1
|
Brigi C, Aghila Rani K, Selvakumar B, Hamad M, Abou Neel EA, Samsudin A. Decoding biomaterial-associated molecular patterns (BAMPs): influential players in bone graft-related foreign body reactions. PeerJ 2025; 13:e19299. [PMID: 40292103 PMCID: PMC12024449 DOI: 10.7717/peerj.19299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Bone grafts frequently induce immune-mediated foreign body reactions (FBR), which hinder their clinical performance and result in failure. Understanding biomaterial-associated molecular patterns (BAMPs), including physicochemical properties of biomaterial, adsorbed serum proteins, and danger signals, is crucial for improving bone graft outcomes. Recent studies have investigated the role of BAMPs in the induction and maintenance of FBR, thereby advancing the understanding of FBR kinetics, triggers, stages, and key contributors. This review outlines the stages of FBR, the components of BAMPs, and their roles in immune activation. It also discusses various bone grafting biomaterials, their physicochemical properties influencing protein adsorption and macrophage modulation, and the key mechanisms of protein adsorption on biomaterial surfaces. Recent advancements in surface modifications and immunomodulatory strategies to mitigate FBR are also discussed. Furthermore, the authors look forward to future studies that will focus on a comprehensive proteomic analysis of adsorbed serum proteins, a crucial component of BAMPs, to identify proteins that promote or limit inflammation. This understanding could facilitate the design of biomaterials that selectively adsorb beneficial proteins, thereby reducing the risk of FBR and enhancing bone regeneration.
Collapse
Affiliation(s)
- Carel Brigi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - K.G. Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Mawieh Hamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ensanya Ali Abou Neel
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - A.R. Samsudin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
2
|
Su X, Wang M, Yuan R, Guo L, Han Y, Huang C, Li A, Kaplan DL, Wang X. Organoids in Dynamic Culture: Microfluidics and 3D Printing Technologies. ACS Biomater Sci Eng 2025. [PMID: 40248908 DOI: 10.1021/acsbiomaterials.4c02245] [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: 04/19/2025]
Abstract
With the rapid advancement of biomaterials and tissue engineering technologies, organoid research and its applications have made significant strides. Organoids are increasingly utilized in pharmacology, regenerative medicine, and precision clinical medicine. Current trends in organoid research are moving toward multifunctional composite three-dimensional cultivation and dynamic cultivation strategies. Key technologies driving this evolution, including 3D printing and microfluidics, continue to impact new areas of discovery and clinical relevance. This review provides a systematic overview of these emerging trends, discussing the strengths and limitations of these critical technologies and offering insight and research directions for professionals working in the organoid field.
Collapse
Affiliation(s)
- Xin Su
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Mingqi Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Ruqiang Yuan
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Lina Guo
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Yinhe Han
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Chun Huang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Ang Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Xiuli Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| |
Collapse
|
3
|
Yu H, Liu K, Hou Y, Guo L, Sun L, Zhou W, Wang L, Pan P, Sun S, Chen J. Facile one-step antibacterial biomineralized scaffolds through regulation of chitosan by imitating bone ingredient bonding to inspire endogenous repair. Int J Biol Macromol 2025; 309:142827. [PMID: 40203943 DOI: 10.1016/j.ijbiomac.2025.142827] [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: 12/27/2024] [Revised: 03/19/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
In situ mineralization based on chitosan (CS) as an organic template achieves uniform distribution of nano-hydroxyapatite (nHAp), and is expected to enhance interface bonding between materials and tissues, therefore promoting endogenous bone repair. However, chitosan-based materials are difficult to provide certain mechanical and antibacterial properties. In this study, Ca2+ and PO43- were used as the precursor of nHAP, Zn2+ as the precursor of nano-ZnO, and synthesized nHAp/ZnO particles in situ on the surface of chitosan to form CS/HAp/ZnO bone repair scaffold. The results indicated that the incorporated Zn2+ was chemically bonded to the system, with formed particles evenly distributed, which achieved more efficient antibacterial activity and osteogenic induction performance in vivo. The material had interconnected pore structure with a porosity of >70 %, which simulates the microenvironment of natural bone. The formed inorganic particles improved the mechanical properties of the chitosan scaffold to match the rate of new bone formation. ZnO endowed scaffolds with antibacterial activity, with antibacterial rate of >95 %. In addition, the material also showed good cell compatibility and biological activity, and promoted the adhesion, migration, proliferation and differentiation of osteoblast-related cells.
Collapse
Affiliation(s)
- Hui Yu
- Marine College, Shandong University, Weihai 264209, China
| | - Kaihua Liu
- Marine College, Shandong University, Weihai 264209, China
| | - Yage Hou
- Marine College, Shandong University, Weihai 264209, China
| | - Liangyu Guo
- Marine College, Shandong University, Weihai 264209, China.
| | - Lixin Sun
- Marine College, Shandong University, Weihai 264209, China
| | - Wutong Zhou
- Marine College, Shandong University, Weihai 264209, China
| | - Lin Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Panpan Pan
- Marine College, Shandong University, Weihai 264209, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai 200444, China.
| | - Shengjun Sun
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250000, China.
| | - Jingdi Chen
- Marine College, Shandong University, Weihai 264209, China; Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai 265599, China.
| |
Collapse
|
4
|
Ling Z, Ge X, Jin C, Song Z, Zhang H, Fu Y, Zheng K, Xu R, Jiang H. Copper doped bioactive glass promotes matrix vesicles-mediated biomineralization via osteoblast mitophagy and mitochondrial dynamics during bone regeneration. Bioact Mater 2025; 46:195-212. [PMID: 39760064 PMCID: PMC11699476 DOI: 10.1016/j.bioactmat.2024.12.010] [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: 08/25/2024] [Revised: 11/05/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Bone defect repair remains a great challenge in the field of orthopedics. Human body essential trace element such as copper is essential for bone regeneration, but how to use it in bone defects and the underlying its mechanisms of promoting bone formation need to be further explored. In this study, by doping copper into mesoporous bioactive glass nanoparticles (Cu-MBGNs), we unveil a previously unidentified role of copper in facilitating osteoblast mitophagy and mitochondrial dynamics, which enhance amorphous calcium phosphate (ACP) release and subsequent biomineralization, ultimately accelerating the process of bone regeneration. Specifically, by constructing conditional knockout mice lacking the autophagy gene Atg5 in osteogenic lineage cells, we first confirmed the role of Cu-MBGNs-promoted bone formation via mediating osteoblast autophagy pathway. Then, the in vitro studies revealed that Cu-MBGNs strengthened mitophagy by inducing ROS production and recruiting PINK1/Parkin, thereby facilitating the efficient release of ACP from mitochondria into matrix vesicles for biomineralization during bone regeneration. Moreover, we found that Cu-MBGNs promoted mitochondrion fission via activating dynamin related protein 1 (Drp1) to reinforce mitophagy pathway. Together, this study highlights the potential of Cu-MBGNs-mediated mitophagy and biomineralization for augmenting bone regeneration, offering a promising avenue for the development of advanced bioactive materials in orthopedic applications.
Collapse
Affiliation(s)
- Ziji Ling
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Xiao Ge
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Chengyu Jin
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Zesheng Song
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Hang Zhang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Yu Fu
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Kai Zheng
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Rongyao Xu
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Hongbing Jiang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, 210029, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| |
Collapse
|
5
|
Li S, Shan Y, Chen J, Su R, Zhao L, He R, Li Y. Piezoelectricity Promotes 3D-Printed BTO/β-TCP Composite Scaffolds with Excellent Osteogenic Performance. ACS APPLIED BIO MATERIALS 2025; 8:2204-2214. [PMID: 40013453 DOI: 10.1021/acsabm.4c01754] [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: 02/28/2025]
Abstract
Piezoelectricity is reported to be able to promote bone scaffolds with excellent osteogenic performance. Herein, barium titanate/β-tricalcium phosphate (BTO/β-TCP) piezoelectric composite scaffolds were 3D printed, and their osteogenic performances were investigated in detail. The fabrication of BTO/β-TCP piezoelectric composite scaffolds employed cutting-edge DLP 3D printing technology. The scaffolds, featuring a triply periodic minimal surface (TPMS) design with a porosity of 60%, offered a unique structural framework. A comprehensive assessment of the composition, piezoelectric properties, and mechanical characteristics of the BTO/β-TCP scaffolds was conducted. Notably, an increase in the BTO volume fraction from 50 to 80 vol % within the scaffolds led to a reduction in compressive strength, decreasing from 2.47 to 1.74 MPa. However, this variation was accompanied by a substantial enhancement in the piezoelectric constant d33, soaring from 1.4 pC/N to 21.6 pC/N. Utilizing mouse osteoblasts (MC3T3-E1) in a live/dead cell staining assay, under the influence of external ultrasound, demonstrated the commendable biocompatibility of these piezoelectric composite ceramic bone scaffolds. Furthermore, thorough analyses of alkaline phosphatase (ALP) activity and polymerase chain reaction (PCR) findings provided compelling evidence of the scaffolds' superior osteogenic properties, underpinning their effectiveness at the cellular protein and gene levels. In conclusion, this study offers a groundbreaking strategy for the employment of BTO/β-TCP piezoelectric composite scaffolds in bone implant applications, harnessing their unique blend of biocompatibility, piezoelectricity, and osteogenic potential.
Collapse
Affiliation(s)
- Suyun Li
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yanbo Shan
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Jingyi Chen
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Ruyue Su
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Lisheng Zhao
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Rujie He
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Ying Li
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
6
|
Seraj FQM, Kheradmand D, Najafi S, Dizaj MJT, Salmasian S, Ghasemi A, Ghasemi R, Zabihyan S, Etemadrezaie H, Ebrahimnia F, Mowla A, Baharvahdat H. Customized 3D-printed Poly ether ether ketone cranial implant for cranioplasty of skull defects. Neurosurg Rev 2025; 48:301. [PMID: 40090957 DOI: 10.1007/s10143-025-03448-7] [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: 11/05/2024] [Revised: 01/28/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Cranioplasty become one of the common surgeries in the neurosurgical field. The complex configuration of the cranium makes the bone reconstruction of the skull defects a challenging procedure. While the patient's own bone flap is the best option, other materials such as titanium and Poly ether ether Ketone (PEEK) were used for cranioplasty when the autogenous flap of the patient is unavailable. Recently, customized 3D printing implants were developed for cranioplasty with favorable outcomes. In this study, we evaluated the outcome of customized 3-dimentional (3D)-printed PEEK implant cranioplasty for reconstructing skull defects, especially large ones. We report a series of 27 patients whose skull defects were reconstructed by a customized 3D-printed PEEK implants. Demographic data and postoperative clinical and imaging findings were reviewed. We analyzed the complications, clinical and aesthetic outcomes during mid-term follow-up. During the surgery, all the implants were perfectly fitted in the skull defect without any major modification. Aesthetic results were satisfactory in 74.1% of patients. 4 patients (14.8%) had complications, including 3 deep cranial infections (11.1%) and one extradural hematoma (3,7%). In 2 patients (7.4%), the implants were removed due to infection. Therefore, when autologous bone is unavailable or, in selected cases with large or complex skull defects, customized 3D-printed PEEK implants could be a proper option to reconstruct these defects.
Collapse
Affiliation(s)
| | - Daniel Kheradmand
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajjad Najafi
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurosurgery Department, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Sanaz Salmasian
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Ghasemi
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Ghasemi
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Zabihyan
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Etemadrezaie
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ashkan Mowla
- Neurosurgery Department, Keck School of Medicine, Los Angeles, USA
| | - Humain Baharvahdat
- Neurosurgery Department, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Interventional Neuroradiology, Rothschild Foundation Hospital, Paris, France.
| |
Collapse
|
7
|
Cen C, Zhang Y, Cao Y, Hu C, Tang L, Liu C, Wang T, Peng W. Construction of a 3D Degradable PLLA/β-TCP/CS Scaffold for Establishing an Induced Membrane Inspired by the Modified Single-Stage Masquelet Technique. ACS Biomater Sci Eng 2025; 11:1629-1645. [PMID: 39943835 PMCID: PMC11900768 DOI: 10.1021/acsbiomaterials.4c01849] [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: 10/04/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/11/2025]
Abstract
Although the Masquelet-induced membrane technique (MIMT) is now employed worldwide for bone defects, it often needs to be repeated and autogenous bone graft. This study aims to investigate the theoretical feasibility of replacing PMMA (poly(methyl methacrylate)) bone cement with PLLA (poly-l-lactic acid)/β -TCP (beta-tricalcium phosphate)/CS (calcium sulfate) scaffold for single-stage bone defect reconstruction, which evoke the induced membrane (IM) formation in the early stage and directly acts as the implantation in the second stage to reconstruct the bone defect. We constructed a corn-like PLLA/β -TCP/CS scaffold by the fused deposition 3D printing method. The characterizations of the scaffolds were investigated systematically. The P/T15/S15 scaffolds (the PLLA/β -TCP/CS scaffold with a 15% mass fraction of β-TCP and 15% mass fraction of CS) were filled into the large-segmental radius bone defects of white rabbits to evoke the formation of IMs. HE (hematoxylin-eosin) and VG (van gieson) staining, along with immunofluorescent staining, were performed to analyze the architecture and cellularity, the expression of BMP-2 (bone morphogenetic protein-2), VEGF (vascular endothelial growth factor), and TGF-β1 (transforming growth factor-β1) was evaluated by IHC (immunohistochemistry) and WB (western-blot) respectively, the ALP (alkaline phosphatase) and ARS (alizarin red S) staining was applied to assess the osteogenic potential. The corn-like PLLA/β-TCP/CS scaffolds with excellent physicochemical properties are successfully constructed using the fused deposition 3D printing technique. The HE and VG staining, along with immunofluorescent staining, suggested that the P/T15/S15 scaffold effectively mediated the formation of IM after 6 weeks of placement. A significant presence of M2 macrophages was observed in IM. The results of IHC and WB demonstrated that the IMs derived from the P/T15/S15 scaffolds exhibited elevated levels of VEGF, BMP-2, and TGF-β1, all of which promote the osteogenic differentiation of BMSCs. The results of cellular immunofluorescence, flow cytometry, and WB indicate that P/T15/S15 regulates the phenotypic polarization of M0 macrophages toward the M2 phenotype via the PI3K/AKT/β-Catenin pathway. These findings suggest that the biodegradable PLLA/β-TCP/CS scaffold may serve as a viable alternative to PMMA bone cement for single-stage bone defect reconstruction, owing to its unique ability to stimulate IM formation and promote the polarization of macrophages toward the M2 phenotype. This work presents innovative materials and strategies for the management of bone defects.
