1
|
Kibritoglu E, Yuksel H. Numerical analysis of coil designs to expedite fracture healing using dielectrophoresis with S method. Comput Biol Med 2025; 192:110213. [PMID: 40279972 DOI: 10.1016/j.compbiomed.2025.110213] [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: 10/21/2024] [Revised: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Classical methods for speeding up fracture healing usually rely on direct electrical stimulation and electromagnetic fields to boost the levels of growth factors at the fracture site. However, these techniques often concentrate on bone cells themselves rather than addressing the critical blood flow dynamics necessary for effective healing. This study introduces a mathematical model designed to explore the potential of dielectrophoretic forces (DEPFs) in improving blood flow at the fracture site. By adjusting blood flow, the model seeks to enhance the delivery of vital nutrients, hormones, and growth factors, including endothelial cells (ECs), vascular endothelial growth factor (VEGF) and oxygen, which are essential for accelerating the fracture healing process. METHOD The proposed approach includes a new technique, termed the S method, which assesses the non-uniformity of DEPFs by algebraically analyzing the electric field lines associated with positive and negative dielectrophoresis. We developed analytical equations to simulate various coil configurations, focusing on long bone fractures where blood flow is vertically oriented. The DEPF Factor (χDEPF) was used to measure the ratio of blood flow velocity in the presence of DEPFs compared to the absence of DEPFs, thus indicating the effectiveness of DEPF in enhancing blood flow. RESULTS The simulation results revealed that DEPF reaches its peak efficacy at the gamma dispersion band, with the most significant enhancement occurring at a frequency of 15 MHz. Specifically, the average values of χDEPF were 1.8, 3.2, and 7.9 for the catenary, lintearia, and valeria coils, respectively. Our computational model, which incorporated VEGF, ECs, and oxygen tension, demonstrated that the catenary coil slightly improved healing rates in impaired fractures, the lintearia coil normalized healing times between impaired and normal fractures, and the valeria coil not only accelerated healing in impaired fractures but also enhanced healing in normal fractures. CONCLUSIONS This paper's findings suggest that the valeria coil exhibits the best DEPF functionality, making it the optimal configuration for future experimental studies aimed at evaluating the efficacy of DEPF in promoting fracture healing. The ability of DEPFs to significantly enhance blood flow could represent a substantial advancement in the treatment of both normal and impaired fractures.
Collapse
Affiliation(s)
- Erman Kibritoglu
- Department of Electrical and Electronics Engineering, Bogazici University, Bebek, İstanbul, 34342, Turkey
| | - Heba Yuksel
- Department of Electrical and Electronics Engineering, Bogazici University, Bebek, İstanbul, 34342, Turkey.
| |
Collapse
|
2
|
Mansouri Moghaddam M, Jooybar E, Imani R. Injectable microgel and micro-granular hydrogels for bone tissue engineering. Biofabrication 2025; 17:032001. [PMID: 40228520 DOI: 10.1088/1758-5090/adcc58] [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: 01/17/2025] [Accepted: 04/14/2025] [Indexed: 04/16/2025]
Abstract
Injectable microgels, made from both natural and synthetic materials, are promising platforms for the encapsulation of cells or bioactive agents, such as drugs and growth factors, for delivery to injury sites. They can also serve as effective micro-scaffolds in bone tissue engineering (BTE), offering a supportive environment for cell proliferation or differentiation into osteoblasts. Microgels can be injected in the injury sites individually or in the form of aggregated/jammed ones named micro-granular hydrogels. This review focuses on common materials and fabrication techniques for preparing injectable microgels, as well as their characteristics and applications in BTE. These applications include their use as cell carriers, delivery systems for bioactive molecules, micro-granular hydrogels, bio-inks for bioprinting, three-dimensional microarrays, and the formation of microtissues. Furthermore, we discuss the current and potential future applications of microgels in bone tissue regeneration.
