1
|
Zaszczyńska A, Zabielski K, Gradys A, Kowalczyk T, Sajkiewicz P. Piezoelectric Scaffolds as Smart Materials for Bone Tissue Engineering. Polymers (Basel) 2024; 16:2797. [PMID: 39408507 PMCID: PMC11479154 DOI: 10.3390/polym16192797] [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: 08/26/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Bone repair and regeneration require physiological cues, including mechanical, electrical, and biochemical activity. Many biomaterials have been investigated as bioactive scaffolds with excellent electrical properties. Amongst biomaterials, piezoelectric materials (PMs) are gaining attention in biomedicine, power harvesting, biomedical devices, and structural health monitoring. PMs have unique properties, such as the ability to affect physiological movements and deliver electrical stimuli to damaged bone or cells without an external power source. The crucial bone property is its piezoelectricity. Bones can generate electrical charges and potential in response to mechanical stimuli, as they influence bone growth and regeneration. Piezoelectric materials respond to human microenvironment stimuli and are an important factor in bone regeneration and repair. This manuscript is an overview of the fundamentals of the materials generating the piezoelectric effect and their influence on bone repair and regeneration. This paper focuses on the state of the art of piezoelectric materials, such as polymers, ceramics, and composites, and their application in bone tissue engineering. We present important information from the point of view of bone tissue engineering. We highlight promising upcoming approaches and new generations of piezoelectric materials.
Collapse
Affiliation(s)
| | | | | | - Tomasz Kowalczyk
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland; (A.Z.); (K.Z.); (A.G.); (P.S.)
| | | |
Collapse
|
2
|
Han K, Mao M, Fu L, Zhang Y, Kang Y, Li D, He J. Multimaterial Printing of Serpentine Microarchitectures with Synergistic Mechanical/Piezoelectric Stimulation for Enhanced Cardiac-Specific Functional Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401561. [PMID: 38899348 DOI: 10.1002/smll.202401561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/21/2024] [Indexed: 06/21/2024]
Abstract
Recreating the natural heart's mechanical and electrical environment is crucial for engineering functional cardiac tissue and repairing infarcted myocardium in vivo. In this study, multimaterial-printed serpentine microarchitectures are presented with synergistic mechanical/piezoelectric stimulation, incorporating polycaprolactone (PCL) microfibers for mechanical support, polyvinylidene fluoride (PVDF) microfibers for piezoelectric stimulation, and magnetic PCL/Fe3O4 for controlled deformation via an external magnet. Rat cardiomyocytes in piezoelectric constructs, subjected to dynamic mechanical stimulation, exhibit advanced maturation, featuring superior sarcomeric structures, improved calcium transients, and upregulated maturation genes compared to non-piezoelectric constructs. Furthermore, these engineered piezoelectric cardiac constructs demonstrate significant structural and functional repair of infarcted myocardium, as evidenced by enhanced ejection and shortening fraction, reduced fibrosis and inflammation, and increased angiogenesis. The findings underscore the therapeutic potential of piezoelectric cardiac constructs for myocardial infarction therapy.
Collapse
Affiliation(s)
- Kang Han
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Mao Mao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Liyan Fu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, 710061, P. R. China
| | - Yabo Zhang
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yuming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi'an, 710061, P. R. China
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
3
|
Martins LA, García-Parra N, Ródenas-Rochina J, Cordón L, Sempere A, Ribeiro C, Lanceros-Méndez S, Gómez-Ribelles JL. Assemblable 3D biomimetic microenvironment for hMSC osteogenic differentiation. Biomed Mater 2024; 19:065013. [PMID: 39303743 DOI: 10.1088/1748-605x/ad7dc4] [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: 04/04/2024] [Accepted: 09/20/2024] [Indexed: 09/22/2024]
Abstract
Adequate simulation mimicking a tissue's native environment is one of the elemental premises in tissue engineering. Although various attempts have been made to induce human mesenchymal stem cells (hMSC) into an osteogenic pathway, they are still far from widespread clinical application. Most strategies focus primarily on providing a specific type of cue, inadequately replicating the complexity of the bone microenvironment. An alternative multifunctional platform for hMSC osteogenic differentiation has been produced. It is based on poly(vinylidene fluoride) (PVDF) and cobalt ferrites magnetoelectric microspheres, functionalized with collagen and gelatin, and packed in a 3D arrangement. This platform is capable of performing mechanical stimulation of piezoelectric PVDF, mimicking the bones electromechanical biophysical cues. Surface functionalization with extracellular matrix biomolecules and osteogenic medium complete this all-round approach. hMSC were cultured in osteogenic inducing conditions and tested for proliferation, surface biomarkers, and gene expression to evaluate their osteogenic commitment.
Collapse
Affiliation(s)
- Luis A Martins
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Nadia García-Parra
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Joaquín Ródenas-Rochina
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Lourdes Cordón
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe (IISLAFE), 46022 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
| | - Amparo Sempere
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe (IISLAFE), 46022 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Clarisse Ribeiro
- CF-UM-UP-Physics Centre of Minho and Porto Universities, University of Minho, 4710-057 Braga, Portugal
- LaPMET-Laboratory of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
| | - Senentxu Lanceros-Méndez
- CF-UM-UP-Physics Centre of Minho and Porto Universities, University of Minho, 4710-057 Braga, Portugal
- BCMaterials-Basque Center for Materials Applications and Nanostructures, University of the Basque Country, 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - José Luis Gómez-Ribelles
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, 46022 Valencia, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Aadinath W, K S P S T, Saravanakumar I, Muthuvijayan V. Iron oxide nanoparticle-stabilized Pickering emulsion-templated porous scaffolds loaded with polyunsaturated fatty acids (PUFAs) for bone tissue engineering. J Mater Chem B 2024; 12:9312-9324. [PMID: 39171408 DOI: 10.1039/d4tb00286e] [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: 08/23/2024]
Abstract
Dietary intake of ω-3-polyunsaturated fatty acids (PUFAs) can significantly improve the expression levels of alkaline phosphatase (ALP) and osteocalcin. However, PUFAs are hydrophobic and highly sensitive to temperature, oxygen concentration, pH, and ionic strength. Hence, it is challenging to use PUFAs as bioactive compounds for bone tissue engineering. Here, we encapsulated PUFAs in liposomes to improve their stability. The hydrodynamic size of the PUFA-loaded liposomes was found to be 121.3 ± 35 nm. GC-MS analysis showed that the encapsulation efficiency of the PUFAs was 19.9 ± 3.4%. These PUFA-loaded liposomes were loaded into porous scaffolds that were prepared by polymerizing glycidyl methacrylate and trimethylolpropane triacrylate monomers using the Pickering emulsion polymerization technique. Oleic acid-coated iron oxide nanoparticles were used as the stabilizing agent to prepare these acrylate-based scaffolds containing PUFA-loaded liposomes (P-Lipo-IO(GMA-TMPTA)). SEM micrographs confirmed the porous nature of the scaffolds and the presence of well-adhered liposomes. An in vitro cytotoxicity study conducted using MG63 cells confirmed that these scaffolds showed desirable cytocompatibility. Cell adhesion study showed a well-spread morphology, indicating firm adhesion of the cells. The alizarin red staining of P-Lipo-IO(GMA-TMPTA) scaffolds showed 3- and 2-fold higher calcium deposition compared to the control on days 7 and 14, respectively. ALP activity was also 2-fold higher than that of the control on day 14. RT-PCR analysis of cells exposed to P-Lipo-IO(GMA-TMPTA) scaffolds showed significantly higher expression of osteogenic markers compared to the control. An antibacterial study conducted on Staphylococcus aureus showed a higher percentage inhibition and reactive oxygen species generation in samples treated with P-Lipo-IO(GMA-TMPTA) scaffolds. These desirable biological properties indicate that the developed scaffolds are suitable for bone tissue engineering.