Collapse
Affiliation(s)
- Chaode Cen
- School
of Clinical Medicine, The Guizhou Medical
University, Guiyang 550025,China
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Yong Zhang
- Department
of gynaecology and obstetrics, Guiyang First
People’s Hospital, Guiyang 550005, China
| | - Yongfei Cao
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Chaoran Hu
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Lingli Tang
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Chengwei Liu
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Tao Wang
- School
of Clinical Medicine, The Guizhou Medical
University, Guiyang 550025,China
- Department
of Emergency Surgery, The Affiliated Hospital
of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Wuxun Peng
- School
of Clinical Medicine, The Guizhou Medical
University, Guiyang 550025,China
- Department
of Emergency Surgery, The Affiliated Hospital
of Guizhou Medical University, Guiyang, Guizhou 550004, China
| |
Collapse
|
8
|
Abedi N, Sadeghian A, Kouhi M, Haugen HJ, Savabi O, Nejatidanesh F. Immunomodulation in Bone Tissue Engineering: Recent Advancements in Scaffold Design and Biological Modifications for Enhanced Regeneration. ACS Biomater Sci Eng 2025; 11:1269-1290. [PMID: 39970366 DOI: 10.1021/acsbiomaterials.4c01613] [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: 02/21/2025]
Abstract
Bone defects, whether caused by trauma, cancer, infectious diseases, or surgery, can significantly impair people's quality of life. Although autografts are the gold standard for treating bone defects, they often fall short in adequately forming bone tissue. The field of bone tissue engineering has made strides in using scaffolds with various biomaterials, stem cells, and growth factors to enhance bone healing. However, some biological structures do not yield satisfactory therapeutic outcomes for new bone formation. Recent studies have shed light on the crucial role of immunomodulation, specifically the interaction between the implanted scaffold and host immune systems, in bone regeneration. Immune cells, particularly macrophages, are pivotal in the inflammatory response, angiogenesis, and osteogenesis. This review delves into the immune system's mechanism toward foreign bodies and the recent advancements in scaffolds' physical and biological properties that foster bone regeneration by modulating macrophage polarization to an anti-inflammatory phenotype and enhancing the osteoimmune microenvironment.
Collapse
Affiliation(s)
- Niloufar Abedi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Aida Sadeghian
- Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Monireh Kouhi
- Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway
| | - Omid Savabi
- Department of Prosthodontics, Dental Research Center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Farahnaz Nejatidanesh
- Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| |
Collapse
|
9
|
Kim J, Lee HJ, Choi EA, Lee JH, Oh J, Byeon DH, Kwak HS, Park CH. Effects of structural design on the mechanical performances of poly-L-lactic acid cardiovascular scaffolds using FEA and in vitro methods. J Mech Behav Biomed Mater 2025; 163:106849. [PMID: 39652986 DOI: 10.1016/j.jmbbm.2024.106849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/24/2024] [Accepted: 11/30/2024] [Indexed: 02/08/2025]
Abstract
OBJECTIVE In this study, we propose distinct and novel types of scaffold geometries to improve the mechanical performance of Poly-L-lactic Acid (PLLA) bioresorbable vascular scaffolds (BVS), investigating how different geometries of PLLA BVS influence their mechanical performances through finite element analysis (FEA) and in vitro experiment methods. METHODS Four different types of scaffold geometries were modelled for FEA and manufactured for in vitro experiments. PLLA tubes with 110 μm thickness were used in manufacturing the scaffolds. For FEA measurements, material properties and bilinear material models were obtained from tensile testing using the PLLA tubes employed for manufacturing. Various measurements were conducted including crush resistance, radial strength in both the laser-cut and deployed state, three-point bending, and scaffold crimping/expansion test. RESULTS Overall, the FEA results were similar to the experimental results. Design A, which had a conventional open-cell geometry with straight bridges, showed inferior crush resistance and radial strength to those of the other tested geometries. Design B exhibited the most well-balanced scaffold performances in terms of radial strengths, crush resistance, three-point bending, and crimping/expansion behaviors. Notably, it showed minimum plastic strain during crimping and expanding deformations in FEA. CONCLUSIONS Findings from such distinct and novel types of scaffold geometries shown by this study may provide a valuable understanding using PLLA scaffolds as cardiovascular devices.
Collapse
Affiliation(s)
- Jinwoo Kim
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Hyeon Ji Lee
- Department of Mechanical Design Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea; Innovative Mechanobio Active Materials Based Medical Device Demonstration Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Eun Ae Choi
- Department of Mechanical Design Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea; Innovative Mechanobio Active Materials Based Medical Device Demonstration Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jung Ho Lee
- Department of Mechanical Design Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea; Innovative Mechanobio Active Materials Based Medical Device Demonstration Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jin Oh
- Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea; Innovative Mechanobio Active Materials Based Medical Device Demonstration Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Dae-Heung Byeon
- CGBio Co. Ltd., 244 Galmachi-ro, Jungwon-u, Seongnam, 13211, Republic of Korea
| | - Hyo Sung Kwak
- Department of Radiology and Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, 54896, Republic of Korea.
| | - Chan Hee Park
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea; Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea; Department of Mechanical Design Engineering, Graduate School, Jeonbuk National University, Jeonju 54896, Republic of Korea; Advanced Mechanical Components Design & Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea; Innovative Mechanobio Active Materials Based Medical Device Demonstration Center, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| |
Collapse
|
10
|
Shuai Y, Yang T, Zheng M, Zheng L, Wang J, Mao C, Yang M. Oriented Cortical-Bone-Like Silk Protein Lamellae Effectively Repair Large Segmental Bone Defects in Pigs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414543. [PMID: 39871679 PMCID: PMC11899506 DOI: 10.1002/adma.202414543] [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: 09/25/2024] [Revised: 12/19/2024] [Indexed: 01/29/2025]
Abstract
Assembling natural proteins into large, strong, bone-mimetic scaffolds for repairing bone defects in large-animal load-bearing sites remain elusive. Here this challenge is tackled by assembling pure silk fibroin (SF) into 3D scaffolds with cortical-bone-like lamellae, superior strength, and biodegradability through freeze-casting. The unique lamellae promote the attachment, migration, and proliferation of tissue-regenerative cells (e.g., mesenchymal stem cells [MSCs] and human umbilical vein endothelial cells) around them, and are capable of developing in vitro into cortical-bone organoids with a high number of MSC-derived osteoblasts. High-SF-content lamellar scaffolds, regardless of MSC inoculation, regenerated more bone than non-lamellar or low-SF-content lamellar scaffolds. They accelerated neovascularization by transforming macrophages from M1 to M2 phenotype, promoting bone regeneration to repair large segmental bone defects (LSBD) in minipigs within three months, even without growth factor supplements. The bone regeneration can be further enhanced by controlling the orientation of the lamella to be parallel to the long axis of bone during implantation. This work demonstrates the power of oriented lamellar bone-like protein scaffolds in repairing LSBD in large animal models.
Collapse
Affiliation(s)
- Yajun Shuai
- Institute of Applied Bioresource ResearchCollege of Animal SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang ProvinceZhejiang UniversityHangzhou310058China
| | - Tao Yang
- Department of Biomedical EngineeringThe Chinese University of Hong KongSha TinHong Kong SARChina
| | - Meidan Zheng
- Institute of Applied Bioresource ResearchCollege of Animal SciencesZhejiang UniversityHangzhou310058China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for BiomedicineLife Sciences InstituteGuangxi Medical UniversityNanning530021China
| | - Jie Wang
- Institute of Applied Bioresource ResearchCollege of Animal SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang ProvinceZhejiang UniversityHangzhou310058China
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongSha TinHong Kong SARChina
| | - Mingying Yang
- Institute of Applied Bioresource ResearchCollege of Animal SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang ProvinceZhejiang UniversityHangzhou310058China
| |
Collapse
|
11
|
Li J, Hietel B, Brunk MGK, Reimers A, Willems C, Groth T, Cynis H, Adelung R, Schütt F, Sacher WD, Poon JKS. 3D-printed microstructured alginate scaffolds for neural tissue engineering. Trends Biotechnol 2025; 43:447-461. [PMID: 39658448 DOI: 10.1016/j.tibtech.2024.10.013] [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: 04/26/2024] [Revised: 10/18/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024]
Abstract
Alginate (Alg) is a versatile biopolymer for scaffold engineering and a bioink component widely used for direct cell printing. However, due to a lack of intrinsic cell-binding sites, Alg must be functionalized for cellular adhesion when used as a scaffold. Moreover, direct cell-laden ink 3D printing requires tedious disinfection procedures and cell viability is compromised by shear stress. Here, we demonstrate proof-of-concept, bioactive additive-free, microstructured Alg (M-Alg) scaffolds for neuron culture. The M-Alg scaffold was formed by introducing tetrapod-shaped ZnO (t-ZnO) microparticles into the ink as structural templates for interconnected channels and textured surfaces in the 3D-printed Alg scaffold, which were subsequently removed. Neurons exhibited significantly improved adhesion and growth on these M-Alg scaffolds compared with pristine Alg (P-Alg) scaffolds, with extensive neurite outgrowth and spontaneous neural activity, indicating the maturation of neuronal networks. These transparent, porous, additive-free Alg-based scaffolds with neuron affinity are promising for neuroregenerative and organoid-related research.
Collapse
Affiliation(s)
- Jianfeng Li
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada.
| | - Benjamin Hietel
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Weinbergweg 22, 06120, Halle, Germany
| | - Michael G K Brunk
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada
| | - Armin Reimers
- Functional Nanomaterials, Department of Materials Science, Kiel University, Kaiserstraße 2, 24143 Kiel, Germany
| | - Christian Willems
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120, Halle, Germany
| | - Thomas Groth
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120, Halle, Germany
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Weinbergweg 22, 06120, Halle, Germany; Junior Research Group, Immunomodulation in Pathophysiological Processes, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120, Halle, Germany
| | - Rainer Adelung
- Functional Nanomaterials, Department of Materials Science, Kiel University, Kaiserstraße 2, 24143 Kiel, Germany
| | - Fabian Schütt
- Functional Nanomaterials, Department of Materials Science, Kiel University, Kaiserstraße 2, 24143 Kiel, Germany
| | - Wesley D Sacher
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada
| | - Joyce K S Poon
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada; Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Wang J, Huang Z, Han Z, Luan J, Li Z, Guo X, Yang D, Cui Y, Han J, Xu D. TPMS-Gyroid Scaffold-Mediated Up-Regulation of ITGB1 for Enhanced Cell Adhesion and Immune-Modulatory Osteogenesis. Adv Healthc Mater 2025:e2404768. [PMID: 39853929 DOI: 10.1002/adhm.202404768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/09/2025] [Indexed: 01/26/2025]
Abstract
The porous structure is crucial in bone tissue engineering for promoting osseointegration. Among various structures, triply periodic minimal surfaces (TPMS) -Gyroid has been extensively studied due to its superior mechanical and biological properties. However, previous studies have given limited attention to the impact of unit cell size on the biological performance of scaffolds. In this research, four TPMS-Gyroid titanium scaffolds with different unit cell sizes (TG15, TG20, TG25, and TG30) are fabricated using Selective Laser Melting (SLM) to explore their effects on osseointegration. Mechanical tests revealed that TG15 and TG20 exhibited superior compressive strength. In vitro experiments demonstrated that TG20 facilitated better cell adhesion through robust integrin protein expression initially, which subsequently enhanced cell proliferation and osteogenic differentiation. Furthermore, macrophages on TG20 showed higher Integrin β1 (ITGB1) expression, promoting their polarization to the M2 phenotype, which suppressed inflammation, fostered bone integration, and angiogenesis. In vivo studies confirmed TG20's effectiveness in promoting bone ingrowth by reducing inflammation. This study highlights TG20's structural advantages, making it a promising bone scaffold with exceptional osteogenic and angiogenic properties through osteoimmune microenvironment modulation. Therefore, TG20 holds significant potential for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, Shandong, 250014, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
- NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Zenan Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, Shandong, 250014, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
- NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Zhenzhong Han
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, Shandong, 250014, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
- NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Jing Luan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, Shandong, 250014, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
- NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Zihan Li
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
| | - Xutong Guo
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
| | - Dongxu Yang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
| | - Yazhou Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, Shandong, 250014, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
- NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
- Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong, 250355, China
| | - Jinxiang Han
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, Shandong, 250014, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
- NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| | - Duo Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, Shandong, 250014, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, 250117, China
- NHC Key Laboratory of biotechnology drugs (Shandong Academy of Medical Sciences), Ji'nan, Shandong, 250117, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, Shandong, 250117, China
| |
Collapse
|
13
|
Sikder P. A comprehensive review on the State of the Art in the research and development of poly-ether-ether-ketone (PEEK) biomaterial-based implants. Acta Biomater 2025; 191:29-52. [PMID: 39579846 DOI: 10.1016/j.actbio.2024.11.033] [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: 08/13/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Polyetheretherketone (PEEK) is a preferred high-performance polymer in the spine, orthopedic, and craniomaxillofacial implant industry. However, despite its commendable mechanical properties, its bioinert nature limits the implants from integrating with neighboring tissues, impacting the implant's long-term performance. To address this limitation, various kinds of surface functionalization techniques have been developed over the years. Noteworthy efforts have been made to incorporate bioactive fillers in the PEEK matrix to develop standalone bioactive composites. In personalized medicine, significant advances have been made in the 3D Printing of PEEK implants. 3D-printed PEEK implants are now being developed at Point-of-Care, significantly reducing manufacturing and logistic time. Given the recent clinical follow-up updates and advancements in PEEK-based implants, PEEK implants are witnessing an important phase in its history. Recognizing this vital phase, this paper aims to comprehensively review the advancements of PEEK implants over the past decade. The review starts with an overview of the clinical impact of varying PEEK implants, followed by PEEK's surface functionalization techniques and engineering of PEEK-based bioactive composites. Next, this review describes the advancements made in the 3D printing of PEEK implants and points out the essential considerations that should be considered when developing 3D-printed PEEK-based implants. Finally, the review ends with an estimated projection about the future of PEEK-based implants. Readers are expected to gain an all-encompassing and in-depth understanding of PEEK biomedical implants' past, present, and future, enabling researchers to advance the research and development of PEEK-based implants in the required direction. STATEMENT OF SIGNIFICANCE: PEEK is a preferred high-performance polymer in the implant industry, with notable benefits over metallic and ceramic implants, such as bone-matching stiffness and durability. Significant strides have been made in the last decade to make PEEK implants bioactive and utilize 3D Printing to develop patient-specific implants. Given the recent advancements in PEEK-based implants, this review aims to provide an all-encompassing and in-depth understanding of PEEK biomedical implants' past, present, and future. It will comprehensively discuss the know-how gained from the clinical follow-up, the strategies to address the limitations of PEEK implants, and the essential considerations in 3D Printing of PEEK implants. This review will enable researchers to advance the research and development of PEEK implants in the required direction.
