybrid's bone-like apatite forming. Kinetics differ significantly between directly and indirectly printed scaffolds, the former requiring longer periods to degrade, while the latter demonstrates faster calcium phosphate (CaP) formation. Remarkably, Zn diffusion and accumulation are observed at the surface within the newly formed active CaP layer. Zn release is found to be dependent on printing method and immersion medium. Investigation of BG at the atomic scale reveals the ambivalent role of Zn, capable of acting both as a network modifier and as a network former linking the BG silicate network. In addition, hMSCs viability assay proves no cytotoxicity of the Zn hybrid. LIVE/DEAD staining demonstrated excellent cell viability and proliferation for over seven weeks. Overall, this hybrid material either non-doped or doped with a metal trace element is a promising candidate to be translated to clinical applications for bone regeneration.
Collapse
Affiliation(s)
- Nafise Elahpour
- Laboratoire de Physique de Clermont (LPC), Université Clermont Auvergne, CNRS/IN2P3, F-63000 Clermont-Ferrand, France; (N.E.)
| | - Isabella Niesner
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Cédric Bossard
- Laboratoire de Physique de Clermont (LPC), Université Clermont Auvergne, CNRS/IN2P3, F-63000 Clermont-Ferrand, France; (N.E.)
| | - Nora Abdellaoui
- Laboratoire de Physique de Clermont (LPC), Université Clermont Auvergne, CNRS/IN2P3, F-63000 Clermont-Ferrand, France; (N.E.)
| | - Valérie Montouillout
- Conditions Extrêmes et Matériaux: Haute Température et Irradiation (CEMHTI), CNRS-UPR3079, Université Orléans, F-45071 Orléans, France
| | - Franck Fayon
- Conditions Extrêmes et Matériaux: Haute Température et Irradiation (CEMHTI), CNRS-UPR3079, Université Orléans, F-45071 Orléans, France
| | - Christine Taviot-Guého
- Institut de Chimie de Clermont-Ferrand, Université Clermont Auvergne, CNRS/UMR 6296, F-63000 Clermont-Ferrand, France
| | - Tina Frankenbach
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Alexander Crispin
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Pardis Khosravani
- Flow Cytometry Core Facility, Biomedical Center, Ludwig-Maximilians-Universität München, 82152 Planegg, Germany
| | - Boris Michael Holzapfel
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Edouard Jallot
- Laboratoire de Physique de Clermont (LPC), Université Clermont Auvergne, CNRS/IN2P3, F-63000 Clermont-Ferrand, France; (N.E.)
| | - Susanne Mayer-Wagner
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Jonathan Lao
- Laboratoire de Physique de Clermont (LPC), Université Clermont Auvergne, CNRS/IN2P3, F-63000 Clermont-Ferrand, France; (N.E.)
| |
Collapse
|
46
|
Zhou P, Yan B, Wei B, Fu L, Wang Y, Wang W, Zhang L, Mao Y. Quercetin-solid lipid nanoparticle-embedded hyaluronic acid functionalized hydrogel for immunomodulation to promote bone reconstruction. Regen Biomater 2023; 10:rbad025. [PMID: 37077623 PMCID: PMC10110271 DOI: 10.1093/rb/rbad025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 04/21/2023] Open
Abstract
Bone defects are a persistent challenge in clinical practice. Although repair therapies based on tissue-engineered materials, which are known to have a crucial role in defective bone regeneration, have gathered increased attention, the current treatments for massive bone defects have several limitations. In the present study, based on the immunomodulatory inflammatory microenvironment properties of quercetin, we encapsulated quercetin-solid lipid nanoparticles (SLNs) in a hydrogel. Temperature-responsive poly(ε-caprolactone-co-lactide)-b-poly(ethylene glycol)-b-poly(ε-caprolactone-co-lactide) modifications were coupled to the main chain of hyaluronic acid hydrogel, constructing a novel, injectable bone immunomodulatory hydrogel scaffold. Extensive in vitro and in vivo data showed that this bone immunomodulatory scaffold forms an anti-inflammatory microenvironment by decreasing M1 polarization, while elevating the M2 polarization. Synergistic effects on angiogenesis and anti-osteoclastic differentiation were observed. These findings further proved that administering quercetin SLNs encapsulated in a hydrogel can aid bone defect reconstruction in rats, providing new insights for large-scale bone defect repair.