Collapse
Affiliation(s)
- W Aadinath
- Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600 036, Tamil Nadu, India.
| | - Teja K S P S
- Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600 036, Tamil Nadu, India.
| | - Iniyan Saravanakumar
- Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600 036, Tamil Nadu, India.
| | - Vignesh Muthuvijayan
- Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600 036, Tamil Nadu, India.
| |
Collapse
|
5
|
Yuan X, Zhu W, Yang Z, He N, Chen F, Han X, Zhou K. Recent Advances in 3D Printing of Smart Scaffolds for Bone Tissue Engineering and Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403641. [PMID: 38861754 DOI: 10.1002/adma.202403641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Indexed: 06/13/2024]
Abstract
The repair and functional reconstruction of bone defects resulting from severe trauma, surgical resection, degenerative disease, and congenital malformation pose significant clinical challenges. Bone tissue engineering (BTE) holds immense potential in treating these severe bone defects, without incurring prevalent complications associated with conventional autologous or allogeneic bone grafts. 3D printing technology enables control over architectural structures at multiple length scales and has been extensively employed to process biomimetic scaffolds for BTE. In contrast to inert and functional bone grafts, next-generation smart scaffolds possess a remarkable ability to mimic the dynamic nature of native extracellular matrix (ECM), thereby facilitating bone repair and regeneration. Additionally, they can generate tailored and controllable therapeutic effects, such as antibacterial or antitumor properties, in response to exogenous and/or endogenous stimuli. This review provides a comprehensive assessment of the progress of 3D-printed smart scaffolds for BTE applications. It begins with an introduction to bone physiology, followed by an overview of 3D printing technologies utilized for smart scaffolds. Notable advances in various stimuli-responsive strategies, therapeutic efficacy, and applications of 3D-printed smart scaffolds are discussed. Finally, the review highlights the existing challenges in the development and clinical implementation of smart scaffolds, as well as emerging technologies in this field.
Collapse
Affiliation(s)
- Xun Yuan
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Wei Zhu
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Zhongyuan Yang
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Ning He
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Feng Chen
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Xiaoxiao Han
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Kun Zhou
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
6
|
Chen A, Wang W, Mao Z, He Y, Chen S, Liu G, Su J, Feng P, Shi Y, Yan C, Lu J. Multimaterial 3D and 4D Bioprinting of Heterogenous Constructs for Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307686. [PMID: 37737521 DOI: 10.1002/adma.202307686] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Indexed: 09/23/2023]
Abstract
Additive manufacturing (AM), which is based on the principle of layer-by-layer shaping and stacking of discrete materials, has shown significant benefits in the fabrication of complicated implants for tissue engineering (TE). However, many native tissues exhibit anisotropic heterogenous constructs with diverse components and functions. Consequently, the replication of complicated biomimetic constructs using conventional AM processes based on a single material is challenging. Multimaterial 3D and 4D bioprinting (with time as the fourth dimension) has emerged as a promising solution for constructing multifunctional implants with heterogenous constructs that can mimic the host microenvironment better than single-material alternatives. Notably, 4D-printed multimaterial implants with biomimetic heterogenous architectures can provide a time-dependent programmable dynamic microenvironment that can promote cell activity and tissue regeneration in response to external stimuli. This paper first presents the typical design strategies of biomimetic heterogenous constructs in TE applications. Subsequently, the latest processes in the multimaterial 3D and 4D bioprinting of heterogenous tissue constructs are discussed, along with their advantages and challenges. In particular, the potential of multimaterial 4D bioprinting of smart multifunctional tissue constructs is highlighted. Furthermore, this review provides insights into how multimaterial 3D and 4D bioprinting can facilitate the realization of next-generation TE applications.
Collapse
Affiliation(s)
- Annan Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- 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
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Wanying Wang
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| | - Zhengyi Mao
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Yunhu He
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Shiting Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Guo Liu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, 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
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Pei Feng
- State Key Laboratory of High-Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, 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
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, 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
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Jian Lu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Hong Kong Branch of National Precious Metals Material Engineering Research, Center (NPMM), City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
7
|
Percival KM, Paul V, Husseini GA. Recent Advancements in Bone Tissue Engineering: Integrating Smart Scaffold Technologies and Bio-Responsive Systems for Enhanced Regeneration. Int J Mol Sci 2024; 25:6012. [PMID: 38892199 PMCID: PMC11172494 DOI: 10.3390/ijms25116012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
In exploring the challenges of bone repair and regeneration, this review evaluates the potential of bone tissue engineering (BTE) as a viable alternative to traditional methods, such as autografts and allografts. Key developments in biomaterials and scaffold fabrication techniques, such as additive manufacturing and cell and bioactive molecule-laden scaffolds, are discussed, along with the integration of bio-responsive scaffolds, which can respond to physical and chemical stimuli. These advancements collectively aim to mimic the natural microenvironment of bone, thereby enhancing osteogenesis and facilitating the formation of new tissue. Through a comprehensive combination of in vitro and in vivo studies, we scrutinize the biocompatibility, osteoinductivity, and osteoconductivity of these engineered scaffolds, as well as their interactions with critical cellular players in bone healing processes. Findings from scaffold fabrication techniques and bio-responsive scaffolds indicate that incorporating nanostructured materials and bioactive compounds is particularly effective in promoting the recruitment and differentiation of osteoprogenitor cells. The therapeutic potential of these advanced biomaterials in clinical settings is widely recognized and the paper advocates continued research into multi-responsive scaffold systems.
Collapse
Affiliation(s)
- Kelly M. Percival
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (K.M.P.); (V.P.)
| | - Vinod Paul
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (K.M.P.); (V.P.)
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (K.M.P.); (V.P.)
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
8
|
Chen Z, Li J, Wang Z, Chen Y, Jin M, Chen S, Xie J, Ge S, He H, Xu J, Wu F. Polydopamine-mediated immobilization of BMP-2 onto electrospun nanofibers enhances bone regeneration. NANOTECHNOLOGY 2024; 35:325101. [PMID: 38688249 DOI: 10.1088/1361-6528/ad4554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 04/30/2024] [Indexed: 05/02/2024]
Abstract
Dealing with bone defects is a significant challenge to global health. Electrospinning in bone tissue engineering has emerged as a solution to this problem. In this study, we designed a PVDF-b-PTFE block copolymer by incorporating TFE, which induced a phase shift in PVDF fromαtoβ, thereby enhancing the piezoelectric effect. Utilizing the electrospinning process, we not only converted the material into a film with a significant surface area and high porosity but also intensified the piezoelectric effect. Then we used polydopamine to immobilize BMP-2 onto PVDF-b-PTFE electrospun nanofibrous membranes, achieving a controlled release of BMP-2. The scaffold's characters were examined using SEM and XRD. To assess its osteogenic effectsin vitro, we monitored the proliferation of MC3T3-E1 cells on the fibers, conducted ARS staining, and measured the expression of osteogenic genes.In vivo, bone regeneration effects were analyzed through micro-CT scanning and HE staining. ELISA assays confirmed that the sustained release of BMP-2 can be maintained for at least 28 d. SEM images and CCK-8 results demonstrated enhanced cell viability and improved adhesion in the experimental group. Furthermore, the experimental group exhibited more calcium nodules and higher expression levels of osteogenic genes, including COL-I, OCN, and RUNX2. HE staining and micro-CT scans revealed enhanced bone tissue regeneration in the defective area of the PDB group. Through extensive experimentation, we evaluated the scaffold's effectiveness in augmenting osteoblast proliferation and differentiation. This study emphasized the potential of piezoelectric PVDF-b-PTFE nanofibrous membranes with controlled BMP-2 release as a promising approach for bone tissue engineering, providing a viable solution for addressing bone defects.