Collapse
Affiliation(s)
- Prabaha Sikder
- Department of Mechanical Engineering, Cleveland State University, Cleveland, OH 44115, United States.
| |
Collapse
|
14
|
Yuan Z, Zhang L, Shafiq M, Wang X, Cai P, Hafeez A, Ding Y, Wang Z, El-Newehy M, Meera Moydeen Abdulhameed, Jiang L, Mo X, Xu Y. Composite superplastic aerogel scaffolds containing dopamine and bioactive glass-based fibers for skin and bone tissue regeneration. J Colloid Interface Sci 2024; 673:411-425. [PMID: 38878375 DOI: 10.1016/j.jcis.2024.06.098] [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: 02/23/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/26/2024]
Abstract
Multifunctional bioactive biomaterials with integrated bone and soft tissue regenerability hold great promise for the regeneration of trauma-affected skin and bone defects. The aim of this research was to fabricate aerogel scaffolds (GD-BF) by blending the appropriate proportions of short bioactive glass fiber (BGF), gelatin (Gel), and dopamine (DA). Electrospun polyvinyl pyrrolidone (PVP)-BGF fibers were converted into short BGF through calcination and homogenization. Microporous GD-BF scaffolds displayed good elastic deformation recovery and promoted neo-tissue formation. The DA could enable thermal crosslinking and enhance the mechanical properties and structural stability of the GD-BF scaffolds. The BGF-mediated release of therapeutic ions shorten hemostatic time (<30 s) in a rat tail amputation model and a rabbit artery injury model alongside inducing the regeneration of skin appendages (e.g., blood vessels, glands, etc.) in a full-thickness excisional defect model in rats (percentage wound closure: GD-BF2, 98 % vs. control group, 83 %) at day 14 in vitro. Taken together, these aerogel scaffolds may have significant promise for soft and hard tissue repair, which may also be worthy for the other related disciplines.
Collapse
Affiliation(s)
- Zhengchao Yuan
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, No. 183, Xinqiao Street, Shapingba District, Chongqing 400037, China; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai 201600, PR China
| | - Lixiang Zhang
- Department of Health Management, Characteristic Medical Center of Chinese People's Armed Police Force, 220 Chenglin Road, Hedong District, Tianjin, China
| | - Muhammad Shafiq
- Innovation Center of Nanomedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Xinyi Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai 201600, PR China
| | - Pengfei Cai
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai 201600, PR China
| | - Abdul Hafeez
- Department of Mechanical Engineering, Faculty of Engineering, University of Engineering & Technology (UET), Lahore 54000, Pakistan
| | - Yangfan Ding
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai 201600, PR China
| | - Zewen Wang
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, No. 183, Xinqiao Street, Shapingba District, Chongqing 400037, China
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Meera Moydeen Abdulhameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Lianyong Jiang
- Department of Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai 201600, PR China.
| | - Yuan Xu
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, No. 183, Xinqiao Street, Shapingba District, Chongqing 400037, China.
| |
Collapse
|
15
|
Luo H, Yang X, Ding Q, Sheng B, Deng J, Yan X, Wu Y, Chen H, Hao C, Yuan S, Zeng J, Zhou W. Tensile properties and deformation by different compatibilizers in bio-based polylactide/poly(4-hydroxybutyrate) blends. Int J Biol Macromol 2024; 281:136550. [PMID: 39426776 DOI: 10.1016/j.ijbiomac.2024.136550] [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: 07/05/2024] [Revised: 09/21/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Blending chemically synthesized poly(4-hydroxybutyrate) (P4HB) with polylactide (PLLA) can overcome PLLA's brittleness, offering fully biobased blends. However, due to low compatibility between PLLA and P4HB, the influence of compatibilizers on the morphology, structure and tensile deformation of PLLA/P4HB blends remains to be unresolved. This article introduces reactive poly(methyl methacrylate-co-styrene-glycidyl methacrylate) (MSG) and non-reactive polyformaldehyde (POM) compatibilizers to improve the compatibility between P4HB and PLLA, achieving the maximal elongation at break exceeding 300 % at 2 wt% MSG or 3 wt% POM. MSG inhibits PLLA crystallization, extending stress stability in the silver streak stage, while POM enhances crystallization, prolonging the strain-hardening stage. Small-angle X-ray scattering (SAXS) and wide-angle X-ray scattering (WAXS) analysis show that pristine PLLA forms voids before fracture, and PLLA/P4HB blends cavitate at the yield point and develop crazes in the silver streak stage. MSG effectively transmits stress and delays cavitation, extending the silver streak stage, whereas POM forms a microcrystalline network, lowering the energy barrier for stretching-induced recrystallization. These findings could provide theoretical guidelines on selecting compatibilizers for different PLLA based blends.
Collapse
Affiliation(s)
- Haoqi Luo
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China
| | - Xiangyan Yang
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China
| | - Qingyi Ding
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China
| | - Bogang Sheng
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China
| | - Jing Deng
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China
| | - Xiaofei Yan
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China
| | - Yang Wu
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China
| | - Han Chen
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Chaowei Hao
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Shuaishuai Yuan
- Key Lab of Biobased Polymer Materials of Shandong Provincial Education Department, College of Polymer Science and Engineering, Qingdao University of Science and Technology, 266042 Qingdao, PR China.
| | - Jianrong Zeng
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, 201204 Shanghai, PR China; Shanghai Institute of Applied Physics, Chinese Academy of Sciences, 201800 Shanghai, PR China.
| | - Weihua Zhou
- Department of Polymer Materials and Engineering, School of Physics and Materials, Nanchang University, 330031 Nanchang, PR China.
| |
Collapse
|
16
|
Wang H, Shu Z, Chen P, Su J, Zhu H, Jiang J, Yan C, Xiao J, Shi Y. Laser powder bed fusion printed poly-ether-ether-ketone/bioactive glass composite scaffolds with dual-scale pores for enhanced osseointegration and bone ingrowth. Acta Biomater 2024; 189:605-620. [PMID: 39389225 DOI: 10.1016/j.actbio.2024.09.055] [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: 07/16/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Although poly-ether-ether-ketone (PEEK) implants hold significant medical promise, their bioinert nature presents challenges in osseointegration and bone ingrowth within clinical contexts. To mitigate these challenges, the present study introduces Diamond PEEK/bioactive glass (BG) composite scaffolds, characterized by macro/micro dual-porous structures, precisely fabricated via laser powder bed fusion (LPBF) technology. The findings indicate that an increase in BG content within these scaffolds significantly augments their hydrophilicity and hydroxyapatite formation capacities. Stress-strain curve analysis demonstrates reliable load-bearing stability across all scaffold types. In vitro assessments confirmed the non-cytotoxicity of PEEK/BG samples and demonstrated improved osteogenic differentiation and mineralization with increased BG incorporation. Further, in vivo experiments illustrated that the Diamond porous structure of these scaffolds facilitated bone growth, an effect notably amplified with higher BG content. Particularly in groups with 15 wt.% and 25 wt.% BG scaffolds, new bone formation was observed not only within the macropores of the Diamond structure but also within the micropores inside the scaffold rod, suggesting an almost seamless fusion with the new bone. This demonstrates the scaffolds' effective osteointegration and bone ingrowth properties. This study conclusively established the effectiveness of Diamond-structured PEEK/BG composite scaffolds, fabricated via LPBF, in bone repair. It highlights the crucial role of BG in enhancing osteogenic potential through interaction with the macro/micro pores of the scaffold. STATEMENT OF SIGNIFICANCE: This study addresses the bioinert nature of PEEK implants by developing Diamond-structured PEEK/bioactive glass (BG) composite scaffolds by laser powder bed fusion. The dual-porous macro/microstructure enhances hydrophilicity and hydroxyapatite formation, vital for bone regeneration. By adjusting the BG content, we controlled the melt viscosity and sintering rate, leading to the formation of beneficial microscale pores. These pores resolve the issue of ineffective bioactive fillers in previous LPBF-fabricated scaffolds, enhancing the osteogenic potential of BG and inducing superior bone ingrowth and osseointegration. In vitro and in vivo analyses show enhanced osteogenic differentiation, mineralization, and bone growth, underscoring the clinical potential of these scaffolds for bone repair.
Collapse
Affiliation(s)
- Haoze Wang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zixing Shu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Chen
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hao Zhu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiawei Jiang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chunze Yan
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Jun Xiao
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
17
|
Chen MC, Chang CC, Wu CL, Chiang PM, Yeh CC, Chen YH, Sheu MT. Augmenting dermal collagen synthesis through hyaluronic acid-based microneedle-mediated delivery of poly(l-lactic acid) microspheres. Int J Biol Macromol 2024; 281:136311. [PMID: 39370068 DOI: 10.1016/j.ijbiomac.2024.136311] [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: 08/11/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Poly(L-lactic acid) (PLLA) can stimulate collagen synthesis through a foreign body response. However, inappropriate injection techniques and localized PLLA clustering can lead to complications and adverse events. This study developed a composite microneedle (MN) device comprising an array of PLLA microsphere (PLLA MP)-loaded hyaluronic acid needle tips with a supporting patch (PLLA MP-MN). This device was designed to deliver PLLA MPs precisely and uniformly to the dermis and to provide dual stimulation through MN puncture and MP implantation, thereby enabling the rapid and long-lasting regeneration of dermal collagen. When applied to rat skin, the MN array evenly distributed the PLLA MPs throughout the penetrated regions, which prevented local PLLA overdosing and elicited a milder inflammatory response compared with that induced by intradermal PLLA MP injections. An in vivo efficacy study revealed that MN-mediated delivery of PLLA MPs not only promptly initiated collagen production through microwound-triggered wound-healing cascades in the early treatment stage but also enabled the long-term stimulation of collagen deposition through MP-induced foreign body reactions, thereby significantly enhancing neocollagenesis. This innovative PLLA MP-MN system can augment the benefits and minimize the adverse effects associated with traditional PLLA fillers, providing a safe and reliable anti-aging therapeutic option.
Collapse
Affiliation(s)
- Mei-Chin Chen
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan.
| | - Chih-Chi Chang
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Lin Wu
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Min Chiang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chien Yeh
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hung Chen
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
18
|
Yu H, Luo X, Li Y, Shao L, Yang F, Pang Q, Zhu Y, Hou R. Advanced Hybrid Strategies of GelMA Composite Hydrogels in Bone Defect Repair. Polymers (Basel) 2024; 16:3039. [PMID: 39518248 PMCID: PMC11548276 DOI: 10.3390/polym16213039] [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: 09/21/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
To date, severe bone defects remain a significant challenge to the quality of life. All clinically used bone grafts have their limitations. Bone tissue engineering offers the promise of novel bone graft substitutes. Various biomaterial scaffolds are fabricated by mimicking the natural bone structure, mechanical properties, and biological properties. Among them, gelatin methacryloyl (GelMA), as a modified natural biomaterial, possesses a controllable chemical network, high cellular stability and viability, good biocompatibility and degradability, and holds the prospect of a wide range of applications. However, because they are hindered by their mechanical properties, degradation rate, and lack of osteogenic activity, GelMA hydrogels need to be combined with other materials to improve the properties of the composites and endow them with the ability for osteogenesis, vascularization, and neurogenesis. In this paper, we systematically review and summarize the research progress of GelMA composite hydrogel scaffolds in the field of bone defect repair, and discuss ways to improve the properties, which will provide ideas for the design and application of bionic bone substitutes.
Collapse
Affiliation(s)
- Han Yu
- Department of Cell Biology and Regenerative Medicine, Health Science Center, Ningbo University, Ningbo 315211, China; (H.Y.); (X.L.); (Y.L.); (F.Y.); (Y.Z.)
| | - Xi Luo
- Department of Cell Biology and Regenerative Medicine, Health Science Center, Ningbo University, Ningbo 315211, China; (H.Y.); (X.L.); (Y.L.); (F.Y.); (Y.Z.)
| | - Yanling Li
- Department of Cell Biology and Regenerative Medicine, Health Science Center, Ningbo University, Ningbo 315211, China; (H.Y.); (X.L.); (Y.L.); (F.Y.); (Y.Z.)
| | - Lei Shao
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, China;
| | - Fang Yang
- Department of Cell Biology and Regenerative Medicine, Health Science Center, Ningbo University, Ningbo 315211, China; (H.Y.); (X.L.); (Y.L.); (F.Y.); (Y.Z.)
| | - Qian Pang
- Department of Cell Biology and Regenerative Medicine, Health Science Center, Ningbo University, Ningbo 315211, China; (H.Y.); (X.L.); (Y.L.); (F.Y.); (Y.Z.)
| | - Yabin Zhu
- Department of Cell Biology and Regenerative Medicine, Health Science Center, Ningbo University, Ningbo 315211, China; (H.Y.); (X.L.); (Y.L.); (F.Y.); (Y.Z.)
| | - Ruixia Hou
- Department of Cell Biology and Regenerative Medicine, Health Science Center, Ningbo University, Ningbo 315211, China; (H.Y.); (X.L.); (Y.L.); (F.Y.); (Y.Z.)
| |
Collapse
|
19
|
Zhang Y, Zhang J, Yang Q, Song Y, Pan M, Kan Y, Xiang L, Li M, Zeng H. Tuning interfacial molecular asymmetry to engineer protective coatings with superior surface anchoring, antifouling and antibacterial properties. Acta Biomater 2024:S1742-7061(24)00598-1. [PMID: 39395705 DOI: 10.1016/j.actbio.2024.10.015] [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: 05/29/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
Multifunctional robust protective coatings that combine biocompatibility, antifouling and antimicrobial properties play an essential role in reducing host reactions and infection on invasive medical devices. However, developing these protective coatings generally faces a paradox: coating materials capable of achieving robust adhesion to substrates via spontaneous deposition inevitably initiate continuous biofoulant adsorption, while those employing strong hydration capability to resist biofoulant attachment have limited substrate binding ability and durability under wear. Herein, we designed a multifunctional terpolymer of poly(dopamine methyacrylamide-co-2-methacryloyloxyethyl phoasphorylcholine-co-2-(dimethylamino)-ethyl methacrylate) (P(DMA-co-MPC-co-DMAEMA)), which integrates desired yet traditionally incompatible functions (i.e., robust adhesion, antifouling, lubrication, and antimicrobial properties). Direct normal and lateral force measurements, dynamic adsorption tests, surface ion conductance mapping were applied to comprehensively investigate the nanomechanics of coating-biofloulant interactions. Catechol groups of DMA act as basal anchors for robust substrate deposition, while the highly hydrated zwitterion of MPC provides apical protection to resist biofouling and wear. Moreover, the antimicrobial property is conferred through the protonation of tertiary amine groups on DMAEMA, inhibiting infection under physiological conditions. This work provides an effective strategy for harmonizing demanded yet incompatible properties in one coating material, with significant implications for the development of multifunctional surfaces towards the advancement of invasive biomedical devices. STATEMENT OF SIGNIFICANCE: Multifunctional robust protective coatings have been widely utilized in invasive medical devices to mitigate host responses and infection. However, modified surface coatings often encounter a trade-off between robust adhesion to substrates and strong hydration capability for antifouling and antimicrobial properties. We propose a universal strategy for surface modification by dopamine-assisted co-deposition with a multifunctional terpolymer of P(DMA-co-MPC-co-DMAEMA) that simultaneously achieves robust adhesion, antifouling, and antimicrobial properties. Through elucidating the nanomechanics with fundamentally understanding the interactions between the coating and biomacromolecules, we highlight the role of DMA for substrate adhesion, MPC for biofouling resistance, and DMAEMA for antimicrobial activity. This approach presents a promising strategy for constructing multifunctional coatings on minimally invasive medical devices by tuning interfacial molecular asymmetricity to reconcile incompatible properties within one coating.