Collapse
Affiliation(s)
- Melika Mansouri Moghaddam
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
3
|
Zhou X, Chen S, Pich A, He C. Advanced Bioresponsive Drug Delivery Systems for Promoting Diabetic Vascularized Bone Regeneration. ACS Biomater Sci Eng 2025; 11:182-207. [PMID: 39666445 DOI: 10.1021/acsbiomaterials.4c02037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
The treatment of bone defects in diabetes mellitus (DM) patients remains a major challenge since the diabetic microenvironments significantly impede bone regeneration. Many abnormal factors including hyperglycemia, elevated oxidative stress, increased inflammation, imbalanced osteoimmune, and impaired vascular system in the diabetic microenvironment will result in a high rate of impaired, delayed, or even nonhealing events of bone tissue. Stimuli-responsive biomaterials that can respond to endogenous biochemical signals have emerged as effective therapeutic systems to treat diabetic bone defects via the combination of microenvironmental regulation and enhanced osteogenic capacity. Following the natural bone healing processes, coupling of angiogenesis and osteogenesis by advanced bioresponsive drug delivery systems has proved to be of significant approach for promoting bone repair in DM. In this Review, we have systematically summarized the mechanisms and therapeutic strategies of DM-induced impaired bone healing, outlined the bioresponsive design for drug delivery systems, and highlighted the vascularization strategies for promoting bone regeneration. Accordingly, we then overview the recent advances in developing bioresponsive drug delivery systems to facilitate diabetic vascularized bone regeneration by remodeling the microenvironment and modulating multiple regenerative cues. Furthermore, we discuss the development of adaptable drug delivery systems with unique features for guiding DM-associated bone regeneration in the future.
Collapse
Affiliation(s)
- Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Shuo Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Andrij Pich
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
4
|
Hui Y, Mao J, Rui M, Huang Y, Jiang X, Xu Y, Wang W, Wu J, Zhou L, Xi K, Huang L, Chen L. Hydrogel Microsphere-Encapsulated Bimetallic Nanozyme for Promoting Diabetic Bone Regeneration via Glucose Consumption and ROS Scavenging. Adv Healthc Mater 2024; 13:e2402596. [PMID: 39252661 DOI: 10.1002/adhm.202402596] [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: 07/15/2024] [Revised: 08/24/2024] [Indexed: 09/11/2024]
Abstract
The healing of bone defects among diabetic patients presents a critical challenge due to the pathological microenvironment, characterized by hyperglycemia, excessive reactive oxygen species (ROS) production, and inflammation. Herein, multifunctional composite microspheres, termed GMAP are developed, using a microfluidic technique by incorporating Au@Pt nanoparticles (NPs) and GelMA hydrogel to modulate the diabetic microenvironment for promoting bone regeneration. The GMAP enables the sustained release of Au@Pt NPs, which function as bimetallic nanozymes with dual enzyme-like activities involving glucose oxidase and catalase. The synergistic effect allows for efficient glucose consumption and ROS elimination concurrently. Thus, the GMAP effectively protects the proliferation of bone marrow mesenchymal stem cells (BMSCs) under adverse high-glucose conditions. Furthermore, it also promotes the osteogenic differentiation and paracrine capabilities of BMSCs, and subsequently inhibits inflammation and enhances angiogenesis. In vivo diabetic rats bone defect model, it is demonstrated that GMAP microspheres significantly improve bone regeneration, as verified by micro-computed tomography and histological examinations. This study provides a novel strategy for bone regeneration by modulating the diabetic microenvironment, presenting a promising approach for addressing the complex challenges associated with bone healing in diabetic patients.