Collapse
Affiliation(s)
- Zhuo Chen
- Department of Orthopaedics and Rehabilitation, Affiliated Huzhou Hospital, Zhejiang University School of Medicine; Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University; Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University; Huzhou Basic and Clinical Translation of Orthopaedics Key Laboratory; Huzhou Shushan Geriatric Hospital, Huzhou, People's Republic of China
| | - Jing Li
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, Huzhou, Zhejiang 313000, People's Republic of China
| | - Zichen Wang
- Department of Orthopaedics and Rehabilitation, Affiliated Huzhou Hospital, Zhejiang University School of Medicine; Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University; Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University; Huzhou Basic and Clinical Translation of Orthopaedics Key Laboratory; Huzhou Shushan Geriatric Hospital, Huzhou, People's Republic of China
| | - Yuehui Chen
- Key Laboratory of Textile Science & Technology, College of Textile, Donghua University, Shanghai, 201620, People's Republic of China
| | - Mingchao Jin
- Department of Orthopaedics and Rehabilitation, Affiliated Huzhou Hospital, Zhejiang University School of Medicine; Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University; Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University; Huzhou Basic and Clinical Translation of Orthopaedics Key Laboratory; Huzhou Shushan Geriatric Hospital, Huzhou, People's Republic of China
| | - Shuo Chen
- Key Laboratory of Textile Science & Technology, College of Textile, Donghua University, Shanghai, 201620, People's Republic of China
| | - Jinlu Xie
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, Huzhou, Zhejiang 313000, People's Republic of China
| | - Shuhui Ge
- Key Laboratory of Textile Science & Technology, College of Textile, Donghua University, Shanghai, 201620, People's Republic of China
| | - Hongyi He
- School of Pharmacy, Hubei University of Science and Technology, Xianning, People's Republic of China
| | - Juntao Xu
- Department of Orthopaedics, Huzhou Traditional Chinese Medicine Hospital, Affiliated to Zhejiang Chinese Medical University, Huzhou, People's Republic of China
| | - Fengfeng Wu
- Department of Orthopaedics and Rehabilitation, Affiliated Huzhou Hospital, Zhejiang University School of Medicine; Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University; Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University; Huzhou Basic and Clinical Translation of Orthopaedics Key Laboratory; Huzhou Shushan Geriatric Hospital, Huzhou, People's Republic of China
| |
Collapse
|
9
|
Sun W, Gao C, Liu H, Zhang Y, Guo Z, Lu C, Qiao H, Yang Z, Jin A, Chen J, Dai Q, Liu Y. Scaffold-Based Poly(Vinylidene Fluoride) and Its Copolymers: Materials, Fabrication Methods, Applications, and Perspectives. ACS Biomater Sci Eng 2024; 10:2805-2826. [PMID: 38621173 DOI: 10.1021/acsbiomaterials.3c01989] [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: 04/17/2024]
Abstract
Tissue engineering involves implanting grafts into damaged tissue sites to guide and stimulate the formation of new tissue, which is an important strategy in the field of tissue defect treatment. Scaffolds prepared in vitro meet this requirement and are able to provide a biochemical microenvironment for cell growth, adhesion, and tissue formation. Scaffolds made of piezoelectric materials can apply electrical stimulation to the tissue without an external power source, speeding up the tissue repair process. Among piezoelectric polymers, poly(vinylidene fluoride) (PVDF) and its copolymers have the largest piezoelectric coefficients and are widely used in biomedical fields, including implanted sensors, drug delivery, and tissue repair. This paper provides a comprehensive overview of PVDF and its copolymers and fillers for manufacturing scaffolds as well as the roles in improving piezoelectric output, bioactivity, and mechanical properties. Then, common fabrication methods are outlined such as 3D printing, electrospinning, solvent casting, and phase separation. In addition, the applications and mechanisms of scaffold-based PVDF in tissue engineering are introduced, such as bone, nerve, muscle, skin, and blood vessel. Finally, challenges, perspectives, and strategies of scaffold-based PVDF and its copolymers in the future are discussed.
Collapse
Affiliation(s)
- Wenbin Sun
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Chuang Gao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Huazhen Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yi Zhang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Zilong Guo
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Chunxiang Lu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Hao Qiao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Zhiqiang Yang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Aoxiang Jin
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Jianan Chen
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Qiqi Dai
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| |
Collapse
|
10
|
Li C, Zhu A, Yang L, Wang X, Guo Z. Advances in magnetoelectric composites for promoting bone regeneration: a review. J Mater Chem B 2024; 12:4361-4374. [PMID: 38639047 DOI: 10.1039/d3tb02617e] [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: 04/20/2024]
Abstract
Repair of large bone defects is one of the clinical problems that have not yet been fully solved. The dynamic balance of bone tissue is regulated by many biological, chemical and physical environmental factors. Simulating the microenvironment of bone tissue in the physiological state through biomimetic materials is an important development direction of tissue engineering in recent years. With the deepening of research, it has been found that when bone tissue is damaged, its surrounding magnetoelectric microenvironment is subsequently destroyed, and providing a magnetoelectric microenvironment in the biomimetic state will be beneficial to promote bone repair. This review describes the piezoelectric effect of natural bone tissue with magnetoelectric stimulation for bone regeneration, provides a detailed account of the historical development of magnetoelectric composites and the current magnetoelectric composites that are most commonly utilized in the field of tissue engineering. Besides, the hypothesized mechanistic pathways through which magnetoelectric composite materials promote bone regeneration are critically examined, including the enhancement of osteogenesis, promotion of cell adhesion and angiogenesis, modulation of bone immunity, and promotion of nerve regeneration.
Collapse
Affiliation(s)
- Chengyu Li
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| | - Andi Zhu
- Department of Implantology and Prosthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China
| | - Liqing Yang
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| | - Xinyi Wang
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| | - Zehong Guo
- Department of Periodontology and Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, P. R. China.
| |
Collapse
|
11
|
Azadani RN, Karbasi S, Poursamar A. Chitosan/MWCNTs nanocomposite coating on 3D printed scaffold of poly 3-hydroxybutyrate/magnetic mesoporous bioactive glass: A new approach for bone regeneration. Int J Biol Macromol 2024; 260:129407. [PMID: 38224805 DOI: 10.1016/j.ijbiomac.2024.129407] [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: 11/08/2023] [Revised: 12/20/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Abstract
The utilization of 3D printing has become increasingly common in the construction of composite scaffolds. In this study, magnetic mesoporous bioactive glass (MMBG) was incorporated into polyhydroxybutyrate (PHB) to construct extrusion-based 3D printed scaffold. After fabrication of the PHB/MMBG composite scaffolds, they were coated with chitosan (Cs) and chitosan/multi-walled carbon nanotubes (Cs/MWCNTs) solutions utilizing deep coating method. FTIR was conducted to confirm the presence of Cs and MWCNTs on the scaffolds' surface. The findings of mechanical analysis illustrated that presence of Cs/MWCNTs on the composite scaffolds increases compressive young modulus significantly, from 16.5 to 42.2 MPa. According to hydrophilicity evaluation, not only MMBG led to decrease the contact angle of pure PHB but also scaffolds surface modification utilization of Cs and MWCNTs, the contact angle decreased significantly from 82.34° to 54.15°. Furthermore, investigation of cell viability, cell metabolism and inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) and interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β) proved that the scaffolds not only do not stimulate the immune system, but also polarize macrophage cells from M1 phase to M2 phase. The present study highlights the suitability of 3D printed scaffold PHB/MMBG with Cs/MWCNTs coating for bone tissue engineering.