Collapse
Affiliation(s)
- Yuhao Zhang
- School of Mechanical Engineering, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, China
| | - Jiawen Zhang
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, China
| | - Qiang Yang
- School of Mechanical Engineering, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, China
| | - Yao Song
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostic, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Mingfei Pan
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostic, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Yajing Kan
- School of Mechanical Engineering, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, China
| | - Li Xiang
- School of Mechanical Engineering, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, China.
| | - Mei Li
- National Demonstration Center for Experimental Basic Medical Education, Nanjing Medical University, Nanjing 211166, China.
| | - Hongbo Zeng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, T6G 1H9, Canada.
| |
Collapse
|
20
|
Wang YC, Cai MT, Chen MH, Tung FI, Chen MH, Liu TY. Europium-Containing Nanospheres for Treating Ovariectomy-Induced Osteoporosis: Targeted Bone Remodeling and Macrophage Polarization Modulation. Int J Nanomedicine 2024; 19:10145-10163. [PMID: 39386058 PMCID: PMC11463175 DOI: 10.2147/ijn.s472253] [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: 04/06/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Purpose Osteoporosis, characterized by reduced bone mass and structural deterioration, poses a significant healthcare challenge. Traditional treatments, while effective in reducing fracture risks, are often limited by side effects. This study introduces a novel nanocomplex, europium (Eu) ions-doped superparamagnetic iron oxide (SPIO) nanocrystals encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanospheres, abbreviated as SPIO:Eu@PLGA nanospheres, as a potential therapeutic agent for osteoporosis by modulating macrophage polarization, enhancing osteoblast differentiation and inhibiting osteoclastogenesis. Methods SPIO and SPIO:Eu nanocrystals were synthesized through pyrolysis and encapsulated in PLGA using an emulsification method. To evaluate the impact of SPIO:Eu@PLGA nanospheres on macrophage reprogramming and reactive oxygen species (ROS) production, flow cytometry analysis was conducted. Furthermore, an ovariectomized (OVX) rat model was employed to assess the therapeutic efficacy of SPIO:Eu@PLGA nanospheres in preventing the deterioration of osteoporosis. Results In vitro, SPIO:Eu@PLGA nanospheres significantly attenuated M1 macrophage activation induced by lipopolysaccharides, promoting a shift towards the M2 phenotype. This action is linked to the modulation of ROS and the NF-κB pathway. Unlike free Eu ions, which do not achieve similar results when not incorporated into the SPIO nanocrystals. SPIO:Eu@PLGA nanospheres enhanced osteoblast differentiation and matrix mineralization while inhibiting RANKL-induced osteoclastogenesis. In vivo studies demonstrated that SPIO:Eu@PLGA nanospheres effectively targeted trabecular bone surfaces in OVX rats under magnetic guidance, preserving their structure and repairing trabecular bone loss by modulating macrophage polarization, thus restoring bone remodeling homeostasis. The study underscores the critical role of Eu doping in boosting the anti-osteoporotic effects of SPIO:Eu@PLGA nanospheres, evident at both cellular and tissue levels in vitro and in vivo. Conclusion The inclusion of Eu into SPIO matrix suggests a novel approach for developing more effective osteoporosis treatments, particularly for conditions induced by OVX. This research provides essential insights into SPIO:Eu@PLGA nanospheres as an innovative osteoporosis treatment, addressing the limitations of conventional therapies through targeted delivery and macrophage polarization modulation.
Collapse
Grants
- the National Science and Technology Council (NSTC 111-2221-E-A49-051-MY2, NSTC 111-2811-E-A49A-007-MY2, NSTC 111-2314-B-038-094, NSTC 113-2314-B-A49-065-MY3, NSTC 113-2811-B-A49A-029), the Far Eastern Memorial Hospital (FEMH-2024-C-013, FEMH-2024-C-057, FEMH-2023-C-081), and the Department of Health, Taipei City Government (11201-62-004, 11301-62-048) for financial support
Collapse
Affiliation(s)
- Yu-Chi Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Meng-Ting Cai
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Ming-Hong Chen
- Division of Neurosurgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, 220216, Taiwan
- Department of Electrical Engineering, Yuan Ze University, Taoyuan City, 320315, Taiwan
| | - Fu-I Tung
- Department of Orthopaedics, Yang-Ming Branch, Taipei City Hospital, Taipei, 111024, Taiwan
- Department of Health and Welfare, College of City Management, University of Taipei, Taipei, 111036, Taiwan
| | - Mei-Hsiu Chen
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, 220216, Taiwan
- Department of Biomedical Engineering, Ming Chuang University, Taoyuan, 333, Taiwan
| | - Tse-Ying Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| |
Collapse
|
21
|
An X, Ma C, Gong L, Liu C, Li N, Liu Z, Li X. Ionic-physical-chemical triple cross-linked all-biomass-based aerogel for thermal insulation applications. J Colloid Interface Sci 2024; 668:678-690. [PMID: 38710124 DOI: 10.1016/j.jcis.2024.04.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/08/2024]
Abstract
Aerogels, as a unique porous material, are expected to be used as insulation materials to solve the global environmental and energy crisis. Using chitosan, citric acid, pectin and phytic acid as raw materials, an all-biomass-based aerogel with high modulus was prepared by the triple strategy of ionic, physical and chemical cross-linking through directional freezing technique. Based on this three-dimensional network, the aerogel exhibited excellent compressive modulus (24.89 ± 1.76 MPa) over a wide temperature range and thermal insulation properties. In the presence of chitosan, citric acid and phytic acid, the aerogel obtained excellent fire safety (LOI value up to 31.2%) and antibacterial properties (antibacterial activity against Staphylococcus aureus and Escherichia coli reached 81.98% and 67.43%). In addition, the modified aerogel exhibited excellent hydrophobicity (hydrophobic angle of 146°) and oil-water separation properties. More importantly, the aerogel exhibited a biodegradation rate of up to 40.31% for 35 days due to its all-biomass nature. This work provides a green and sustainable strategy for the production of highly environmentally friendly thermal insulation materials with high strength, flame retardant, antibacterial and hydrophobic properties.
Collapse
Affiliation(s)
- Xinyu An
- Material Science and Engineering College, Northeast Forestry University, Harbin 150040, China
| | - Chang Ma
- Material Science and Engineering College, Northeast Forestry University, Harbin 150040, China
| | - Ling Gong
- Material Science and Engineering College, Northeast Forestry University, Harbin 150040, China
| | - Chang Liu
- Material Science and Engineering College, Northeast Forestry University, Harbin 150040, China
| | - Ning Li
- Material Science and Engineering College, Northeast Forestry University, Harbin 150040, China
| | - Zhiming Liu
- Material Science and Engineering College, Northeast Forestry University, Harbin 150040, China.
| | - Xu Li
- Material Science and Engineering College, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
22
|
Hao D, Zhang Y, Ding Y, Yan Q. Preparation and properties of silver-carrying nano-titanium dioxide antimicrobial agents and silicone composite. Sci Rep 2024; 14:18870. [PMID: 39143137 PMCID: PMC11324888 DOI: 10.1038/s41598-024-69787-5] [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: 06/12/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
The characteristics of dopamine self-polymerization were used to cover the nano-titanium dioxide (TiO2) surface and produce nano-titanium dioxide-polydopamine (TiO2-PDA). The reducing nature of dopamine was then used to reduce silver nitrate to silver elemental particles on the modified nano-titanium dioxide: The resulting TiO2-PDA-Ag nanoparticles were used as antimicrobial agents. Finally, the antibacterial agent was mixed with silicone to obtain an antibacterial silicone composite material. The composition and structure of antibacterial agents were analyzed by scanning electron microscopy, transmission electron microscopy, X-ray photoelectron energy spectroscopy, and X-ray diffraction. Microscopy and the antibacterial properties of the silicone antibacterial composites were studied as well. The TiO2-PDA-Ag antimicrobial agent had good dispersion versus nano-TiO2. The three were strongly combined with obvious characteristic peaks. The antibacterial agents were evenly dispersed in silicone, and the silicone composite has excellent antibacterial properties. Bacillus subtilis (B. subtilis) adhesion was reduced from 246 × 104 cfu/cm2 to 2 × 104 cfu/cm2, and colibacillus (E. coli) reduced from 228 × 104 cfu/cm2 leading to bacteria-free adhesion.
Collapse
Affiliation(s)
- Dongdong Hao
- Changzhou University Huaide College, Jingjiang, 214500, China
| | - Yuxuan Zhang
- Changzhou Vocational Institute of Textile and Garment, Changzhou, 213000, China.
| | | | - Qiuyu Yan
- Changzhou University Huaide College, Jingjiang, 214500, China
| |
Collapse
|
23
|
Tang X, Wang Y, Liu N, Deng X, Zhou Z, Yu C, Wang Y, Fang K, Wu T. Methacrylated Carboxymethyl Chitosan Scaffold Containing Icariin-Loaded Short Fibers for Antibacterial, Hemostasis, and Bone Regeneration. ACS Biomater Sci Eng 2024; 10:5181-5193. [PMID: 38935742 DOI: 10.1021/acsbiomaterials.4c00707] [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: 06/29/2024]
Abstract
Bone defects typically result in bone nonunion, delayed or nonhealing, and localized dysfunction, and commonly used clinical treatments (i.e., autologous and allogeneic grafts) have limited results. The multifunctional bone tissue engineering scaffold provides a new treatment for the repair of bone defects. Herein, a three-dimensional porous composite scaffold with stable mechanical support, effective antibacterial and hemostasis properties, and the ability to promote the rapid repair of bone defects was synthesized using methacrylated carboxymethyl chitosan and icariin-loaded poly-l-lactide/gelatin short fibers (M-CMCS-SFs). Icariin-loaded SFs in the M-CMCS scaffold resulted in the sustained release of osteogenic agents, which was beneficial for mechanical reinforcement. Both the porous structure and the use of chitosan facilitate the effective absorption of blood and fluid exudates. Moreover, its superior antibacterial properties could prevent the occurrence of inflammation and infection. When cultured with bone mesenchymal stem cells, the composite scaffold showed a promotion in osteogenic differentiation. Taken together, such a multifunctional composite scaffold showed comprehensive performance in antibacterial, hemostasis, and bone regeneration, thus holding promising potential in the repair of bone defects and related medical treatments.
Collapse
Affiliation(s)
- Xunmeng Tang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
| | - Yawen Wang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China
| | - Na Liu
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xinyuan Deng
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
| | - Ziyi Zhou
- Department of Plastic, Reconstructive and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Chenghao Yu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China
| | - Yuanfei Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China
| | - Kuanjun Fang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
- Laboratory for Manufacturing Low Carbon and Functionalized Textiles in the Universities of Shandong Province, State Key Laboratory for Biofibers and Eco-textiles, College of Textiles & Clothing, Qingdao University, Qingdao 266071, China
| | - Tong Wu
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China
| |
Collapse
|
24
|
Pan S, Zou Z, Zhou X, Wei J, Liu H, Su Z, Liao G, Huang G, Huang Z, Xu Y, Lu M, Gu R. Therapeutic impacts of GNE‑477‑loaded H 2O 2 stimulus‑responsive dodecanoic acid‑phenylborate ester‑dextran polymeric micelles on osteosarcoma. Int J Mol Med 2024; 54:69. [PMID: 38940336 PMCID: PMC11232662 DOI: 10.3892/ijmm.2024.5393] [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: 12/19/2023] [Accepted: 04/25/2024] [Indexed: 06/29/2024] Open
Abstract
Osteosarcoma (OS) is a highly malignant primary bone neoplasm that is the leading cause of cancer‑associated death in young people. GNE‑477 belongs to the second generation of mTOR inhibitors and possesses promising potential in the treatment of OS but dose tolerance and drug toxicity limit its development and utilization. The present study aimed to prepare a novel H2O2 stimulus‑responsive dodecanoic acid (DA)‑phenylborate ester‑dextran (DA‑B‑DEX) polymeric micelle delivery system for GNE‑477 and evaluate its efficacy. The polymer micelles were characterized by morphology, size and critical micelle concentration. The GNE‑477 loaded DA‑B‑DEX (GNE‑477@DBD) tumor‑targeting drug delivery system was established and the release of GNE‑477 was measured. The cellular uptake of GNE‑477@DBD by three OS cell lines (MG‑63, U2OS and 143B cells) was analyzed utilizing a fluorescent tracer technique. The hydroxylated DA‑B was successfully grafted onto dextran at a grafting rate of 3%, suitable for forming amphiphilic micelles. Following exposure to H2O2, the DA‑B‑DEX micelles ruptured and released the drug rapidly, leading to increased uptake of GNE‑477@DBD by cells with sustained release of GNE‑477. The in vitro experiments, including MTT assay, flow cytometry, western blotting and RT‑qPCR, demonstrated that GNE‑477@DBD inhibited tumor cell viability, arrested cell cycle in G1 phase, induced apoptosis and blocked the PI3K/Akt/mTOR cascade response. In vivo, through the observation of mice tumor growth and the results of H&E staining, the GNE‑477@DBD group exhibited more positive therapeutic outcomes than the free drug group with almost no adverse effects on other organs. In conclusion, H2O2‑responsive DA‑B‑DEX presents a promising delivery system for hydrophobic anti‑tumor drugs for OS therapy.