Collapse
Affiliation(s)
- Yujian Hui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiangyin, 214400, P. R. China
| | - Jiannan Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiangyin, 214400, P. R. China
| | - Min Rui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopedics, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiangyin, 214400, P. R. China
| | - Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jie Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lixin Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| |
Collapse
|
5
|
Chen R, Jin Y, Lian R, Yang J, Liao Z, Jin Y, Deng Z, Feng S, Feng Z, Wei Y, Zhang Z, Zhao L. CRIP1 regulates osteogenic differentiation of bone marrow stromal cells and pre-osteoblasts via the Wnt signaling pathway. Biochem Biophys Res Commun 2024; 727:150277. [PMID: 38936225 DOI: 10.1016/j.bbrc.2024.150277] [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: 04/12/2024] [Revised: 06/02/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
With the aging of the global demographic, the prevention and treatment of osteoporosis are becoming crucial issues. The gradual loss of self-renewal and osteogenic differentiation capabilities in bone marrow stromal cells (BMSCs) is one of the key factors contributing to osteoporosis. To explore the regulatory mechanisms of BMSCs differentiation, we collected bone marrow cells of femoral heads from patients undergoing total hip arthroplasty for single-cell RNA sequencing analysis. Single-cell RNA sequencing revealed significantly reduced CRIP1 (Cysteine-Rich Intestinal Protein 1) expression and osteogenic capacity in the BMSCs of osteoporosis patients compared to non-osteoporosis group. CRIP1 is a gene that encodes a member of the LIM/double zinc finger protein family, which is involved in the regulation of various cellular processes including cell growth, development, and differentiation. CRIP1 knockdown resulted in decreased alkaline phosphatase activity, mineralization and expression of osteogenic markers, indicating impaired osteogenic differentiation. Conversely, CRIP1 overexpression, both in vitro and in vivo, enhanced osteogenic differentiation and rescued bone mass reduction in ovariectomy-induced osteoporosis mice model. The study further established CRIP1's modulation of osteogenesis through the Wnt signaling pathway, suggesting that targeting CRIP1 could offer a novel approach for osteoporosis treatment by promoting bone formation and preventing bone loss.
Collapse
Affiliation(s)
- Ruge Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yangchen Jin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ru Lian
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jie Yang
- Department of Chinese Medicine, Chinese People's Liberation Army Air Force Special Medical Center, Beijing, 100142, China
| | - Zheting Liao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yu Jin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhonghao Deng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Shuhao Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zihang Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yiran Wei
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Liang Zhao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
6
|
Xiong C, Shang J, Yu Z, Zhang J, Miao K, Yu C, Huang Y, Weng Y, Zhou X. Britanin alleviates chondrocyte ferroptosis in osteoarthritis by regulating the Nrf2-GPX4 axis. ARAB J CHEM 2024; 17:105918. [DOI: 10.1016/j.arabjc.2024.105918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025] Open
|
7
|
Xia Y, Chen Z, Zheng Z, Chen H, Chen Y. Nanomaterial-integrated injectable hydrogels for craniofacial bone reconstruction. J Nanobiotechnology 2024; 22:525. [PMID: 39217329 PMCID: PMC11365286 DOI: 10.1186/s12951-024-02801-0] [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: 06/30/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The complex anatomy and biology of craniofacial bones pose difficulties in their effective and precise reconstruction. Injectable hydrogels (IHs) with water-swollen networks are emerging as a shape-adaptive alternative for noninvasively rebuilding craniofacial bones. The advent of versatile nanomaterials (NMs) customizes IHs with strengthened mechanical properties and therapeutically favorable performance, presenting excellent contenders over traditional substitutes. Structurally, NM-reinforced IHs are energy dissipative and covalently crosslinked, providing the mechanics necessary to support craniofacial structures and physiological functions. Biofunctionally, incorporating unique NMs into IH expands a plethora of biological activities, including immunomodulatory, osteogenic, angiogenic, and antibacterial effects, further favoring controllable dynamic tissue regeneration. Mechanistically, NM-engineered IHs optimize the physical traits to direct cell responses, regulate intracellular signaling pathways, and control the release of biomolecules, collectively bestowing structure-induced features and multifunctionality. By encompassing state-of-the-art advances in NM-integrated IHs, this review offers a foundation for future clinical translation of craniofacial bone reconstruction.