Collapse
Affiliation(s)
- Reyhaneh Nasr Azadani
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Karbasi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Dental Implants Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ali Poursamar
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Wang B, Ye X, Chen G, Zhang Y, Zeng Z, Liu C, Tan Z, Jie X. Fabrication and properties of PLA/β-TCP scaffolds using liquid crystal display (LCD) photocuring 3D printing for bone tissue engineering. Front Bioeng Biotechnol 2024; 12:1273541. [PMID: 38440328 PMCID: PMC10910430 DOI: 10.3389/fbioe.2024.1273541] [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: 08/06/2023] [Accepted: 01/08/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction: Bone defects remain a thorny challenge that clinicians have to face. At present, scaffolds prepared by 3D printing are increasingly used in the field of bone tissue repair. Polylactic acid (PLA) has good thermoplasticity, processability, biocompatibility, and biodegradability, but the PLA is brittle and has poor osteogenic performance. Beta-tricalcium phosphate (β-TCP) has good mechanical properties and osteogenic induction properties, which can make up for the drawbacks of PLA. Methods: In this study, photocurable biodegradable polylactic acid (bio-PLA) was utilized as the raw material to prepare PLA/β-TCP slurries with varying β-TCP contents (β-TCP dosage at 0%, 10%, 20%, 30%, 35% of the PLA dosage, respectively). The PLA/β-TCP scaffolds were fabricated using liquid crystal display (LCD) light-curing 3D printing technology. The characterization of the scaffolds was assessed, and the biological activity of the scaffold with the optimal compressive strength was evaluated. The biocompatibility of the scaffold was assessed through CCK-8 assays, hemocompatibility assay and live-dead staining experiments. The osteogenic differentiation capacity of the scaffold on MC3T3-E1 cells was evaluated through alizarin red staining, alkaline phosphatase (ALP) detection, immunofluorescence experiments, and RT-qPCR assays. Results: The prepared scaffold possesses a three-dimensional network structure, and with an increase in the quantity of β-TCP, more β-TCP particles adhere to the scaffold surface. The compressive strength of PLA/β-TCP scaffolds exhibits a trend of initial increase followed by decrease with an increasing amount of β-TCP, reaching a maximum value of 52.1 MPa at a 10% β-TCP content. Degradation rate curve results indicate that with the passage of time, the degradation rate of the scaffold gradually increases, and the pH of the scaffold during degradation shows an alkaline tendency. Additionally, Live/dead staining and blood compatibility experiments suggest that the prepared PLA/β-TCP scaffold demonstrates excellent biocompatibility. CCK-8 experiments indicate that the PLA/β-TCP group promotes cell proliferation, and the prepared PLA/β-TCP scaffold exhibits a significant ability to enhance the osteogenic differentiation of MC3T3-E1 cells in vitro. Discussion: 3D printed LCD photocuring PLA/β-TCP scaffolds could improve surface bioactivity and lead to better osteogenesis, which may provide a unique strategy for developing bioactive implants in orthopedic applications.
Collapse
Affiliation(s)
- Boqun Wang
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, Guangdong, China
- School of Intelligent Manufacturing, Dongguan Polytechnic, Dongguan, Guangdong, China
| | - Xiangling Ye
- Dongguan Hospital, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guocai Chen
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yongqiang Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhikui Zeng
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Cansen Liu
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Zhichao Tan
- Dongguan Hospital, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
| | - Xiaohua Jie
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Careta O, Nicolenco A, Perdikos F, Blanquer A, Ibañez E, Pellicer E, Stefani C, Sepúlveda B, Nogués J, Sort J, Nogués C. Enhanced Proliferation and Differentiation of Human Osteoblasts by Remotely Controlled Magnetic-Field-Induced Electric Stimulation Using Flexible Substrates. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58054-58066. [PMID: 38051712 PMCID: PMC10739596 DOI: 10.1021/acsami.3c09428] [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: 06/30/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023]
Abstract
With the progressive aging of the population, bone fractures are an increasing major health concern. Diverse strategies are being studied to reduce the recovery times using nonaggressive treatments. Electrical stimulation (either endogenous or externally applied electric pulses) has been found to be effective in accelerating bone cell proliferation and differentiation. However, the direct insertion of electrodes into tissues can cause undesirable inflammation or infection reactions. As an alternative, magnetoelectric heterostructures (wherein magnetic fields are applied to induce electric polarization) could be used to achieve electric stimulation without the need for implanted electrodes. Here, we develop a magnetoelectric platform based on flexible kapton/FeGa/P(VDF-TrFE) (flexible substrate/magnetostrictive layer/ferroelectric layer) heterostructures for remote magnetic-field-induced electric field stimulation of human osteoblast cells. We show that the use of flexible supports overcomes the clamping effects that typically occur when analogous magnetoelectric structures are grown onto rigid substrates (which preclude strain transfer from the magnetostrictive to the ferroelectric layers). The study of the diverse proliferation and differentiation markers evidence that in all the stages of bone formation (cell proliferation, extracellular matrix maturation, and mineralization), the electrical stimulation of the cells results in a remarkably better performance. The results pave the way for novel strategies for remote cell stimulation based on flexible platforms not only in bone regeneration but also in many other applications where electrical cell stimulation may be beneficial (e.g., neurological diseases or skin regeneration).
Collapse
Affiliation(s)
- Oriol Careta
- Departament
de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
| | - Aliona Nicolenco
- Departament
de Física, Universitat Autònoma
de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
- CIDETEC,
Parque Científico y Tecnológico de Gipuzkoa, Paseo Miramón, 191, San Sebastián 20014, Spain
| | - Filippos Perdikos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona E-08193, Spain
| | - Andreu Blanquer
- Departament
de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
| | - Elena Ibañez
- Departament
de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
| | - Eva Pellicer
- Departament
de Física, Universitat Autònoma
de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
| | - Christina Stefani
- Departament
de Física, Universitat Autònoma
de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
| | - Borja Sepúlveda
- Instituto
de Microelectronica de Barcelona (IMB-CNM, CSIC), Campus UAB, Bellaterra, Barcelona E-08193, Spain
| | - Josep Nogués
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona E-08193, Spain
- Institució
Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, Barcelona E-08010, Spain
| | - Jordi Sort
- Departament
de Física, Universitat Autònoma
de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
- Institució
Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, Barcelona E-08010, Spain
| | - Carme Nogués
- Departament
de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès E-08193, Spain
| |
Collapse
|
14
|
Zhu T, Zhou H, Chen X, Zhu Y. Recent advances of responsive scaffolds in bone tissue engineering. Front Bioeng Biotechnol 2023; 11:1296881. [PMID: 38047283 PMCID: PMC10691504 DOI: 10.3389/fbioe.2023.1296881] [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: 09/19/2023] [Accepted: 11/09/2023] [Indexed: 12/05/2023] Open
Abstract
The investigation of bone defect repair has been a significant focus in clinical research. The gradual progress and utilization of different scaffolds for bone repair have been facilitated by advancements in material science and tissue engineering. In recent times, the attainment of precise regulation and targeted drug release has emerged as a crucial concern in bone tissue engineering. As a result, we present a comprehensive review of recent developments in responsive scaffolds pertaining to the field of bone defect repair. The objective of this review is to provide a comprehensive summary and forecast of prospects, thereby contributing novel insights to the field of bone defect repair.
Collapse
Affiliation(s)
| | | | | | - Yuanjing Zhu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
16
|
Marrella A, Suarato G, Fiocchi S, Chiaramello E, Bonato M, Parazzini M, Ravazzani P. Magnetoelectric nanoparticles shape modulates their electrical output. Front Bioeng Biotechnol 2023; 11:1219777. [PMID: 37691903 PMCID: PMC10485842 DOI: 10.3389/fbioe.2023.1219777] [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: 05/09/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Core-shell magnetoelectric nanoparticles (MENPs) have recently gained popularity thanks to their capability in inducing a local electric polarization upon an applied magnetic field and vice versa. This work estimates the magnetoelectrical behavior, in terms of magnetoelectric coupling coefficient (αME), via finite element analysis of MENPs with different shapes under either static (DC bias) and time-variant (AC bias) external magnetic fields. With this approach, the dependence of the magnetoelectrical performance on the MENPs geometrical features can be directly derived. Results show that MENPs with a more elongated morphology exhibits a superior αME if compared with spherical nanoparticles of similar volume, under both stimulation conditions analyzed. This response is due to the presence of a larger surface area at the interface between the magnetostrictive core and piezoelectric shell, and to the MENP geometrical orientation along the direction of the magnetic field. These findings pave a new way for the design of novel high-aspect ratio magnetic nanostructures with an improved magnetoelectric behaviour.