Collapse
Affiliation(s)
- Songmu Pan
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Zhuan Zou
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Xiaofeng Zhou
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Jiyong Wei
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Huijiang Liu
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Zhongyi Su
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Gui Liao
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Guangyu Huang
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Zonggui Huang
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Yi Xu
- Department of Pharmacy, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Minan Lu
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Ronghe Gu
- Department of Orthopedic Surgery, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
- Guangxi Key Laboratory of Intelligent Precision Medicine, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| |
Collapse
|
25
|
Negi D, Bhavya K, Pal D, Singh Y. Acemannan coated, cobalt-doped biphasic calcium phosphate nanoparticles for immunomodulation regulated bone regeneration. Biomater Sci 2024; 12:3672-3685. [PMID: 38864476 DOI: 10.1039/d4bm00482e] [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: 06/13/2024]
Abstract
Biomaterials are used as scaffolds in bone regeneration to facilitate the restoration of bone tissues. The local immune microenvironment affects bone repair but the role of immune response in biomaterial-facilitated osteogenesis has been largely overlooked and it presents a major knowledge gap in the field. Nanomaterials that can modulate M1 to M2 macrophage polarization and, thus, promote bone repair are known. This study investigates a novel approach to accelerate bone healing by using acemannan coated, cobalt-doped biphasic calcium phosphate nanoparticles to promote osteogenesis and modulate macrophage polarization to provide a prohealing microenvironment for bone regeneration. Different concentrations of cobalt were doped in biphasic calcium phosphate nanoparticles, which were further coated with acemannan polymer and characterized. The nanoparticles showed >90% cell viability and enhanced cell proliferation along with osteogenic differentiation as demonstrated by the enhanced alkaline phosphatase activity and osteogenic calcium deposition. The morphology of MC3T3-E1 cells remained unchanged even after treatment with nanoparticles. Acemannan coated nanoparticles were also able to decrease the expression of M1 markers, iNOS, and CD68 and enhance the expression of M2 markers, CD206, CD163, and Arg-1 as indicated by RT-qPCR, flow cytometry, and ICC studies. The findings show that acemannan coated nanoparticles can create a supportive immune milieu by inducing and promoting the release of osteogenic markers, and by causing a reduction in inflammatory markers, thus helping in efficient bone regeneration. As per our knowledge, this is the first study showing the combined effect of acemannan and cobalt for bone regeneration using immunomodulation. The work presents a novel approach for enhancing osteogenesis and macrophage polarization, thus, offering a potent strategy for effective bone regeneration.
Collapse
Affiliation(s)
- Deepa Negi
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar-140 001, Punjab, India.
| | - Kumari Bhavya
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar-140 001, Punjab, India.
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar-140 001, Punjab, India.
| | - Yashveer Singh
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar-140 001, Punjab, India.
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar-140 001, Punjab, India
| |
Collapse
|
26
|
Du H, Li B, Yu R, Lu X, Li C, Zhang H, Yang F, Zhao R, Bao W, Yin X, Wang Y, Zhou J, Xu J. ETV2 regulating PHD2-HIF-1α axis controls metabolism reprogramming promotes vascularized bone regeneration. Bioact Mater 2024; 37:222-238. [PMID: 38549772 PMCID: PMC10973785 DOI: 10.1016/j.bioactmat.2024.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/23/2024] [Accepted: 02/11/2024] [Indexed: 01/05/2025] Open
Abstract
The synchronized development of mineralized bone and blood vessels is a fundamental requirement for successful bone tissue regeneration. Adequate energy production forms the cornerstone supporting new bone formation. ETS variant 2 (ETV2) has been identified as a transcription factor that promotes energy metabolism reprogramming and facilitates the coordination between osteogenesis and angiogenesis. In vitro molecular experiments have demonstrated that ETV2 enhances osteogenic differentiation of dental pulp stem cells (DPSCs) by regulating the ETV2- prolyl hydroxylase 2 (PHD2)- hypoxia-inducible factor-1α (HIF-1α)- vascular endothelial growth factor A (VEGFA) axis. Notably, ETV2 achieves the rapid reprogramming of energy metabolism by simultaneously accelerating mitochondrial aerobic respiration and glycolysis, thus fulfilling the energy requirements essential to expedite osteogenic differentiation. Furthermore, decreased α-ketoglutarate release from ETV2-modified DPSCs contributes to microcirculation reconstruction. Additionally, we engineered hydroxyapatite/chitosan microspheres (HA/CS MS) with biomimetic nanostructures to facilitate multiple ETV2-DPSC functions and further enhanced the osteogenic differentiation. Animal experiments have validated the synergistic effect of ETV2-modified DPSCs and HA/CS MS in promoting the critical-size bone defect regeneration. In summary, this study offers a novel treatment approach for vascularized bone tissue regeneration that relies on energy metabolism activation and the maintenance of a stable local hypoxia signaling state.
Collapse
Affiliation(s)
- HaoRan Du
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Bang Li
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Rui Yu
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Xiaoxuan Lu
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - ChengLin Li
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - HuiHui Zhang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Fan Yang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - RongQuan Zhao
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - WeiMin Bao
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Xuan Yin
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - YuanYin Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Jian Zhou
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
- Department of VIP Dental Service, School of Stomatology, Capital Medical University, Beijing, 100050, China
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianguang Xu
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| |
Collapse
|
27
|
Wu X, Li W, Herlah L, Koch M, Wang H, Schirhagl R, Włodarczyk-Biegun MK. Melt electrowritten poly-lactic acid /nanodiamond scaffolds towards wound-healing patches. Mater Today Bio 2024; 26:101112. [PMID: 38873104 PMCID: PMC11170272 DOI: 10.1016/j.mtbio.2024.101112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/18/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
Multifunctional wound dressings, enriched with biologically active agents for preventing or treating infections and promoting wound healing, along with cell delivery capability, are highly needed. To address this issue, composite scaffolds with potential in wound dressing applications were fabricated in this study. The poly-lactic acid/nanodiamonds (PLA/ND) scaffolds were first printed using melt electrowriting (MEW) and then coated with quaternized β-chitin (QβC). The NDs were well-dispersed in the printed filaments and worked as fillers and bioactive additions to PLA material. Additionally, they improved coating effectiveness due to the interaction between their negative charges (from NDs) and positive charges (from QβC). NDs not only increased the thermal stability of PLA but also benefitted cellular behavior and inhibited the growth of bacteria. Scaffolds coated with QβC increased the effect of bacteria growth inhibition and facilitated the proliferation of human dermal fibroblasts. Additionally, we have observed rapid extracellular matrix (ECM) remodeling on QβC-coated PLA/NDs scaffolds. The scaffolds provided support for cell adhesion and could serve as a valuable tool for delivering cells to chronic wound sites. The proposed PLA/ND scaffold coated with QβC holds great potential for achieving fast healing in various types of wounds.
Collapse
Affiliation(s)
- Xixi Wu
- Department of Biomedical Engineering, University Medical Centre, Ant. Deusinglaan 1, 9713, AW, Groningen, the Netherlands
- Polymer Science, Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
| | - Wenjian Li
- Advanced Production Engineering, Engineering and Technology Institute of Groningen, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
| | - Lara Herlah
- Department of Biomedical Engineering, University Medical Centre, Ant. Deusinglaan 1, 9713, AW, Groningen, the Netherlands
| | - Marcus Koch
- INM – Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Hui Wang
- Nanostructured Materials and Interfaces, Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
| | - Romana Schirhagl
- Department of Biomedical Engineering, University Medical Centre, Ant. Deusinglaan 1, 9713, AW, Groningen, the Netherlands
| | - Małgorzata K. Włodarczyk-Biegun
- Polymer Science, Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
- Biotechnology Centre, The Silesian University of Technology, Krzywoustego 8, 44-100, Gliwice, Poland
| |
Collapse
|
28
|
Liu Z, Li S, Xu Z, Li L, Liu Y, Gao X, Diao Y, Chen L, Sun J. Preparation and Characterization of Carboxymethyl Chitosan/Sodium Alginate Composite Hydrogel Scaffolds Carrying Chlorhexidine and Strontium-Doped Hydroxyapatite. ACS OMEGA 2024; 9:22230-22239. [PMID: 38799338 PMCID: PMC11112597 DOI: 10.1021/acsomega.4c01237] [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: 02/07/2024] [Revised: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
Herein, we introduce a novel composite hydrogel scaffold designed for addressing infectious jaw defects, a significant challenge in clinical settings caused by the inherent limited self-regenerative capacity of bone tissues. The scaffold was engineered from a blend of carboxymethyl chitosan (CMCS)/sodium alginate (SA) hydrogel (CSH), β-cyclodextrin/chlorhexidine clathrate (β-CD-CHX), and strontium-nanohydroxyapatite nanoparticles (Sr-nHA). The β-CD-CHX and Sr-nHA components were synthesized using a saturated aqueous solution and a coprecipitation method, respectively. Subsequently, these elements were encapsulated within the CSH matrix. Comprehensive characterization of the CMCS/SA/β-CD-CHX/Sr-nHA composite hydrogel scaffold via scanning electron microscopy, X-ray photoelectron spectroscopy, and Fourier-transform infrared spectroscopy validated the successful synthesis. The swelling and in vitro degradation behaviors proved that the composite hydrogel had good physical properties, while in vitro evaluations demonstrated favorable biocompatibility and osteoinductive properties. Additionally, antibacterial assessments revealed its effectiveness against common pathogens, Staphylococcus aureus and Escherichia coli. Overall, our results indicate that the CMCS/SA/β-CD-CHX/Sr-nHA composite hydrogel scaffolds exhibit significant potential for effectively treating infection-prone jaw defects.
Collapse
Affiliation(s)
- Zijian Liu
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
| | - Shangbo Li
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
| | - Zexian Xu
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
| | - Li Li
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
| | - Yanshan Liu
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
- Dental
Digital Medicine and 3D Printing Engineering Laboratory of Qingdao, Qingdao 266003, China
| | - Xiaohan Gao
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
| | - Yaru Diao
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
| | - Liqiang Chen
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
- Dental
Digital Medicine and 3D Printing Engineering Laboratory of Qingdao, Qingdao 266003, China
- The
Climbing Peak Discipline Project of Qingdao, Qingdao 266003, China
| | - Jian Sun
- Department
of Oral and Maxillofacial Surgery, The Affiliated
Hospital of Qingdao University, Qingdao 266003, China
- School
of Stomatology, Qingdao University, Qingdao 266003, China
- Dental
Digital Medicine and 3D Printing Engineering Laboratory of Qingdao, Qingdao 266003, China
- The
Climbing Peak Discipline Project of Qingdao, Qingdao 266003, China
| |
Collapse
|
29
|
Mim JJ, Hasan M, Chowdhury MS, Ghosh J, Mobarak MH, Khanom F, Hossain N. A comprehensive review on the biomedical frontiers of nanowire applications. Heliyon 2024; 10:e29244. [PMID: 38628721 PMCID: PMC11016983 DOI: 10.1016/j.heliyon.2024.e29244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/19/2024] Open
Abstract
This comprehensive review examines the immense capacity of nanowires, nanostructures characterized by unbounded dimensions, to profoundly transform the field of biomedicine. Nanowires, which are created by combining several materials using techniques such as electrospinning and vapor deposition, possess distinct mechanical, optical, and electrical properties. As a result, they are well-suited for use in nanoscale electronic devices, drug delivery systems, chemical sensors, and other applications. The utilization of techniques such as the vapor-liquid-solid (VLS) approach and template-assisted approaches enables the achievement of precision in synthesis. This precision allows for the customization of characteristics, which in turn enables the capability of intracellular sensing and accurate drug administration. Nanowires exhibit potential in biomedical imaging, neural interfacing, and tissue engineering, despite obstacles related to biocompatibility and scalable manufacturing. They possess multifunctional capabilities that have the potential to greatly influence the intersection of nanotechnology and healthcare. Surmounting present obstacles has the potential to unleash the complete capabilities of nanowires, leading to significant improvements in diagnostics, biosensing, regenerative medicine, and next-generation point-of-care medicines.
Collapse
Affiliation(s)
- Juhi Jannat Mim
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Mehedi Hasan
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Md Shakil Chowdhury
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Jubaraz Ghosh
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Md Hosne Mobarak
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Fahmida Khanom
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Nayem Hossain
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| |
Collapse
|
30
|
Zhao W, Xu F, Shen Y, Ding Q, Wang Y, Liang L, Dai W, Chen Y. Temporal control in shell-core structured nanofilm for tracheal cartilage regeneration: synergistic optimization of anti-inflammation and chondrogenesis. Regen Biomater 2024; 11:rbae040. [PMID: 38769993 PMCID: PMC11105955 DOI: 10.1093/rb/rbae040] [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: 01/11/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 05/22/2024] Open
Abstract
Cartilage tissue engineering offers hope for tracheal cartilage defect repair. Establishing an anti-inflammatory microenvironment stands as a prerequisite for successful tracheal cartilage restoration, especially in immunocompetent animals. Hence, scaffolds inducing an anti-inflammatory response before chondrogenesis are crucial for effectively addressing tracheal cartilage defects. Herein, we develop a shell-core structured PLGA@ICA-GT@KGN nanofilm using poly(lactic-co-glycolic acid) (PLGA) and icariin (ICA, an anti-inflammatory drug) as the shell layer and gelatin (GT) and kartogenin (KGN, a chondrogenic factor) as the core via coaxial electrospinning technology. The resultant PLGA@ICA-GT@KGN nanofilm exhibited a characteristic fibrous structure and demonstrated high biocompatibility. Notably, it showcased sustained release characteristics, releasing ICA within the initial 0 to 15 days and gradually releasing KGN between 11 and 29 days. Subsequent in vitro analysis revealed the potent anti-inflammatory capabilities of the released ICA from the shell layer, while the KGN released from the core layer effectively induced chondrogenic differentiation of bone marrow stem cells (BMSCs). Following this, the synthesized PLGA@ICA-GT@KGN nanofilms were loaded with BMSCs and stacked layer by layer, adhering to a 'sandwich model' to form a composite sandwich construct. This construct was then utilized to repair circular tracheal defects in a rabbit model. The sequential release of ICA and KGN facilitated by the PLGA@ICA-GT@KGN nanofilm established an anti-inflammatory microenvironment before initiating chondrogenic induction, leading to effective tracheal cartilage restoration. This study underscores the significance of shell-core structured nanofilms in temporally regulating anti-inflammation and chondrogenesis. This approach offers a novel perspective for addressing tracheal cartilage defects, potentially revolutionizing their treatment methodologies.