Collapse
Affiliation(s)
- Yong Xia
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zihan Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zebin Zheng
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Huimin Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yuming Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
8
|
Mahmoudi C, Tahraoui Douma N, Mahmoudi H, Iurciuc (Tincu) CE, Popa M. Hydrogels Based on Proteins Cross-Linked with Carbonyl Derivatives of Polysaccharides, with Biomedical Applications. Int J Mol Sci 2024; 25:7839. [PMID: 39063081 PMCID: PMC11277554 DOI: 10.3390/ijms25147839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Adding carbonyl groups into the hydrogel matrix improves the stability and biocompatibility of the hydrogels, making them suitable for different biomedical applications. In this review article, we will discuss the use of hydrogels based on polysaccharides modified by oxidation, with particular attention paid to the introduction of carbonyl groups. These hydrogels have been developed for several applications in tissue engineering, drug delivery, and wound healing. The review article discusses the mechanism by which oxidized polysaccharides can introduce carbonyl groups, leading to the development of hydrogels through cross-linking with proteins. These hydrogels have tunable mechanical properties and improved biocompatibility. Hydrogels have dynamic properties that make them promising biomaterials for various biomedical applications. This paper comprehensively analyzes hydrogels based on cross-linked proteins with carbonyl groups derived from oxidized polysaccharides, including microparticles, nanoparticles, and films. The applications of these hydrogels in tissue engineering, drug delivery, and wound healing are also discussed.
Collapse
Affiliation(s)
- Chahrazed Mahmoudi
- Laboratory of Water and Environment, Faculty of Technology, University Hassiba Benbouali of Chlef, Chlef 02000, Algeria
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
| | - Naïma Tahraoui Douma
- Laboratory of Water and Environment, Faculty of Technology, University Hassiba Benbouali of Chlef, Chlef 02000, Algeria
| | - Hacene Mahmoudi
- National Higher School of Nanosciences and Nanotechnologies, Algiers 16000, Algeria;
| | - Camelia Elena Iurciuc (Tincu)
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, University Street, No. 16, 700115 Iasi, Romania
| | - Marcel Popa
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov, 050044 Bucharest, Romania
| |
Collapse
|
9
|
Sanati M, Amin Yavari S. Liposome-integrated hydrogel hybrids: Promising platforms for cancer therapy and tissue regeneration. J Control Release 2024; 368:703-727. [PMID: 38490373 DOI: 10.1016/j.jconrel.2024.03.008] [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: 10/22/2023] [Revised: 02/10/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Drug delivery platforms have gracefully emerged as an indispensable component of novel cancer chemotherapy, bestowing targeted drug distribution, elevating therapeutic effects, and reducing the burden of unwanted side effects. In this context, hybrid delivery systems artfully harnessing the virtues of liposomes and hydrogels bring remarkable benefits, especially for localized cancer therapy, including intensified stability, excellent amenability to hydrophobic and hydrophilic medications, controlled liberation behavior, and appropriate mucoadhesion to mucopenetration shift. Moreover, three-dimensional biocompatible liposome-integrated hydrogel networks have attracted unprecedented interest in tissue regeneration, given their tunable architecture and physicochemical properties, as well as enhanced mechanical support. This review elucidates and presents cutting-edge developments in recruiting liposome-integrated hydrogel systems for cancer treatment and tissue regeneration.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
10
|
Yao J, Huo Z, Xu J, Shang J, Weng Y, Xu D, Liu T, Huang Y, Zhou X. Enhanced Surface Immunomodification of Engineered Hydrogel Materials through Chondrocyte Modulation for the Treatment of Osteoarthritis. COATINGS 2024; 14:308. [DOI: 10.3390/coatings14030308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Osteoarthritis (OA) is characterized by cartilage degeneration and synovial inflammation, with chondrocytes playing a pivotal role in this disease. However, inflammatory mediators, mechanical stress, and oxidative stress can compromise functionality. The occurrence and progression of OA are intrinsically linked to the immune response. Current research on the treatment of OA mainly concentrates on the synergistic application of drugs and tissue engineering. The surface of engineered hydrogel materials can be immunomodified to affect the function of chondrocytes in drug therapy, gene therapy, and cell therapy. Prior studies have concentrated on the drug-loading function of hydrogels but overlooked the immunomodulatory role of chondrocytes. These modifications can inhibit the proliferation and differentiation of chondrocytes, reduce the inflammatory response, and promote cartilage regeneration. The surface immunomodification of engineered hydrogel materials can significantly enhance their efficacy in the treatment of OA. Thus, immunomodulatory tissue engineering has significant potential for treating osteoarthritis.