Collapse
Affiliation(s)
| | - G. Suarato
- *Correspondence: A. Marrella, ; G. Suarato,
| | | | | | | | | | | |
Collapse
|
17
|
Zhang YQ, Geng Q, Li C, Wang HC, Ren C, Zhang YF, Bai JS, Pan HB, Cui X, Yao MX, Chen W. Application of piezoelectric materials in the field of bone: a bibliometric analysis. Front Bioeng Biotechnol 2023; 11:1210637. [PMID: 37600300 PMCID: PMC10436523 DOI: 10.3389/fbioe.2023.1210637] [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: 04/23/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
In the past 4 decades, many articles have reported on the effects of the piezoelectric effect on bone formation and the research progress of piezoelectric biomaterials in orthopedics. The purpose of this study is to comprehensively evaluate all existing research and latest developments in the field of bone piezoelectricity, and to explore potential research directions in this area. To assess the overall trend in this field over the past 40 years, this study comprehensively collected literature reviews in this field using a literature retrieval program, applied bibliometric methods and visual analysis using CiteSpace and R language, and identified and investigated publications based on publication year (1984-2022), type of literature, language, country, institution, author, journal, keywords, and citation counts. The results show that the most productive countries in this field are China, the United States, and Italy. The journal with the most publications in the field of bone piezoelectricity is the International Journal of Oral & Maxillofacial Implants, followed by Implant Dentistry. The most productive authors are Lanceros-Méndez S, followed by Sohn D.S. Further research on the results obtained leads to the conclusion that the research direction of this field mainly includes piezoelectric surgery, piezoelectric bone tissue engineering scaffold, manufacturing artificial cochleae for hearing loss patients, among which the piezoelectric bone tissue engineering scaffold is the main research direction in this field. The piezoelectric materials involved in this direction mainly include polyhydroxybutyrate valerate, PVDF, and BaTiO3.
Collapse
Affiliation(s)
- Yu-Qin Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| | - Qian Geng
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chao Li
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| | - Hai-Cheng Wang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| | - Chuan Ren
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| | - Yi-Fan Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| | - Jun-Sheng Bai
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| | - Hao-Bo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xu Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Meng-Xuan Yao
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| | - Wei Chen
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, Hebei, China
| |
Collapse
|
18
|
Zhang Y, Tang J, Fang W, Zhao Q, Lei X, Zhang J, Chen J, Li Y, Zuo Y. Synergetic Effect of Electrical and Topographical Cues in Aniline Trimer-Based Polyurethane Fibrous Scaffolds on Tissue Regeneration. J Funct Biomater 2023; 14:jfb14040185. [PMID: 37103277 PMCID: PMC10146274 DOI: 10.3390/jfb14040185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023] Open
Abstract
Processibility and biodegradability of conductive polymers are major concerns when they are applied to tissue regeneration. This study synthesizes dissolvable and conductive aniline trimer-based polyurethane copolymers (DCPU) and processes them into scaffolds by using electrospinning with different patterns (random, oriented, and latticed). The effects of topographic cue changes on electrical signal transmission and further regulation of cell behaviors concerning bone tissue are researched. Results show that DCPU fibrous scaffolds possessed good hydrophilicity, swelling capacity, elasticity, and fast biodegradability in enzymatic liquid. In addition, the conductivity and efficiency of electrical signal transmission can be tuned by changing the surface’s topological structure. Among them, oriented DCPU scaffolds (DCPU-O) showed the best conductivity with the lowest ionic resistance value. Furthermore, the viability and proliferation results of bone mesenchymal stem cells (BMSCs) demonstrate a significant increase on three DCPU scaffolds compared to AT-free scaffolds (DPU-R). Especially, DCPU-O scaffolds exhibit superior abilities to promote cell proliferation because of their unique surface topography and excellent electroactivity. Concurrently, the DCPU-O scaffolds can synergistically promote osteogenic differentiation in terms of osteogenic differentiation and gene expression levels when combined with electrical stimulation. Together, these results suggest a promising use of DCPU-O fibrous scaffolds in the application of tissue regeneration.
Collapse
|
19
|
Durán-Rey D, Brito-Pereira R, Ribeiro C, Ribeiro S, Sánchez-Margallo JA, Crisóstomo V, Irastorza I, Silván U, Lanceros-Méndez S, Sánchez-Margallo FM. Development of Silk Fibroin Scaffolds for Vascular Repair. Biomacromolecules 2023; 24:1121-1130. [PMID: 36754364 PMCID: PMC10016106 DOI: 10.1021/acs.biomac.2c01124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/05/2023] [Indexed: 02/10/2023]
Abstract
Silk fibroin (SF) is a biocompatible natural protein with excellent mechanical characteristics. SF-based biomaterials can be structured using a number of techniques, allowing the tuning of materials for specific biomedical applications. In this study, SF films, porous membranes, and electrospun membranes were produced using solvent-casting, salt-leaching, and electrospinning methodologies, respectively. SF-based materials were subjected to physicochemical and biological characterizations to determine their suitability for tissue regeneration applications. Mechanical analysis showed stress-strain curves of brittle materials in films and porous membranes, while electrospun membranes featured stress-strain curves typical of ductile materials. All samples showed similar chemical composition, melting transition, hydrophobic behavior, and low cytotoxicity levels, regardless of their architecture. Finally, all of the SF-based materials promote the proliferation of human umbilical vein endothelial cells (HUVECs). These findings demonstrate the different relationship between HUVEC behavior and the SF sample's topography, which can be taken advantage of for the design of vascular implants.
Collapse
Affiliation(s)
- David Durán-Rey
- Jesús
Usón Minimally Invasive Surgery Centre, Cáceres 10004, Spain
| | - Ricardo Brito-Pereira
- CMEMS-UMinho, University of Minho, 4800-058 Guimarães, Portugal
- LABBELS-Associate
Laboratory, 4710-057 Braga/Guimarães, Portugal
- CF−UM-UP−Physics
Centre of Minho and Porto Universities and LaPMET−Laboratory
of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
- IB-S,
Institute of Science and Innovation for Bio-Sustainability, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Clarisse Ribeiro
- CF−UM-UP−Physics
Centre of Minho and Porto Universities and LaPMET−Laboratory
of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
| | - Sylvie Ribeiro
- CF−UM-UP−Physics
Centre of Minho and Porto Universities and LaPMET−Laboratory
of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
| | - Juan A. Sánchez-Margallo
- Jesús
Usón Minimally Invasive Surgery Centre, Cáceres 10004, Spain
- RICORS-TERAV
Network, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Verónica Crisóstomo
- Jesús
Usón Minimally Invasive Surgery Centre, Cáceres 10004, Spain
- Centro
de
Investigación Biomédica en Red de Enfermedades Cardiovasculares
(CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
- RICORS-TERAV
Network, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Igor Irastorza
- CF−UM-UP−Physics
Centre of Minho and Porto Universities and LaPMET−Laboratory
of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
- Cell
Biology and Histology Department, Faculty
of Medicine, Leioa 48940, Spain
| | - Unai Silván
- BCMaterials,
Basque Center for Materials, Applications
and Nanostructures, UPV/EHU
Science Park, Leioa 48940, Spain
- Ikerbasque,
Basque Foundation for Science, Bilbao 48009, Spain
| | - Senentxu Lanceros-Méndez
- CF−UM-UP−Physics
Centre of Minho and Porto Universities and LaPMET−Laboratory
of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
- BCMaterials,
Basque Center for Materials, Applications
and Nanostructures, UPV/EHU
Science Park, Leioa 48940, Spain
- Ikerbasque,
Basque Foundation for Science, Bilbao 48009, Spain
| | - Francisco M. Sánchez-Margallo
- Jesús
Usón Minimally Invasive Surgery Centre, Cáceres 10004, Spain
- Centro
de
Investigación Biomédica en Red de Enfermedades Cardiovasculares
(CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
- RICORS-TERAV
Network, Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
20
|
Timralieva A, Moskalenko IV, Nesterov PV, Shilovskikh VV, Novikov AS, Konstantinova EA, Kokorin AI, Skorb EV. Melamine Barbiturate as a Light-Induced Nanostructured Supramolecular Material for a Bioinspired Oxygen and Organic Radical Trap and Stabilization. ACS OMEGA 2023; 8:8276-8284. [PMID: 36910956 PMCID: PMC9996620 DOI: 10.1021/acsomega.2c06510] [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: 10/10/2022] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Use of coantioxidant systems is a prospective way to increase the effectiveness of antioxidant species in tissue repair and regeneration. In this paper, we introduce a novel scheme of a reactive oxygen species (ROS) trap and neutralization during self-assembly of supramolecular melamine barbiturate material. The performed reaction chain mimics the biological process of ROS generation in key stages and enables one to obtain stable hydroperoxyl and organic radicals in a melamine barbiturate structure. Melamine barbiturate also neutralizes hydroxyl radicals, and the effectiveness of the radical trap is controlled with ROS scavenger incorporation. The number of radicals dramatically increases during light-inducing and depends on pH. The proposed scheme of the ROS trap and neutralization opens a way to the use of supramolecular assemblies as a component of coantioxidant systems and a source of organic radicals.