Collapse
Affiliation(s)
- Pinghui Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Bomin Yan
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Bangguo Wei
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Liangmin Fu
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Ying Wang
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Wenrui Wang
- School of Life Science, Bengbu Medical College, Bengbu 233030, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui 233030, China
| | - Li Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Yingji Mao
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- School of Life Science, Bengbu Medical College, Bengbu 233030, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
47
|
Wang X, Zhu X, Wang D, Li X, Wang J, Yin G, Huang Z, Pu X. Identification of a Specific Phage as Growth Factor Alternative Promoting the Recruitment and Differentiation of MSCs in Bone Tissue Regeneration. ACS Biomater Sci Eng 2023; 9:2426-2437. [PMID: 37023478 DOI: 10.1021/acsbiomaterials.2c01538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Inefficient use and loss of exogenously implanted mesenchymal stem cells (MSCs) are major concerns in MSCs-based bone tissue engineering. It is a promising approach to overcome the above issues by recruiting and regulation of endogenous MSCs. However, there are few substances that can recruit MSCs effectively and specifically to the site of bone injury. In this study, we identified a phage clone (termed P11) with specific affinity for MSCs through phage display biopanning, and further investigated the effects of P11 on the cytological behavior of MSCs and macrophages. The results showed that P11 could bind MSCs specifically and promote the proliferation and migration of MSCs. Meanwhile, P11 could polarize macrophages to the M1 phenotype and significantly changed their morphology, which further enhanced the chemotaxis of MSCs. Additionally, RNA-seq results revealed that P11 could promote the secretion of osteogenesis-related markers in MSCs through the TPL2-MEK-ERK signaling pathway. Altogether, P11 has great potential to be used as growth factor alternatives in bone tissue engineering, with the advantages of cheaper and stable activity. Our study also advances the understanding of the effects of phages on macrophages and MSCs, and provides a new idea for the development in the field of phage-based tissue engineering.
Collapse
Affiliation(s)
- Xingming Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiupeng Zhu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Danni Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaoxu Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
48
|
Zhang QY, Tan J, Nie R, Song YT, Zhou XL, Feng ZY, Huang K, Zou CY, Yuan QJ, Zhao LM, Zhang XZ, Jiang YL, Liu LM, Li-Ling J, Xie HQ. Acceleration of wound healing by composite small intestinal submucosa hydrogels through immunomodulation. COMPOSITES PART B: ENGINEERING 2023; 254:110550. [DOI: 10.1016/j.compositesb.2023.110550] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
|
49
|
Zhang QY, Tan J, Huang K, Nie R, Feng ZY, Zou CY, Li QJ, Chen J, Sheng N, Qin BQ, Gu ZP, Liu LM, Xie HQ. Polyphenolic-modified cellulose acetate membrane for bone regeneration through immunomodulation. Carbohydr Polym 2023; 305:120546. [PMID: 36737196 DOI: 10.1016/j.carbpol.2023.120546] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/27/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023]
Abstract
To enhance the bioactivity of cellulosic derivatives has become an important strategy to promote their value for clinical applications. Herein, protocatechualdehyde (PCA), a polyphenolic molecule, was used to modify a cellulose acetate (CA) membrane by combining with metal ions to confer an immunomodulatory activity. The PCA-modified CA membrane has shown a significant radical scavenging activity, thereby suppressed the inflammatory response and created a favorable immune microenvironment for osteogenesis and mineralization. Moreover, addition of metal ions could further stimulate the osteogenic differentiation of stem cells and accelerate bone regeneration both in vitro and in vivo. This study may provide a strategy to promote the immunomodulatory activity of cellulose-based biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jie Tan
- Department of Spine Surgery, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, PR China
| | - Kai Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Rong Nie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zi-Yuan Feng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Chen-Yu Zou
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qian-Jin Li
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jun Chen
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ning Sheng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Bo-Quan Qin
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhi-Peng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China
| | - Li-Min Liu
- Department of Orthopedics Surgery, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
50
|
Dong C, Tan G, Zhang G, Lin W, Wang G. The function of immunomodulation and biomaterials for scaffold in the process of bone defect repair: A review. Front Bioeng Biotechnol 2023; 11:1133995. [PMID: 37064239 PMCID: PMC10090379 DOI: 10.3389/fbioe.2023.1133995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
The process of bone regeneration involves the interaction of the skeletal, blood, and immune systems. Bone provides a solid barrier for the origin and development of immune cells in the bone marrow. At the same time, immune cells secrete related factors to feedback on the remodeling of the skeletal system. Pathological or traumatic injury of bone tissue involves changes in blood supply, cell behavior, and cytokine expression. Immune cells and their factors play an essential role in repairing foreign bodies in bone injury or implantation of biomaterials, the clearance of dead cells, and the regeneration of bone tissue. This article reviews the bone regeneration application of the bone tissue repair microenvironment in bone cells and immune cells in the bone marrow and the interaction of materials and immune cells.
Collapse
Affiliation(s)
- Changchao Dong
- Trauma Medical Center, Department of Orthopedics Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Orthopedics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gang Tan
- Department of Orthopedics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guangyan Zhang
- Department of Respiratory Medicine, The 7th Hospital of Chengdu, Chengdu, Sichuan, China
| | - Wei Lin
- Department of Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Wei Lin, ; Guanglin Wang,
| | - Guanglin Wang
- Trauma Medical Center, Department of Orthopedics Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Orthopedics, West China Hospital, Orthopedics Research Institute, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Wei Lin, ; Guanglin Wang,
| |
Collapse
|