Collapse
Affiliation(s)
- Jiapei Yao
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Zhennan Huo
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jie Xu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jingjing Shang
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
- Department of Pharmacy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yiping Weng
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Dongmei Xu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Ting Liu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture 811800, China
| |
Collapse
|
11
|
Lu G, Zhao G, Wang S, Li H, Yu Q, Sun Q, Wang B, Wei L, Fu Z, Zhao Z, Yang L, Deng L, Zheng X, Cai M, Lu M. Injectable Nano-Micro Composites with Anti-bacterial and Osteogenic Capabilities for Minimally Invasive Treatment of Osteomyelitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306964. [PMID: 38234236 DOI: 10.1002/advs.202306964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/17/2023] [Indexed: 01/19/2024]
Abstract
The effective management of osteomyelitis remains extremely challenging due to the difficulty associated with treating bone defects, the high probability of recurrence, the requirement of secondary surgery or multiple surgeries, and the difficulty in eradicating infections caused by methicillin-resistant Staphylococcus aureus (MRSA). Hence, smart biodegradable biomaterials that provide effective and precise local anti-infection effects and can promote the repair of bone defects are actively being developed. Here, a novel nano-micro composite is fabricated by combining calcium phosphate (CaP) nanosheets with drug-loaded GelMA microspheres via microfluidic technology. The microspheres are covalently linked with vancomycin (Van) through an oligonucleotide (oligo) linker using an EDC/NHS carboxyl activator. Accordingly, a smart nano-micro composite called "CaP@MS-Oligo-Van" is synthesized. The porous CaP@MS-Oligo-Van composites can target and capture bacteria. They can also release Van in response to the presence of bacterial micrococcal nuclease and Ca2+, exerting additional antibacterial effects and inhibiting the inflammatory response. Finally, the released CaP nanosheets can promote bone tissue repair. Overall, the findings show that a rapid, targeted drug release system based on CaP@MS-Oligo-Van can effectively target bone tissue infections. Hence, this agent holds potential in the clinical treatment of osteomyelitis caused by MRSA.
Collapse
Affiliation(s)
- Guanghua Lu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Gang Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Shen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hanqing Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Qiang Yu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Li Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Zi Fu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Linshan Yang
- Taikang Bybo Dental, Shanghai, 200001, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Xianyou Zheng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| |
Collapse
|
12
|
Yang Z, Jiang W, Xiong C, Shang J, Huang Y, Zhou X, Zhang S. Calcipotriol suppresses GPX4-mediated ferroptosis in OA chondrocytes by blocking the TGF-β1 pathway. Cytokine 2023; 171:156382. [PMID: 37782985 DOI: 10.1016/j.cyto.2023.156382] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/02/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Globally, tens of millions of individuals experience osteoarthritis (OA), a degenerative joint condition for which a definitive cure is currently lacking. This condition is characterized by joint inflammation and the progressive deterioration of articular cartilage. In this study, western blotting, quantitative reverse-transcription polymerase chain reaction, and immunofluorescence analysis were performed to elucidate the molecular mechanisms by which calcipotriol alleviates chondrocyte ferroptosis. The effect of calcipotriol on reactive oxygen species and lipid peroxidation levels in chondrocytes was assessed using dihydroethidium staining and the fluorescent dye BODIPY. To replicate OA, the destabilized medial meniscus model was employed, followed by the injection of calcipotriol into the knee articular cavity. Morphological analysis was conducted through hematoxylin and eosin staining, safranin O-Fast green staining, and micro-computed tomography analysis. Immunohistochemical analysis was performed to validate the effect of calcipotriol in vivo. Our results demonstrate that the expression of SOX9, col2a1, and Aggrecan, as well as MMP13 and ADAMTS5 protein expression levels, decrease upon treatment with calcipotriol in interleukin-1β stimulated chondrocytes. Despite these promising outcomes, the exact mechanism underlying calcipotriol's therapeutic effect on OA remains uncertain. We discovered that calcipotriol inhibits chondrocyte GPX4-mediated ferroptosis by suppressing the expression of transforming growth factor-β1. Furthermore, our study established an in vivo model of OA using rats with medial meniscus instability. Our experiments on rats with OA revealed that intra-articular calcipotriol injection significantly reduces cartilage degradation caused by the disease. Our findings suggest that calcipotriol can mitigate OA by impeding GPX4-mediated ferroptosis of chondrocytes, achieved through the suppression of the TGF-β1 pathway.