Collapse
Affiliation(s)
- Alexandra
A. Timralieva
- Infochemistry
Scientific Center of ITMO University, Lomonosova str. 9, St. Petersburg 191002, Russia
| | - Ivan V. Moskalenko
- Infochemistry
Scientific Center of ITMO University, Lomonosova str. 9, St. Petersburg 191002, Russia
| | - Pavel V. Nesterov
- Infochemistry
Scientific Center of ITMO University, Lomonosova str. 9, St. Petersburg 191002, Russia
| | - Vladimir V. Shilovskikh
- Infochemistry
Scientific Center of ITMO University, Lomonosova str. 9, St. Petersburg 191002, Russia
| | - Alexander S. Novikov
- Infochemistry
Scientific Center of ITMO University, Lomonosova str. 9, St. Petersburg 191002, Russia
| | - Elizaveta A. Konstantinova
- Physics
Department, M. V. Lomonosov Moscow State
University, Leninskie Gory 1/2, Moscow 119991, Russia
- Institute
of Nano-, Bio-, Information, Cognitive and Socio-humanistic Sciences
and Technologies, Moscow Institute of Physics
and Technology, Dolgoprudny 141701 Moscow Region, Russia
| | - Alexander I. Kokorin
- Infochemistry
Scientific Center of ITMO University, Lomonosova str. 9, St. Petersburg 191002, Russia
- N.
N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Kosygin st. 4, Moscow 119991, Russia
- Plekhanov
Russian University of Economics, Stremyannyi per. 36, Moscow 115093, Russia
| | - Ekaterina V. Skorb
- Infochemistry
Scientific Center of ITMO University, Lomonosova str. 9, St. Petersburg 191002, Russia
| |
Collapse
|
21
|
Huang H, Lyu Y, Nan K. Soft robot-enabled controlled release of oral drug formulations. SOFT MATTER 2023; 19:1269-1281. [PMID: 36723379 DOI: 10.1039/d2sm01624a] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The creation of highly effective oral drug delivery systems (ODDSs) has long been the main objective of pharmaceutical research. Multidisciplinary efforts involving materials, electronics, control, and pharmaceutical sciences encourage the development of robot-enabled ODDSs. Compared with conventional rigid robots, soft robots potentially offer better mechanical compliance and biocompatibility with biological tissues, more versatile shape control and maneuverability, and multifunctionality. In this paper, we first describe and highlight the importance of manipulating drug release kinetics, i.e. pharmaceutical kinetics. We then introduce an overview of state-of-the-art soft robot-based ODDSs comprising resident, shape-programming, locomotive, and integrated soft robots. Finally, the challenges and outlook regarding future soft robot-based ODDS development are discussed.
Collapse
Affiliation(s)
- Hao Huang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yidan Lyu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Kewang Nan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
22
|
Marques-Almeida T, Fernandes HJR, Lanceros-Mendez S, Ribeiro C. Surface charge and dynamic mechanoelectrical stimuli improves adhesion, proliferation and differentiation of neuron-like cells. J Mater Chem B 2022; 11:144-153. [PMID: 36441601 DOI: 10.1039/d2tb01933g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neuronal diseases and trauma are among the current major health-care problems. Patients frequently develop an irreversible state of neuronal disfunction that lacks treatment, strongly reducing life quality and expectancy. Novel strategies are thus necessary and tissue engineering research is struggling to provide alternatives to current treatments, making use of biomaterials capable to provide cell supports and active stimuli to develop permissive environments for neural regeneration. As neuronal cells are naturally found in electrical microenvironments, the electrically active materials can pave the way for new and effective neuroregenerative therapies. In this work the influence of piezoelectric poly(vinylidene fluoride) with different surface charges and dynamic mechanoelectrical stimuli on neuron-like cells adhesion, proliferation and differentiation was addressed. It is successfully demonstrated that both surface charge and electrically active dynamic microenvironments can be suitable to improve neuron-like cells adhesion, proliferation, and differentiation. These findings provide new knowledge to develop effective approaches for preclinical applications.
Collapse
Affiliation(s)
- T Marques-Almeida
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, 4710-057, Braga, Portugal. .,LaPMET-Laboratory of Physics for Materials and Emergent Technologies, University of Minho, 4710-057, Braga, Portugal
| | - H J R Fernandes
- UK Dementia Research Institute, University of Cambridge, Department of Clinical Neurosciences, Cambridge Biomedical Campus, Cambridge, CB2 0AH, UK
| | - S Lanceros-Mendez
- BCMaterials, Basque Centre for Materials and Applications, UPV/EHU Science Park, Leioa, 48940, Spain. .,IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain
| | - C Ribeiro
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, 4710-057, Braga, Portugal. .,LaPMET-Laboratory of Physics for Materials and Emergent Technologies, University of Minho, 4710-057, Braga, Portugal
| |
Collapse
|
23
|
Cojocaru FD, Balan V, Verestiuc L. Advanced 3D Magnetic Scaffolds for Tumor-Related Bone Defects. Int J Mol Sci 2022; 23:16190. [PMID: 36555827 PMCID: PMC9788029 DOI: 10.3390/ijms232416190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/04/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The need for bone substitutes is a major challenge as the incidence of serious bone disorders is massively increasing, mainly attributed to modern world problems, such as obesity, aging of the global population, and cancer incidence. Bone cancer represents one of the most significant causes of bone defects, with reserved prognosis regarding the effectiveness of treatments and survival rate. Modern therapies, such as hyperthermia, immunotherapy, targeted therapy, and magnetic therapy, seem to bring hope for cancer treatment in general, and bone cancer in particular. Mimicking the composition of bone to create advanced scaffolds, such as bone substitutes, proved to be insufficient for successful bone regeneration, and a special attention should be given to control the changes in the bone tissue micro-environment. The magnetic manipulation by an external field can be a promising technique to control this micro-environment, and to sustain the proliferation and differentiation of osteoblasts, promoting the expression of some growth factors, and, finally, accelerating new bone formation. By incorporating stimuli responsive nanocarriers in the scaffold's architecture, such as magnetic nanoparticles functionalized with bioactive molecules, their behavior can be rigorously controlled under external magnetic driving, and stimulates the bone tissue formation.
Collapse
Affiliation(s)
| | | | - Liliana Verestiuc
- Biomedical Sciences Department, Faculty of Medical Bioengineering, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 9-13 Kogalniceanu Street, 700454 Iasi, Romania
| |
Collapse
|
24
|
Kumari S, Katiyar S, Darshna, Anand A, Singh D, Singh BN, Mallick SP, Mishra A, Srivastava P. Design strategies for composite matrix and multifunctional polymeric scaffolds with enhanced bioactivity for bone tissue engineering. Front Chem 2022; 10:1051678. [PMID: 36518978 PMCID: PMC9742444 DOI: 10.3389/fchem.2022.1051678] [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: 09/25/2022] [Accepted: 11/14/2022] [Indexed: 09/19/2023] Open
Abstract
Over the past few decades, various bioactive material-based scaffolds were investigated and researchers across the globe are actively involved in establishing a potential state-of-the-art for bone tissue engineering applications, wherein several disciplines like clinical medicine, materials science, and biotechnology are involved. The present review article's main aim is to focus on repairing and restoring bone tissue defects by enhancing the bioactivity of fabricated bone tissue scaffolds and providing a suitable microenvironment for the bone cells to fasten the healing process. It deals with the various surface modification strategies and smart composite materials development that are involved in the treatment of bone tissue defects. Orthopaedic researchers and clinicians constantly focus on developing strategies that can naturally imitate not only the bone tissue architecture but also its functional properties to modulate cellular behaviour to facilitate bridging, callus formation and osteogenesis at critical bone defects. This review summarizes the currently available polymeric composite matrices and the methods to improve their bioactivity for bone tissue regeneration effectively.