Collapse
Affiliation(s)
- Wen Zhao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of Thoracic Surgery, Tongren Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200050, China
| | - Fanglan Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yumei Shen
- Operation Room Department, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Qifeng Ding
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yifei Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Leilei Liang
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, 310005, China
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Yongbing Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| |
Collapse
|
31
|
Meng C, Liu X, Li J. Hierarchical porous PLLA/ACP fibrous membrane towards bone tissue scaffold. J Mech Behav Biomed Mater 2024; 152:106455. [PMID: 38335647 DOI: 10.1016/j.jmbbm.2024.106455] [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: 12/11/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Electrospun fibres have emerged as vital components in developing tissue engineering scaffolds. Calcium phosphate-based materials, renowned for their bioactivity and biocompatibility, have garnered considerable attention in biomedical applications. This study focuses on the incorporation of amorphous calcium phosphate (ACP) nanoparticles into poly(L-lactic acid) (PLLA) to produce electrospun PLLA/ACP fibrous membranes. Subsequent treatment with acetone yielded a hierarchical porous structure, boasting an ultra-high surface area of 94.7753 ± 0.3884 m2/g. The ACP nanoparticles, initially encapsulated by PLLA, were exposed on the fibre surface after acetone treatment. Furthermore, the porous PLLA/ACP fibrous membrane exhibited superior mechanical properties (Young's modulus = 0.148 GPa, tensile strength = 3.05 MPa) and enhanced wettability. In a 7-day in vitro cell culture with human osteoblast-like cells, the porous PLLA/ACP fibrous membrane demonstrated a significant improvement in osteoblast adhesion and proliferation, with a proliferation rate increase of 252.0% and 298.7% at day 4 and day 7, respectively. These findings underscore the potential of the porous PLLA/ACP fibrous membrane as a promising candidate for bone tissue scaffolds.
Collapse
Affiliation(s)
- Chen Meng
- Department of Materials, The University of Manchester, Manchester, M13 9PL, UK
| | - Xuzhao Liu
- Department of Materials, The University of Manchester, Manchester, M13 9PL, UK; Photon Science Institute, The University of Manchester, Manchester, M13 9PL, UK
| | - Jiashen Li
- Department of Materials, The University of Manchester, Manchester, M13 9PL, UK.
| |
Collapse
|
32
|
Escobar Jaramillo M, Covarrubias C, Patiño González E, Ossa Orozco CP. Optimization by mixture design of chitosan/multi-phase calcium phosphate/BMP-2 biomimetic scaffolds for bone tissue engineering. J Mech Behav Biomed Mater 2024; 152:106423. [PMID: 38290393 DOI: 10.1016/j.jmbbm.2024.106423] [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: 11/12/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/01/2024]
Abstract
The modulation of cell behavior during culture is one of the most important aspects of bone tissue engineering because of the necessity for a complex mechanical and biochemical environment. This study aimed to improve the physicochemical properties of chitosan/multi-phase calcium phosphate (MCaP) scaffolds using an optimized mixture design experiment and evaluate the effect of biofunctionalization of the obtained scaffolds with the bone morphogenetic protein BMP-2 on stem cell behavior. The present study evaluated the compressive strength, elastic modulus, porosity, pore diameter, and degradation in simulated body fluids and integrated these responses using desirability. The properties of the scaffolds with the best desirability (18.4% of MCaP) were: compressive strength of 23 kPa, elastic modulus of 430 kPa, pore diameter of 163 μm, porosity of 92%, and degradation of 20% after 21 days. Proliferation and differentiation experiments were conducted using dental pulp stem cells after grafting BMP-2 onto scaffolds via the carbodiimide route. These experiments showed that MCaP promoted cell proliferation and increased alkaline phosphatase activity, whereas BMP-2 enhanced cell differentiation. This study demonstrates that optimizing the composition of a mixture of chitosan and MCaP improves the physicochemical and biological properties of scaffolds, indicating that this solution is viable for application in bone tissue engineering.
Collapse
Affiliation(s)
- Mateo Escobar Jaramillo
- Grupo de Investigación en Biomateriales, Programa de Bioingeniería, Facultad de Ingeniería, Universidad de Antioquia, Medellín, Antioquia, Colombia.
| | - Cristian Covarrubias
- Laboratorio de Nanobiomateriales, Universidad de, Chile, Santiago de Chile, Chile
| | - Edwin Patiño González
- Grupo de Bioquímica Estructural de Macromoléculas, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Claudia Patricia Ossa Orozco
- Grupo de Investigación en Biomateriales, Programa de Bioingeniería, Facultad de Ingeniería, Universidad de Antioquia, Medellín, Antioquia, Colombia
| |
Collapse
|
33
|
Shi Z, Yang F, Hu Y, Pang Q, Shi L, Du T, Cao Y, Song B, Yu X, Cao Z, Ye Z, Liu C, Yu R, Chen X, Zhu Y, Pang Q. An oxidized dextran-composite self-healing coated magnesium scaffold reduces apoptosis to induce bone regeneration. Carbohydr Polym 2024; 327:121666. [PMID: 38171658 DOI: 10.1016/j.carbpol.2023.121666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024]
Abstract
Self-healing coatings have shown promise in controlling the degradation of scaffolds and addressing coating detachment issues. However, developing a self-healing coating for magnesium (Mg) possessing multiple biological functions in infectious environments remains a significant challenge. In this study, a self-healing coating was developed for magnesium scaffolds using oxidized dextran (OD), 3-aminopropyltriethoxysilane (APTES), and nano-hydroxyapatite (nHA) doped micro-arc oxidation (MHA), named OD-MHA/Mg. The results demonstrated that the OD-MHA coating effectively addresses coating detachment issues and controls the degradation of Mg in an infectious environment through self-healing mechanisms. Furthermore, the OD-MHA/Mg scaffold exhibits antibacterial, antioxidant, and anti-apoptotic properties, it also promotes bone repair by upregulating the expression of osteogenesis genes and proteins. The findings of this study indicate that the OD-MHA coated Mg scaffold possessing multiple biological functions presents a promising approach for addressing infectious bone defects. Additionally, the study showcases the potential of polysaccharides with multiple biological functions in facilitating tissue healing even in challenging environments.
Collapse
Affiliation(s)
- Zewen Shi
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China; Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fang Yang
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yiwei Hu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Qian Pang
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Lin Shi
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China
| | - Tianyu Du
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yuhao Cao
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Baiyang Song
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Xueqiang Yu
- Department of Radiology, Ningbo No. 2 Hospital, Ningbo 315000, China
| | - Zhaoxun Cao
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhewei Ye
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chen Liu
- Ningbo Branch of Chinese Academy of Ordnance Science, Ningbo 315100, China
| | - Rongyao Yu
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China
| | - Xianjun Chen
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China.
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo 315211, China.
| | - Qingjiang Pang
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
34
|
Dai Y, Xie Q, Zhang Y, Sun Y, Zhu S, Wang C, Tan Y, Gou X. Neoteric Semiembedded β-Tricalcium Phosphate Promotes Osteogenic Differentiation of Mesenchymal Stem Cells under Cyclic Stretch. ACS APPLIED MATERIALS & INTERFACES 2024; 16:8289-8300. [PMID: 38329794 DOI: 10.1021/acsami.3c15090] [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: 02/09/2024]
Abstract
β-Tricalcium phosphate (β-TCP) is a bioactive material for bone regeneration, but its brittleness limits its use as a standalone scaffold. Therefore, continuous efforts are necessary to effectively integrate β-TCP into polymers, facilitating a sturdy ion exchange for cell regulation. Herein, a novel semiembedded technique was utilized to anchor β-TCP nanoparticles onto the surface of the elastic polymer, followed by hydrophilic modification with the polymerization of dopamine. Cell adhesion and osteogenic differentiation of mesenchymal stem cells (MSCs) under static and dynamic uniaxial cyclic stretching conditions were investigated. The results showed that the new strategy was effective in promoting cell adhesion, proliferation, and osteogenic induction by the sustained release of Ca2+ in the vicinity and creating a reasonable roughness. Specifically, released Ca2+ from β-TCP could activate the calcium signaling pathway, which further upregulated calmodulin and calcium/calmodulin-dependent protein kinase II genes in MSCs. Meanwhile, the roughness of the membrane and the uniaxial cyclic stretching activated the PIEZO1 signaling pathway. Chemical and mechanical stimulation promotes osteogenic differentiation and increases the expression of related genes 2-8-fold. These findings demonstrated that the neoteric semiembedded structure was a promising strategy in controlling both chemical and mechanical factors of biomaterials for cell regulation.
Collapse
Affiliation(s)
- Yujie Dai
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Qingyun Xie
- Department of Orthopedics, General Hospital of Western Theater Command, Chengdu 610031, China
| | - Yimeng Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Yiwan Sun
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Shaomei Zhu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Chongyu Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Xue Gou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| |
Collapse
|
35
|
Patel R, Gómez-Cerezo MN, Huang H, Grøndahl L, Lu M. Degradation behaviour of porous poly(hydroxybutyrate-co-hydroxyvalerate) (PHBV) scaffolds in cell culture. Int J Biol Macromol 2024; 257:128644. [PMID: 38065444 DOI: 10.1016/j.ijbiomac.2023.128644] [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: 07/21/2023] [Revised: 11/16/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024]
Abstract
Exploring the degradation behaviour of biomaterials in a complex in vitro physiological environment can assist in predicting their performance in vivo, yet this aspect remains largely unexplored. In this study, the in vitro degradation over 12 weeks of porous poly(hydroxybutyrate-co-hydroxyvalerate) (PHBV) bone scaffolds in human osteoblast (hOB) culture was investigated. The objective was to evaluate how the presence of cells influenced both the degradation behaviour and mechanical stability of these scaffolds. The molecular weight (Mw) of the scaffolds decreased with increasing incubation time and the Mw reduction rate (6.2 ± 0.4 kg mol-1 week-1) was similar to that observed when incubated in phosphate buffered saline (PBS) solution, implying that the scaffolds underwent hydrolytic degradation in hOB culture. The mass of the scaffolds increased by 0.8 ± 0.2 % in the first 4 weeks, attributed to cells attachment and extracellular matrix (ECM) deposition including biomineralisation. During the first 8 weeks, the nominal compressive modulus, E⁎, of the scaffolds remained constant. However, it increased significantly from Week 8 to 12, with increments of 55 % and 42 % in normal and lateral directions, respectively, attributed to the reinforcement effect of cells, ECM and minerals attached on the surface of the scaffold. This study has highlighted, that while the use of PBS in degradation studies is suitable for evaluating Mw changes it cannot predict changes in mechanical properties to PHBV scaffolds in the presence of cells and culture media. Furthermore, the PHBV scaffolds had mechanical stability in cell culture for 12 weeks validating their suitability for tissue engineering applications.
Collapse
Affiliation(s)
- Rushabh Patel
- School of Mechanical and Mining Engineering, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Maria Natividad Gómez-Cerezo
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Han Huang
- School of Mechanical and Mining Engineering, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Lisbeth Grøndahl
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Mingyuan Lu
- School of Mechanical and Mining Engineering, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
36
|
Chen S, Cheng D, Bao W, Ding R, Shen Z, Huang W, Lu Y, Zhang P, Sun Y, Chen H, Shen C, Wang Y. Polydopamine-Functionalized Strontium Alginate/Hydroxyapatite Composite Microhydrogel Loaded with Vascular Endothelial Growth Factor Promotes Bone Formation and Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4462-4477. [PMID: 38240605 DOI: 10.1021/acsami.3c16822] [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: 02/01/2024]
Abstract
Critical-size bone defects are a common and intractable clinical problem that typically requires filling in with surgical implants to facilitate bone regeneration. Considering the limitations of autologous bone and allogeneic bone in clinical applications, such as secondary damage or immunogenicity, injectable microhydrogels with osteogenic and angiogenic effects have received considerable attention. Herein, polydopamine (PDA)-functionalized strontium alginate/nanohydroxyapatite (Sr-Alg/nHA) composite microhydrogels loaded with vascular endothelial growth factor (VEGF) were prepared using microfluidic technology. This composite microhydrogel released strontium ions stably for at least 42 days to promote bone formation. The PDA coating can release VEGF in a controlled manner, effectively promote angiogenesis around bone defects, and provide nutritional support for new bone formation. In in vitro experiments, the composite microhydrogels had good biocompatibility. The PDA coating greatly improves cell adhesion on the composite microhydrogel and provides good controlled release of VEGF. Therefore, this composite microhydrogel effectively promotes osteogenic differentiation and vascularization. In in vivo experiments, composite microhydrogels were injected into critical-size bone defects in the skull of rats, and they were shown by microcomputed tomography and tissue sections to be effective in promoting bone regeneration. These findings demonstrated that this novel microhydrogel effectively promotes bone formation and angiogenesis at the site of bone defects.
Collapse
Affiliation(s)
- Shi Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, P. R. China
| | - Dawei Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P. R. China
| | - Weimin Bao
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei 230032, P. R. China
| | - Ruyuan Ding
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, P. R. China
| | - Zhenguo Shen
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei 230032, P. R. China
| | - Wenkai Huang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei 230032, P. R. China
| | - Yifan Lu
- Applied Oral Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P. R. China
| | - Panpan Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, P. R. China
| | - Yiwei Sun
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, P. R. China
| | - Hemu Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, P. R. China
| | - Cailiang Shen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, P. R. China
| | - Yuanyin Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, P. R. China
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
37
|
Ye L, Liu X, Li K, Li X, Zhu J, Yang S, Xu L, Yang M, Yan Y, Yan J. A bioinspired synthetic fused protein adhesive from barnacle cement and spider dragline for potential biomedical materials. Int J Biol Macromol 2023; 253:127125. [PMID: 37776922 DOI: 10.1016/j.ijbiomac.2023.127125] [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: 07/26/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Biomaterials with excellent biocompatibility, mechanical performance, and self-recovery properties are urgently needed for tissue regeneration. Inspired by barnacle cement and spider silk, we genetically designed and overexpressed a fused protein (cp19k-MaSp1) composed of Megabalanus rosa (cp19k) and Nephila clavata dragline silk protein (MaSp1) in Pichia pastoris. The recombinant cp19k-MaSp1 exhibited enhanced adhesion capability beyond those of the individual proteins in both aqueous and non-aqueous conditions. cp19k-MaSp1 protein fiber scaffolds prepared through electrospinning have adequate hydrophilicity compared to cp19k and MaSp1 protein fiber scaffolds, and offer improved overall porosity compared to MaSp1 protein fiber scaffolds. The cp19k-MaSp1 protein fiber scaffolds showed excellent proteolytically stable properties because of only 9.6 % depletion after incubation in a biodegradation solution for 56 d. The cp19k-MaSp1 protein fiber scaffolds present remarkably high extreme tensile strength (112.7 ± 11.6 MPa) and superior ductility (438.4 ± 43.9 %) compared with cp19k (34.4 ± 8.1 MPa, 115.4 ± 32.7 %) and MaSp1 protein fiber scaffolds (65.8 ± 9.3 MPa, 409.6 ± 23.1 %), also 68.4 % of tensile strength was recovered by incubation in K+ buffer after multiple stretches, which create a favorable cell adhesion, growth, and proliferation environment for human umbilical vein endothelial cells (HUVECs). The improved biocompatibility, extensive adhesion, mechanical strength, and self-recovery properties make the bioinspired synthetic cp19k-MaSp1 a potential candidate for biomedical tissue reconstruction.