Collapse
Affiliation(s)
- Zhicheng Yang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China
| | - Wei Jiang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui 230022, China
| | - Chenwei Xiong
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China; Department of Orthopedics, Zhangjiajie People's Hospital, Zhangjiajie 427000, China
| | - JingJing Shang
- Department of Pharmacy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China; Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai Province 811800, China.
| | - Su Zhang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
13
|
Yu Z, Jiang X, Yin J, Han L, Xiong C, Huo Z, Xu J, Shang J, Xi K, Nong L, Huang Y, Zhou X. CK1ε drives osteogenic differentiation of bone marrow mesenchymal stem cells via activating Wnt/β-catenin pathway. Aging (Albany NY) 2023; 15:10193-10212. [PMID: 37787983 PMCID: PMC10599756 DOI: 10.18632/aging.205067] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023]
Abstract
The treatment of bone defects is a difficult problem in orthopedics. At present, the treatment mainly relies on autologous or allogeneic bone transplantation, which may lead to some complications such as foreign body rejection, local infection, pain, or numbness at the bone donor site. Local injection of conservative therapy to treat bone defects is one of the research hotspots at present. Bone marrow mesenchymal stem cells (BMSCs) can self-renew, significantly proliferate, and differentiate into various types of cells. Although it has been reported that CK1ε could mediate the Wnt/β-catenin pathway, leading to the development of the diseases, whether CK1ε plays a role in bone regeneration through the Wnt/β-catenin pathway has rarely been reported. The purpose of this study was to investigate whether CK1ε was involved in the osteogenic differentiation (OD) of BMSCs through the Wnt/β-catenin pathway and explore the mechanism. We used quantitative reverse transcription-polymerase chain reaction (qRT-qPCR), Western blots, immunofluorescence, alkaline phosphatase, and alizarin red staining to detect the effect of CK1ε on the OD of BMSCs and the Wnt/β-catenin signaling pathway. CK1ε was highly expressed in BMSCs with OD, and our study further demonstrated that CK1ε might promote the OD of BMSCs by activating DLV2 phosphorylation, initiating Wnt signaling downstream, and activating β-catenin nuclear transfer. In addition, by locally injecting a CK1ε-carrying adeno-associated virus (AAV5- CK1ε) into a femoral condyle defect rat model, the overexpression of CK1ε significantly promoted bone repair. Our data show that CK1ε was involved in the regulation of OD by mediating Wnt/β-catenin. This may provide a new strategy for the treatment of bone defects.