Collapse
Affiliation(s)
- Shikha Kumari
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Soumya Katiyar
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Darshna
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Aditya Anand
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Divakar Singh
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Bhisham Narayan Singh
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sarada Prasanna Mallick
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh, India
| | - Abha Mishra
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | | |
Collapse
|
25
|
Pryadko A, Mukhortova YR, Chernozem RV, Shlapakova LE, Wagner DV, Romanyuk K, Gerasimov EY, Kholkin A, Surmenev RA, Surmeneva MA. Comprehensive Study on the Reinforcement of Electrospun PHB Scaffolds with Composite Magnetic Fe 3O 4-rGO Fillers: Structure, Physico-Mechanical Properties, and Piezoelectric Response. ACS OMEGA 2022; 7:41392-41411. [PMID: 36406497 PMCID: PMC9670262 DOI: 10.1021/acsomega.2c05184] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
This is a comprehensive study on the reinforcement of electrospun poly(3-hydroxybutyrate) (PHB) scaffolds with a composite filler of magnetite-reduced graphene oxide (Fe3O4-rGO). The composite filler promoted the increase of average fiber diameters and decrease of the degree of crystallinity of hybrid scaffolds. The decrease in the fiber diameter enhanced the ductility and mechanical strength of scaffolds. The surface electric potential of PHB/Fe3O4-rGO composite scaffolds significantly increased with increasing fiber diameter owing to a greater number of polar functional groups. The changes in the microfiber diameter did not have any influence on effective piezoresponses of composite scaffolds. The Fe3O4-rGO filler imparted high saturation magnetization (6.67 ± 0.17 emu/g) to the scaffolds. Thus, magnetic PHB/Fe3O4-rGO composite scaffolds both preserve magnetic properties and provide a piezoresponse, whereas varying the fiber diameter offers control over ductility and surface electric potential.
Collapse
Affiliation(s)
- Artyom
S. Pryadko
- Physical
Materials Science and Composite Materials Center, Research School
of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| | - Yulia R. Mukhortova
- Physical
Materials Science and Composite Materials Center, Research School
of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| | - Roman V. Chernozem
- Physical
Materials Science and Composite Materials Center, Research School
of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| | - Lada E. Shlapakova
- Physical
Materials Science and Composite Materials Center, Research School
of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| | | | - Konstantin Romanyuk
- Department
of Physics & CICECO−Aveiro Institute of Materials, University of Aveiro, Aveiro3810-193, Portugal
- International
Research & Development Center of Piezo- and Magnetoelectric Materials,
Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| | | | - Andrei Kholkin
- School
of Natural Sciences and Mathematics, Ural
Federal University, Ekaterinburg620000, Russia
- International
Research & Development Center of Piezo- and Magnetoelectric Materials,
Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| | - Roman A. Surmenev
- Physical
Materials Science and Composite Materials Center, Research School
of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
- International
Research & Development Center of Piezo- and Magnetoelectric Materials,
Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| | - Maria A. Surmeneva
- Physical
Materials Science and Composite Materials Center, Research School
of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
- International
Research & Development Center of Piezo- and Magnetoelectric Materials,
Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk634050, Russia
| |
Collapse
|
26
|
Effect of Piezoelectric BaTiO 3 Filler on Mechanical and Magnetoelectric Properties of Zn 0.25Co 0.75Fe 2O 4/PVDF-TrFE Composites. Polymers (Basel) 2022; 14:polym14224807. [PMID: 36432934 PMCID: PMC9695481 DOI: 10.3390/polym14224807] [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: 10/12/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Polymer-based multiferroics, combining magnetic and piezoelectric properties, are studied experimentally-from synthesis to multi-parameter characterization-in view of their prospects for fabricating biocompatible scaffolds. The main advantage of these systems is facile generation of mechanical deformations and electric signals in response to external magnetic fields. Herein, we address the composites based on PVDF-TrFE polymer matrices filled with a combination of piezoelectric (BaTiO3, BTO) and/or ferrimagnetic (Zn0.25Co0.75Fe2O4, ZCFO) particles. It is shown that the presence of BTO micron-size particles favors stripe-type structuring of the ZCFO filler and enhances the magnetoelectric response of the sample up to 18.6 mV/(cm∙Oe). Besides that, the admixing of BTO particles is crucial because the mechanical properties of the composite filled with only ZCFO is much less efficient in transforming magnetic excitations into the mechanical and electric responses. Attention is focused on the local surfacial mechanical properties since those, to a great extent, determine the fate of stem cells cultivated on these surfaces. The nano-indentation tests are accomplished with the aid of scanning probe microscopy technique. With their proven suitable mechanical properties, a high level of magnetoelectric conversion and also biocompatibility, the composites of the considered type are enticing as the materials for multiferroic-based polymer scaffolds.
Collapse
|
27
|
Guillot-Ferriols M, García-Briega MI, Tolosa L, Costa CM, Lanceros-Méndez S, Gómez Ribelles JL, Gallego Ferrer G. Magnetically Activated Piezoelectric 3D Platform Based on Poly(Vinylidene) Fluoride Microspheres for Osteogenic Differentiation of Mesenchymal Stem Cells. Gels 2022; 8:680. [PMID: 36286181 PMCID: PMC9602007 DOI: 10.3390/gels8100680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) osteogenic commitment before injection enhances bone regeneration therapy results. Piezoelectric stimulation may be an effective cue to promote MSCs pre-differentiation, and poly(vinylidene) fluoride (PVDF) cell culture supports, when combined with CoFe2O4 (CFO), offer a wireless in vitro stimulation strategy. Under an external magnetic field, CFO shift and magnetostriction deform the polymer matrix varying the polymer surface charge due to the piezoelectric effect. To test the effect of piezoelectric stimulation on MSCs, our approach is based on a gelatin hydrogel with embedded MSCs and PVDF-CFO electroactive microspheres. Microspheres were produced by electrospray technique, favouring CFO incorporation, crystallisation in β-phase (85%) and a crystallinity degree of around 55%. The absence of cytotoxicity of the 3D construct was confirmed 24 h after cell encapsulation. Cells were viable, evenly distributed in the hydrogel matrix and surrounded by microspheres, allowing local stimulation. Hydrogels were stimulated using a magnetic bioreactor, and no significant changes were observed in MSCs proliferation in the short or long term. Nevertheless, piezoelectric stimulation upregulated RUNX2 expression after 7 days, indicating the activation of the osteogenic differentiation pathway. These results open the door for optimising a stimulation protocol allowing the application of the magnetically activated 3D electroactive cell culture support for MSCs pre-differentiation before transplantation.