Collapse
Affiliation(s)
- Luona Ye
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Liu
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Kai Li
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Li
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Jiarui Zhu
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Shu Yang
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Min Yang
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China
| | - Yunjun Yan
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China.
| | - Jinyong Yan
- Key Lab of Molecular Biophysics of Ministry of Education, College of Life Science and Technology Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
38
|
Han X, Saiding Q, Cai X, Xiao Y, Wang P, Cai Z, Gong X, Gong W, Zhang X, Cui W. Intelligent Vascularized 3D/4D/5D/6D-Printed Tissue Scaffolds. NANO-MICRO LETTERS 2023; 15:239. [PMID: 37907770 PMCID: PMC10618155 DOI: 10.1007/s40820-023-01187-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023]
Abstract
Blood vessels are essential for nutrient and oxygen delivery and waste removal. Scaffold-repairing materials with functional vascular networks are widely used in bone tissue engineering. Additive manufacturing is a manufacturing technology that creates three-dimensional solids by stacking substances layer by layer, mainly including but not limited to 3D printing, but also 4D printing, 5D printing and 6D printing. It can be effectively combined with vascularization to meet the needs of vascularized tissue scaffolds by precisely tuning the mechanical structure and biological properties of smart vascular scaffolds. Herein, the development of neovascularization to vascularization to bone tissue engineering is systematically discussed in terms of the importance of vascularization to the tissue. Additionally, the research progress and future prospects of vascularized 3D printed scaffold materials are highlighted and presented in four categories: functional vascularized 3D printed scaffolds, cell-based vascularized 3D printed scaffolds, vascularized 3D printed scaffolds loaded with specific carriers and bionic vascularized 3D printed scaffolds. Finally, a brief review of vascularized additive manufacturing-tissue scaffolds in related tissues such as the vascular tissue engineering, cardiovascular system, skeletal muscle, soft tissue and a discussion of the challenges and development efforts leading to significant advances in intelligent vascularized tissue regeneration is presented.
Collapse
Affiliation(s)
- Xiaoyu Han
- 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, People's Republic of China
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Qimanguli Saiding
- 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, People's Republic of China
| | - Xiaolu Cai
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, People's Republic of China
| | - Yi Xiao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Peng Wang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China
| | - Zhengwei Cai
- 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, People's Republic of China
| | - Xuan Gong
- University of Texas Southwestern Medical Center, Dallas, TX, 75390-9096, USA
| | - Weiming Gong
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, People's Republic of China.
| | - Xingcai Zhang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| | - 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, People's Republic of China.
| |
Collapse
|
39
|
Lin H, Zhang L, Zhang Q, Wang Q, Wang X, Yan G. Mechanism and application of 3D-printed degradable bioceramic scaffolds for bone repair. Biomater Sci 2023; 11:7034-7050. [PMID: 37782081 DOI: 10.1039/d3bm01214j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Bioceramics have attracted considerable attention in the field of bone repair because of their excellent osteogenic properties, degradability, and biocompatibility. To resolve issues regarding limited formability, recent studies have introduced 3D printing technology for the fabrication of bioceramic bone repair scaffolds. Nevertheless, the mechanisms by which bioceramics promote bone repair and clinical applications of 3D-printed bioceramic scaffolds remain elusive. This review provides an account of the fabrication methods of 3D-printed degradable bioceramic scaffolds. In addition, the types and characteristics of degradable bioceramics used in clinical and preclinical applications are summarized. We have also highlighted the osteogenic molecular mechanisms in biomaterials with the aim of providing a basis and support for future research on the clinical applications of degradable bioceramic scaffolds. Finally, new developments and potential applications of 3D-printed degradable bioceramic scaffolds are discussed with reference to experimental and theoretical studies.
Collapse
Affiliation(s)
- Hui Lin
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Liyun Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiyue Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, China
| | - Xue Wang
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| | - Guangqi Yan
- School and Hospital of Stomatology, China Medical University, Shenyang, China.
| |
Collapse
|
40
|
Zhang Z, Zhang X, Zheng Z, Xin J, Han S, Qi J, Zhang T, Wang Y, Zhang S. Latest advances: Improving the anti-inflammatory and immunomodulatory properties of PEEK materials. Mater Today Bio 2023; 22:100748. [PMID: 37600350 PMCID: PMC10432209 DOI: 10.1016/j.mtbio.2023.100748] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
Excellent biocompatibility, mechanical properties, chemical stability, and elastic modulus close to bone tissue make polyetheretherketone (PEEK) a promising orthopedic implant material. However, biological inertness has hindered the clinical applications of PEEK. The immune responses and inflammatory reactions after implantation would interfere with the osteogenic process. Eventually, the proliferation of fibrous tissue and the formation of fibrous capsules would result in a loose connection between PEEK and bone, leading to implantation failure. Previous studies focused on improving the osteogenic properties and antibacterial ability of PEEK with various modification techniques. However, few studies have been conducted on the immunomodulatory capacity of PEEK. New clinical applications and advances in processing technology, research, and reports on the immunomodulatory capacity of PEEK have received increasing attention in recent years. Researchers have designed numerous modification techniques, including drug delivery systems, surface chemical modifications, and surface porous treatments, to modulate the post-implantation immune response to address the regulatory factors of the mechanism. These studies provide essential ideas and technical preconditions for the development and research of the next generation of PEEK biological implant materials. This paper summarizes the mechanism by which the immune response after PEEK implantation leads to fibrous capsule formation; it also focuses on modification techniques to improve the anti-inflammatory and immunomodulatory abilities of PEEK. We also discuss the limitations of the existing modification techniques and present the corresponding future perspectives.
Collapse
Affiliation(s)
- Zilin Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Xingmin Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Zhi Zheng
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Jingguo Xin
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Song Han
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Jinwei Qi
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Tianhui Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Yongjie Wang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Shaokun Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| |
Collapse
|
41
|
Dehghanpour P, Emadi R, Salimijazi H. Influence of mechanochemically fabricated nano-hardystonite reinforcement in polycaprolactone scaffold for potential use in bone tissue engineering: Synthesis and characterization. J Mech Behav Biomed Mater 2023; 146:106100. [PMID: 37660447 DOI: 10.1016/j.jmbbm.2023.106100] [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: 07/06/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Bone tissue engineering (BTE) has gained significant attention for the regeneration of bone tissue, particularly for critical-size bone defects. The aim of this research was first to synthesize nanopowders of hardystonite (HT) through ball milling and then to manufacture composite scaffolds for BTE use out of polycaprolactone (PCL) containing 0, 3, 5, and 10 wt% HT by electrospinning method. The crystallite size of the synthesized HT nanopowders was 42.8 nm. including up to 5 wt% HT into PCL scaffolds resulted in significant improvements, such as a reduction in the fiber diameter from 186.457±15.74 to 150.021±21.99 nm, a decrease in porosity volume from 85.2±2.5 to 80.3±3.3 %, an improvement in the mechanical properties (ultimate tensile strength: 5.7±0.2 MPa, elongation: 47.5±3.5 %, tensile modulus: 32.7±0.9 MPa), an improvement in the hydrophilicity, and biodegradability. Notably, PCL/5%HT exhibited the maximum cell viability (194±14 %). Additionally, following a 4-week of submersion in simulated body fluid (SBF), the constructed PCL/HT composite scaffolds showed a remarkable capacity to stimulate the development of hydroxyapatite (HA), which increased significantly for the 5 wt% HT scaffolds. However, at 10 wt% HT, nanopowder agglomeration led to an increase in the fiber diameter and a decrease in the mechanical characteristics. Collectively, the PCL/5%HT composite scaffolds can therefore help with the regeneration of the critical-size bone defects and offer tremendous potential for BTE applications.
Collapse
Affiliation(s)
- Pegah Dehghanpour
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 8415683111, Iran.
| | - Rahmatollah Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 8415683111, Iran.
| | - Hamidreza Salimijazi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 8415683111, Iran
| |
Collapse
|
42
|
Chen J, Zhou H, Fan Y, Gao G, Ying Y, Li J. 3D printing for bone repair: Coupling infection therapy and defect regeneration. CHEMICAL ENGINEERING JOURNAL 2023; 471:144537. [DOI: 10.1016/j.cej.2023.144537] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
|
43
|
Jin P, Xia M, Hasany M, Feng P, Bai J, Gao J, Zhang W, Mehrali M, Wang R. A tough injectable self‐setting cement‐based hydrogel for noninvasive bone augmentation. INTERDISCIPLINARY MATERIALS 2023; 2:771-788. [DOI: 10.1002/idm2.12119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/05/2023] [Indexed: 03/06/2025]
Abstract
AbstractComposite hydrogels with excellent properties can open new opportunities to terminate the need for auto/allografts in bone augmentations. However, their clinical application has been limited by their insufficient mechanical strength and lack of osteoinductivity. Here we report a new strategy to design an injectable bioactive double network hydrogel reinforced by inorganic calcium/magnesium phosphate cement (CMPC) hydrates to meet the mechanical performance requirements for bone regeneration. The engineered CMPC hydration endows the composite hydrogel with an appropriate gelation time and temperature for injection, which shows no harm in the defect site. CMPC hydrates could also provide a lower swelling ratio and higher biodegradation rate fitting the in vivo bone regeneration needs. In vitro and in vivo experiments prove that the ions released from inorganic particles endow biocompatibility, cell migration, adhesion, differentiation, and significantly higher bone regeneration capacity. Taken together, the simple addition of CMPC particles imparts in‐demand features that bring us closer to the clinical utilization of hydrogel‐based materials for bone regeneration.
Collapse
Affiliation(s)
- Peng Jin
- School of Materials Science and Engineering Nanjing China
- Jiangsu Key Laboratory of Construction Materials Southeast University Nanjing China
- Department of Civil and Mechanical Engineering Technical University of Denmark Kgs Lyngby Denmark
| | - Mingjie Xia
- Department of Orthopedics, Nanjing First Hospital Nanjing Medical University Nanjing China
| | - Masoud Hasany
- Department of Civil and Mechanical Engineering Technical University of Denmark Kgs Lyngby Denmark
| | - Pan Feng
- School of Materials Science and Engineering Nanjing China
- Jiangsu Key Laboratory of Construction Materials Southeast University Nanjing China
| | - Jing Bai
- School of Materials Science and Engineering Nanjing China
- Institution of Medical Devices (Suzhou) Southeast University Nanjing China
| | - Jian Gao
- School of Materials Science and Engineering Nanjing China
- Jiangsu Key Laboratory of Construction Materials Southeast University Nanjing China
| | - Wei Zhang
- School of Materials Science and Engineering Nanjing China
- Jiangsu Key Laboratory of Advanced Metallic Materials Southeast University Nanjing China
| | - Mehdi Mehrali
- Department of Civil and Mechanical Engineering Technical University of Denmark Kgs Lyngby Denmark
| | - Ruixing Wang
- School of Materials Science and Engineering Nanjing China
- Jiangsu Key Laboratory of Construction Materials Southeast University Nanjing China
| |
Collapse
|
44
|
Su Q, Qiao Y, Xiao Y, Yang S, Wu H, Li J, He X, Hu X, Yang H, Yong X. Research progress of 3D printed poly (ether ether ketone) in the reconstruction of craniomaxillofacial bone defects. Front Bioeng Biotechnol 2023; 11:1259696. [PMID: 37662437 PMCID: PMC10469012 DOI: 10.3389/fbioe.2023.1259696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
The clinical challenge of bone defects in the craniomaxillofacial region, which can lead to significant physiological dysfunction and psychological distress, persists due to the complex and unique anatomy of craniomaxillofacial bones. These critical-sized defects require the use of bone grafts or substitutes for effective reconstruction. However, current biomaterials and methods have specific limitations in meeting the clinical demands for structural reinforcement, mechanical support, exceptional biological performance, and aesthetically pleasing reconstruction of the facial structure. These drawbacks have led to a growing need for novel materials and technologies. The growing development of 3D printing can offer significant advantages to address these issues, as demonstrated by the fabrication of patient-specific bioactive constructs with controlled structural design for complex bone defects in medical applications using this technology. Poly (ether ether ketone) (PEEK), among a number of materials used, is gaining recognition as a feasible substitute for a customized structure that closely resembles natural bone. It has proven to be an excellent, conformable, and 3D-printable material with the potential to replace traditional autografts and titanium implants. However, its biological inertness poses certain limitations. Therefore, this review summarizes the distinctive features of craniomaxillofacial bones and current methods for bone reconstruction, and then focuses on the increasingly applied 3D printed PEEK constructs in this field and an update on the advanced modifications for improved mechanical properties, biological performance, and antibacterial capacity. Exploring the potential of 3D printed PEEK is expected to lead to more cost-effective, biocompatible, and personalized treatment of craniomaxillofacial bone defects in clinical applications.
Collapse
Affiliation(s)
- Qiao Su
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yixin Qiao
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yile Xiao
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shuhao Yang
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Haoming Wu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Jianan Li
- State Key Laboratory of Biotherapy, State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xinlong He
- State Key Laboratory of Biotherapy, State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, State Key Laboratory and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Gu L, Huang R, Ni N, Gu P, Fan X. Advances and Prospects in Materials for Craniofacial Bone Reconstruction. ACS Biomater Sci Eng 2023; 9:4462-4496. [PMID: 37470754 DOI: 10.1021/acsbiomaterials.3c00399] [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: 07/21/2023]
Abstract
The craniofacial region is composed of 23 bones, which provide crucial function in keeping the normal position of brain and eyeballs, aesthetics of the craniofacial complex, facial movements, and visual function. Given the complex geometry and architecture, craniofacial bone defects not only affect the normal craniofacial structure but also may result in severe craniofacial dysfunction. Therefore, the exploration of rapid, precise, and effective reconstruction of craniofacial bone defects is urgent. Recently, developments in advanced bone tissue engineering bring new hope for the ideal reconstruction of the craniofacial bone defects. This report, presenting a first-time comprehensive review of recent advances of biomaterials in craniofacial bone tissue engineering, overviews the modification of traditional biomaterials and development of advanced biomaterials applying to craniofacial reconstruction. Challenges and perspectives of biomaterial development in craniofacial fields are discussed in the end.