Collapse
Affiliation(s)
- Zhentang Yu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Orthopedics, Yibin Integrated Traditional Chinese and Western Medicine Hospital, Yibin 644104, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Xijia Jiang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jianjian Yin
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Lei Han
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Chengwei Xiong
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Zhennan Huo
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jie Xu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jingjing Shang
- Department of Pharmacy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou 215006, China
| | - Luming Nong
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai 811800, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| |
Collapse
|
14
|
Shang J, Yu Z, Xiong C, Zhang J, Gong J, Yu C, Huang Y, Zhou X. Resistin targets TAZ to promote osteogenic differentiation through PI3K/AKT/mTOR pathway. iScience 2023; 26:107025. [PMID: 37389179 PMCID: PMC10300212 DOI: 10.1016/j.isci.2023.107025] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/06/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Osteogenic differentiation (OD) of bone marrow mesenchymal stem cells (BMSCs) contributes significantly to the regeneration of bone defects. Resistin, an adipose tissue-specific secretory factor, has been shown to involve many different functions, including metabolism, inflammation, cancer, and bone remodeling. However, the effects and mechanisms of resistin on OD of BMSCs remain unclear. Herein, we demonstrated that resistin was highly expressed in BMSCs with OD. Upregulation of resistin contributed to the progression of OD of BMSCs by activating PI3K/AKT/mTOR signaling pathway. In addition, resistin facilitated OD by targeting transcriptional co-activator with PDZ-binding motif (TAZ). In a rat femoral condyle bone defect model, local injection of resistin significantly promoted bone repair and improved bone formation. This work contributes to better understanding the mechanism of resistin directly involved in the OD and might provide a new therapeutic strategy for bone defect regeneration.
Collapse
Affiliation(s)
- JingJing Shang
- Department of Pharmacy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Zhentang Yu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Department of Graduate School, Dalian Medical University, Dalian, Liaoning 116000, China
| | - Chengwei Xiong
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Junjie Zhang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Jinhong Gong
- Department of Pharmacy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Changlin Yu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai 811800, China
| |
Collapse
|
15
|
Tang J, Sang Z, Zhang X, Song C, Tang W, Luo X, Yan M. Impacts of residual 3D printing metal powders on immunological response and bone regeneration: an in vivo study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:29. [PMID: 37227574 DOI: 10.1007/s10856-023-06727-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/03/2023] [Indexed: 05/26/2023]
Abstract
Residual powder is a defect in powder bed fusion-based additive manufacturing (3D printing), and it is difficult to completely remove it from as-printed materials. In addition, it is not necessary to apply 3D printed implants with residual powder in the clinic. The immunological response triggered by the residual powder is an important area of study in medical research. To further understand the possible immunological reactions and hidden dangers caused by residual powders in vivo, this study compared the immunological reactions and osteolysis caused by typical powders for four implant materials: 316 L stainless steel, CoCrMo, CP-Ti, and Ti-6Al-4V (particle size range of 15-45 μm), in a mouse skull model. Furthermore, the possible immunological responses and bone regeneration induced by the four 3D printed implants with residual powder in a rat femur model were compared. In the mouse skull model, it was found that the 316L-S, CoCrMo-S, and especially the 316L-M powders, upregulated the expression of pro-inflammatory factors, increased the ratio of RANKL/OPG, and activated more functional osteoclasts, resulting in more severe bone resorption compared with those in other groups. In the rat femur model, which is more suitable for clinical practice, there is no bone resorption in implants with residual powders, but they show good bone regeneration and integration ability because of their original roughness. The results indicate that the expressions of inflammatory cytokines in all experimental groups were the same as those in the control group, showing good biological safety. The results answered some critical questions related to additively manufactured medical materials in vivo and indicated that as-printed implants may have great potential in future clinical applications.
Collapse
Affiliation(s)
- Jincheng Tang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhuo Sang
- The Eighth Affiliated Hospital, Sun Yat- sen University, Shenzhen, 518033, China.
| | - Xiaolei Zhang
- The Eighth Affiliated Hospital, Sun Yat- sen University, Shenzhen, 518033, China
| | - Changhui Song
- Department of Mechanical and Automotive Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Wei Tang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiaoping Luo
- Nanjing Stomatological Hospital Medical School of Nanjing University, Nanjing, 210008, China
| | - Ming Yan
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.
- Jiaxing Research Institute, Southern University of Science and Technology, Jiaxing, 314001, China.
| |
Collapse
|