Collapse
Affiliation(s)
- Li Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Rui Huang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ni Ni
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
46
|
Peng S, He T, Liu Y, Zheng L, Zhong Y, Niu Z, Zhang M, Yang S. Lnc-PPP2R1B Mediates the Alternative Splicing of PPP2R1B by Interacting and Stabilizing HNRNPLL and Promotes Osteogenesis of MSCs. Stem Cell Rev Rep 2023; 19:1981-1993. [PMID: 37243830 DOI: 10.1007/s12015-023-10559-5] [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] [Accepted: 05/08/2023] [Indexed: 05/29/2023]
Abstract
Osteogeinc differentiation from mesenchymal stem cells (MSCs) into osteoblasts is a key step for bone tissue engineering in regenerative medicine. The insight into regulatory mechanism of osteogenesis of MSCs facilitates achieving better recovery effect. Long non-coding RNAs are regarded as a family of important moderators in osteogenesis. In this study, we found a novel lncRNA, lnc-PPP2R1B was up-regulated during osteogenesis of MSCs by Illumina HiSeq transcritome sequencing. We demonstrated lnc-PPP2R1B overexpression promoted osteogenesis and knockdown of lnc-PPP2R1B inhibited osteogenesis of MSCs. Mechanically, it physically interacted with and up-regulated heterogeneous nuclear ribonucleoprotein L Like (HNRNPLL), which is a master regulator of activation-induced alternative splicing in T cells. We found lnc-PPP2R1B knockdown or HNRNPLL knockdown decreased transcript-201 of Protein Phosphatase 2A, Regulatory Subunit A, Beta Isoform (PPP2R1B) while increased transcript-203 of PPP2R1B, and did not affect transcript-202/204/206. PPP2R1B is a constant regulatory subunit of protein phosphatase 2 (PP2A), which activates Wnt/β-catenin pathway by removing phosphorylation and stabilization of β-catenin and translocation into nucleus. The transcript-201 retained exon 2 and 3, compared to transcript-203. And it was reported the exon 2 and 3 of PPP2R1B were one part of B subunit binding domain on A subunit in PP2A trimer, and therefore retaining exon 2 and 3 promised formation and enzyme function of PP2A. Finally, lnc-PPP2R1B promoted ectopic osteogenesis in vivo. Conclusively, lnc-PPP2R1B mediated alternative splicing of PPP2R1B through retaining exon 2 and 3 by interacting with HNRNPLL and then promoted osteogenesis, which may facilitate an in-depth understanding of function and mechanism of lncRNAs in osteogenesis. Lnc-PPP2R1B interacted with HNRNPLL, and regulated alternative splicing of PPP2R1B through retaining exon 2 and 3, which preserved enzyme function of PP2A and enhanced dephosphorylation and nuclear translocation of β-catenin, thereby promoting Runx2 and OSX expression and then osteogenesis. And it provided experimental data and potential target for promoting bone formation and bone regeneration.
Collapse
Affiliation(s)
- Shuping Peng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Tiantian He
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ying Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Leliang Zheng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yancheng Zhong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhiyuan Niu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mojian Zhang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Sheng Yang
- The Reproduction Medical Center, the Third Affiliated Hospital of Shenzhen University, Shenzhen, China.
| |
Collapse
|
47
|
Smith JA, Petersmann S, Arbeiter F, Schäfer U. Optimization and manufacture of polyetheretherketone patient specific cranial implants by material extrusion - A clinical perspective. J Mech Behav Biomed Mater 2023; 144:105965. [PMID: 37343357 DOI: 10.1016/j.jmbbm.2023.105965] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023]
Abstract
Polyetheretherketone (PEEK) is a high performing thermoplastic that has established itself as a 'gold-standard' material for cranial reconstruction. Traditionally, milled PEEK patient specific cranial implants (PSCIs) exhibit uniform levels of smoothness (excusing suture/drainage holes) to the touch (<1 μm) and homogenous coloration throughout. They also demonstrate predictable and repeatable levels of mechanical performance, as they are machined from isotropic material blocks. The combination of such factors inspires confidence from the surgeon and in turn, approval for implantation. However, manufacturing lead-times and affiliated costs to fabricate a PSCI are high. To simplify their production and reduce expenditure, hospitals are exploring the production of in-house PEEK PSCIs by material extrusion-based additive manufacturing. From a geometrical and morphological perspective, such implants have been produced with good-to-satisfactory clinical results. However, lack of clinical adoption persists. To determine the reasoning behind this, it was necessary to assess the benefits and limitations of current printed PEEK PSCIs in order to establish the status quo. Afterwards, a review on individual PEEK printing variables was performed in order to identify a combination of parameters that could enhance the aesthetics and performance of the PSCIs to that of milled implants/cranial bone. The findings from this review could be used as a baseline to help standardize the production of PEEK PSCIs by material extrusion in the hospital.
Collapse
Affiliation(s)
- James A Smith
- Research Unit Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 2(9), 8036, Graz, Austria.
| | - Sandra Petersmann
- Materials Science and Testing of Polymers, Montanuniversitaet Leoben, Otto Gloeckel-Straße 2, 8700, Leoben, Austria
| | - Florian Arbeiter
- Materials Science and Testing of Polymers, Montanuniversitaet Leoben, Otto Gloeckel-Straße 2, 8700, Leoben, Austria
| | - Ute Schäfer
- Research Unit Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 2(9), 8036, Graz, Austria; BioTechMed-Graz, Graz, Austria
| |
Collapse
|
48
|
Bi X, Li M, Zhang Y, Yin M, Che W, Bi Z, Yang Y, Ouyang J. Polyetheretherketone (PEEK) as a Potential Material for the Repair of Maxillofacial Defect Compared with E-poly(tetrafluoroethylene) (e-PTFE) and Silicone. ACS Biomater Sci Eng 2023; 9:4328-4340. [PMID: 37276458 DOI: 10.1021/acsbiomaterials.2c00744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Silicone and e-poly(tetrafluoroethylene) (e-PTFE) are the most commonly used artificial materials for repairing maxillofacial bone defects caused by facial trauma and tumors. However, their use is limited by poor histocompatibility, unsatisfactory support, and high infection rates. Polyetheretherketone (PEEK) has excellent mechanical strength and biocompatibility, but its application to the repair of maxillofacial bone defects lacks a theoretical basis. The microstructure and mechanical properties of e-PTFE, silicone, and PEEK were evaluated by electron microscopy, BOSE machine, and Fourier transformed infrared spectroscopy. Mouse fibroblast L929 cells were incubated on the surface of the three materials to assess cytotoxicity and adhesion. The three materials were implanted onto the left femoral surface of 90 male mice, and samples of the implants and surrounding soft tissues were evaluated histologically at 1, 2, 4, 8, and 12 weeks post-surgery. PEEK had a much higher Young's modulus than either e-PTFE or silicone (p < 0.05 each), and maintained high stiffness without degradation long after implantation. Both PEEK and e-PTFE facilitated L929 cell adhesion, with PEEK having lower cytotoxicity than e-PTFE and silicone (p < 0.05 each). All three materials similarly hindered the motor function of mice 12 weeks after implantation (p > 0.05 each). Connective tissue ingrowth was observed in PEEK and e-PTFE, whereas a fibrotic peri-prosthetic capsule was observed on the surface of silicone. The postoperative infection rate was significantly lower for both PEEK and silicone than for e-PTFE (p < 0.05 each). PEEK shows excellent biocompatibility and mechanical stability, suggesting that it can be effective as a novel implant to repair maxillofacial bone defects.
Collapse
Affiliation(s)
- Xin Bi
- Guangdong Provincial Key Laboratory of Medical Biomechanics & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, No. 1038, Guangzhou Road North, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Mingdong Li
- Department of Orthopedics and Traumatology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Xiuying District, Haikou, Hainan 570300, China
| | - Yuchen Zhang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, No. 1038, Guangzhou Road North, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Ming Yin
- Department of Imaging, Nanfang Hospital, Southern Medical University, No. 1038, Guangzhou Road North, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Wuqiang Che
- Guangdong Provincial Key Laboratory of Medical Biomechanics & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Zhenyu Bi
- Guangdong Provincial Key Laboratory of Medical Biomechanics & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, Guangdong 510515, China
| |
Collapse
|
49
|
Shu R, Sun J, Li B, Gao X, He M, Chan YK, Shi J, Bai D, Yang W, Deng Y. Self‐Tandem Bio‐Heterojunctions Empower Orthopedic Implants with Amplified Chemo‐Photodynamic Anti‐Pathogenic Therapy and Boosted Diabetic Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202214873] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Indexed: 01/03/2025]
Abstract
AbstractHyperglycemic microenvironment in diabetes mellitus inevitably stalls the normal orchestrated course of bone regeneration and encourages pathogenic multiplication. Photodynamic therapy (PDT) and chemo‐dynamic therapy (CDT) are extensively harnessed to combat pathogens, yet deep‐seated diabetic bone defect has difficulty in supplying sufficient oxygen (O2) and hydrogen peroxide (H2O2) stocks, resulting in inferior therapeutic efficiency. To address the tough plaguing, the self‐tandem bio‐heterojunctions (bio‐HJs) consisting of molybdenum disulfide (MoS2), graphene oxide (GO), and glucose oxidase (GOx) are constructed on orthopedic polyetheretherketone (PEEK) implants (SP‐Mo/G@GOx) for amplified chemo‐photodynamic anti‐pathogenic therapy and boosted osseointegration in the deep‐seated diabetic micromilieu. In this system, GOx exhausts glucose to generate H2O2, which provides an abundant stock for CDT. Besides, the bio‐HJs produce hyperthermia upon near‐infrared light (NIR) to accelerate the dynamic process, which amplifies the antibacterial potency of PDT by promoting the vast yield of singlet oxygen (1O2) in a self‐tandem manner. More importantly, in vivo and in vitro assays demonstrate that the engineered implants exert a captivated bactericidal ability and significantly boost osseointegration in an infectious diabetic bone defect model. As envisaged, this study furnishes a novel tactic to arm orthopedic implants with self‐tandem capability for the remedy of infectious diabetic bone defects.
Collapse
Affiliation(s)
- Rui Shu
- College of Biomedical Engineering State Key Laboratory of Oral Diseases West China Hospital of Stomatology School of Chemical Engineering Sichuan University Chengdu 610065 P. R. China
- National Clinical Research Center for Oral Diseases Department of Pediatric Dentistry West China Hospital of Stomatology Sichuan University Chengdu 610065 P. R. China
| | - Jiamin Sun
- College of Biomedical Engineering State Key Laboratory of Oral Diseases West China Hospital of Stomatology School of Chemical Engineering Sichuan University Chengdu 610065 P. R. China
| | - Bin Li
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Department of Orthodontics West China Hospital of Stomatology Sichuan University Chengdu 610065 P. R. China
| | - Xiangyu Gao
- College of Biomedical Engineering State Key Laboratory of Oral Diseases West China Hospital of Stomatology School of Chemical Engineering Sichuan University Chengdu 610065 P. R. China
| | - Miaomiao He
- College of Biomedical Engineering State Key Laboratory of Oral Diseases West China Hospital of Stomatology School of Chemical Engineering Sichuan University Chengdu 610065 P. R. China
| | - Yau Kei Chan
- Department of Ophthalmology The University of Hong Kong Hong Kong SAR 999077 P. R. China
| | - Jiacheng Shi
- College of Biomedical Engineering State Key Laboratory of Oral Diseases West China Hospital of Stomatology School of Chemical Engineering Sichuan University Chengdu 610065 P. R. China
| | - Ding Bai
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Department of Orthodontics West China Hospital of Stomatology Sichuan University Chengdu 610065 P. R. China
| | - Weizhong Yang
- College of Biomedical Engineering State Key Laboratory of Oral Diseases West China Hospital of Stomatology School of Chemical Engineering Sichuan University Chengdu 610065 P. R. China
| | - Yi Deng
- College of Biomedical Engineering State Key Laboratory of Oral Diseases West China Hospital of Stomatology School of Chemical Engineering Sichuan University Chengdu 610065 P. R. China
- Department of Mechanical Engineering The University of Hong Kong Hong Kong SAR 999077 P. R. China
- State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
| |
Collapse
|
50
|
Sharma S, Mudgal D, Gupta V. Advancement in biological and mechanical behavior of 3D printed poly lactic acid bone plates using polydopamine coating: Innovation for healthcare. J Mech Behav Biomed Mater 2023; 143:105929. [PMID: 37263171 DOI: 10.1016/j.jmbbm.2023.105929] [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: 04/05/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 06/03/2023]
Abstract
The metallic biomaterials have been proclaimed to exhibit stress shielding with discharge of toxic ions, leading to polymeric implants attracting interest in 3D Printing domain. In this study, Poly Lactic Acid based 336 bone plates are fabricated using Fused Filament Fabrication with printing parameters being varied. Polydopamine, being biocompatible, is deposited on fabricated bone plates at varying submersion time, shaker speed and coating solutions concentration. The study involves witnessing the effect of printing and coating parameters on biological behavior of bone plates upon preservation in Simulated Body Fluid and Hank's Balanced Salt Solution. The findings propose the close relation of degradation with apatite growth. The highest degradation rate with significant reduction in mechanical characteristics are shown by uncoated bone plates. These bone plates have porous structure at 20% infill density, 0.5 mm layer height, 0.4 mm wall thickness and 100 mm/s print speed which could result in complete degradation with partial healing of bone fracture. The study suggests the preservation of bone plates coated at 120 h' submersion time and 120 RPM shaker speed in 3 mg/ml concentrated solution which showed lower apatite formation. Thus, the coating would slow down degradation of PLA bone plates, resulting in complete healing of bone fracture.
Collapse
Affiliation(s)
- Shrutika Sharma
- Mechanical Engineering Department, Thapar Institute of Engineering and Technology, Patiala, 147004, Punjab, India
| | - Deepa Mudgal
- Mechanical Engineering Department, Thapar Institute of Engineering and Technology, Patiala, 147004, Punjab, India
| | - Vishal Gupta
- Mechanical Engineering Department, Thapar Institute of Engineering and Technology, Patiala, 147004, Punjab, India.
| |
Collapse
|