Collapse
Affiliation(s)
- Maria Guillot-Ferriols
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Carlos III Health Institute (CIBER-BBN, ISCIII), 46022 Valencia, Spain
| | - María Inmaculada García-Briega
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Carlos III Health Institute (CIBER-BBN, ISCIII), 46022 Valencia, Spain
| | - Laia Tolosa
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Carlos III Health Institute (CIBER-BBN, ISCIII), 46022 Valencia, Spain
- Experimental Hepatology Unit, Health Research Institute La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Carlos M. Costa
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, 4710-057 Braga, Portugal
- Laboratory of Physics for Materials and Emergent Technologies, LapMET, University of Minho, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, 4710-057 Braga, Portugal
| | - Senentxu Lanceros-Méndez
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, 4710-057 Braga, Portugal
- Laboratory of Physics for Materials and Emergent Technologies, LapMET, University of Minho, 4710-057 Braga, Portugal
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - José Luis Gómez Ribelles
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Carlos III Health Institute (CIBER-BBN, ISCIII), 46022 Valencia, Spain
| | - Gloria Gallego Ferrer
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Carlos III Health Institute (CIBER-BBN, ISCIII), 46022 Valencia, Spain
| |
Collapse
|
28
|
Durán-Rey D, Brito-Pereira R, Ribeiro C, Ribeiro S, Sánchez-Margallo JA, Crisóstomo V, Irastorza I, Silván U, Lanceros-Méndez S, Sánchez-Margallo FM. Development and evaluation of different electroactive poly(vinylidene fluoride) architectures for endothelial cell culture. Front Bioeng Biotechnol 2022; 10:1044667. [PMID: 36338140 PMCID: PMC9626752 DOI: 10.3389/fbioe.2022.1044667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/07/2022] [Indexed: 11/28/2022] Open
Abstract
Tissue engineering (TE) aims to develop structures that improve or even replace the biological functions of tissues and organs. Mechanical properties, physical-chemical characteristics, biocompatibility, and biological performance of the materials are essential factors for their applicability in TE. Poly(vinylidene fluoride) (PVDF) is a thermoplastic polymer that exhibits good mechanical properties, high biocompatibility and excellent thermal properties. However, PVDF structuring, and the corresponding processing methods used for its preparation are known to significantly influence these characteristics. In this study, doctor blade, salt-leaching, and electrospinning processing methods were used to produce PVDF-based structures in the form of films, porous membranes, and fiber scaffolds, respectively. These PVDF scaffolds were subjected to a variety of characterizations and analyses, including physicochemical analysis, contact angle measurement, cytotoxicity assessment and cell proliferation. All prepared PVDF scaffolds are characterized by a mechanical response typical of ductile materials. PVDF films displayed mostly vibration modes for the a-phase, while the remaining PVDF samples were characterized by a higher content of electroactive β-phase due the low temperature solvent evaporation during processing. No significant variations have been observed between the different PVDF membranes with respect to the melting transition. In addition, all analysed PVDF samples present a hydrophobic behavior. On the other hand, cytotoxicity assays confirm that cell viability is maintained independently of the architecture and processing method. Finally, all the PVDF samples promote human umbilical vein endothelial cells (HUVECs) proliferation, being higher on the PVDF film and electrospun randomly-oriented membranes. These findings demonstrated the importance of PVDF topography on HUVEC behavior, which can be used for the design of vascular implants.
Collapse
Affiliation(s)
- David Durán-Rey
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Ricardo Brito-Pereira
- CMEMS-UMinho, University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga/Guimarães, Portugal
- CF-UM-UP, Physics Centre of Minho and Porto Universities, University of Minho—Campus de Gualtar, Braga, Portugal
- IB-S Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Clarisse Ribeiro
- CF-UM-UP, Physics Centre of Minho and Porto Universities, University of Minho—Campus de Gualtar, Braga, Portugal
- LaPMET—Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, Portugal
| | - Sylvie Ribeiro
- CF-UM-UP, Physics Centre of Minho and Porto Universities, University of Minho—Campus de Gualtar, Braga, Portugal
- LaPMET—Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, Portugal
| | - Juan A. Sánchez-Margallo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
- RICORS-TERAV Network, Instituto de Salud Carlos III, Madrid, Spain
| | - Verónica Crisóstomo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- RICORS-TERAV Network, Instituto de Salud Carlos III, Madrid, Spain
| | - Igor Irastorza
- CF-UM-UP, Physics Centre of Minho and Porto Universities, University of Minho—Campus de Gualtar, Braga, Portugal
- Cell Biology and Histology Department, Faculty of Medicine, Leioa, Spain
| | - Unai Silván
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Senentxu Lanceros-Méndez
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Francisco M. Sánchez-Margallo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- RICORS-TERAV Network, Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Francisco M. Sánchez-Margallo,
| |
Collapse
|
29
|
Nikitin AA, Ivanova AV, Semkina AS, Lazareva PA, Abakumov MA. Magneto-Mechanical Approach in Biomedicine: Benefits, Challenges, and Future Perspectives. Int J Mol Sci 2022; 23:11134. [PMID: 36232435 PMCID: PMC9569787 DOI: 10.3390/ijms231911134] [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: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
The magneto-mechanical approach is a powerful technique used in many different applications in biomedicine, including remote control enzyme activity, cell receptors, cancer-selective treatments, mechanically-activated drug releases, etc. This approach is based on the use of a combination of magnetic nanoparticles and external magnetic fields that have led to the movement of such nanoparticles with torques and forces (enough to change the conformation of biomolecules or even break weak chemical bonds). However, despite many theoretical and experimental works on this topic, it is difficult to predict the magneto-mechanical effects in each particular case, while the important results are scattered and often cannot be translated to other experiments. The main reason is that the magneto-mechanical effect is extremely sensitive to changes in any parameter of magnetic nanoparticles and the environment and changes in the parameters of the applied magnetic field. Thus, in this review, we (1) summarize and propose a simplified theoretical explanation of the main factors affecting the efficiency of the magneto-mechanical approach; (2) discuss the nature of the MNP-mediated mechanical forces and their order of magnitude; (3) show some of the main applications of the magneto-mechanical approach in the control over the properties of biological systems.
Collapse
Affiliation(s)
- Aleksey A. Nikitin
- Laboratory of Biomedical Nanomaterials, National University of Science and Technology (MISIS), 119049 Moscow, Russia
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Anna V. Ivanova
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Alevtina S. Semkina
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Polina A. Lazareva
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Maxim A. Abakumov
- Department of Medical Nanobiotechnology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
30
|
Zhang Y, Li J, Habibovic P. Magnetically responsive nanofibrous ceramic scaffolds for on-demand motion and drug delivery. Bioact Mater 2022; 15:372-381. [PMID: 35386339 PMCID: PMC8958423 DOI: 10.1016/j.bioactmat.2022.02.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/04/2022] [Accepted: 02/25/2022] [Indexed: 12/17/2022] Open
Abstract
Smart biomaterials, featuring not only bioactivity, but also dynamic responsiveness to external stimuli, are desired for biomedical applications, such as regenerative medicine, and hold great potential to expand the boundaries of the modern clinical practice. Herein, a magnetically responsive three-dimensional scaffold with sandwich structure is developed by using hydroxyapatite (HA) nanowires and ferrosoferric oxide (Fe3O4) nanoparticles as building blocks. The magnetic HA/Fe3O4 scaffold is fully inorganic in nature, but shows polymeric hydrogel-like characteristics including a 3D fibrous network that is highly porous (>99.7% free volume), deformable (50% deformation) and elastic, and tunable stiffness. The magnetic HA/Fe3O4 scaffold has been shown to execute multimodal motion upon exposure to an external magnetic field including shape transformation, rolling and somersault. In addition, we have demonstrated that the magnetic scaffold can serve as a smart carrier for remotely controlled, on-demand delivery of compounds including an organic dye and a protein. Finally, the magnetic scaffold has exhibited good biocompatibility, supporting the attachment and proliferation of human mesenchymal stromal cells, thereby showing great potential as smart biomaterials for a variety of biomedical applications.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Instructive Biomaterials Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Jiaping Li
- Department of Instructive Biomaterials Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
- Department of Complex Tissue Regeneration, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
31
|
Meira RM, Correia DM, García Díez A, Lanceros-Mendez S, Ribeiro C. Ionic liquid-based electroactive materials: a novel approach for cardiac tissue engineering strategies. J Mater Chem B 2022; 10:6472-6482. [PMID: 35968772 DOI: 10.1039/d2tb01155g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cardiac tissue regeneration strategies are increasingly taking advantage of electroactive scaffolds to actively recreate the tissue microenvironment. In this context, this work reports on advanced materials based on two different ionic liquids (ILs), 2-hydroxyethyl-trimethylammonium dihydrogen phosphate ([Ch][DHP]) and choline bis(trifluoromethylsulfonyl)imide ([Ch][TFSI]), combined with poly(vinylidene fluoride-co-trifluoroethylene) (P(VDF-TrFE)) for the development of ionic electroactive IL/polymer hybrid materials for cardiac tissue engineering (TE). The morphological, physico-chemical, thermal and electrical properties of the hybrid materials, as well as their potential use as scaffolds for cardiac TE applications, were evaluated. Besides inducing changes in surface topography, roughness and wettability of the composites, the incorporation of [Ch][DHP] and [Ch][TFSI] leads to the increase in surface (σsurface) and volume (σvolume) electrical conductivities. Furthermore, washing the hybrid samples with phosphate-buffered saline solution strongly decreases the σsurface, whereas σsurface and σvolume of the composites remain almost unaltered after exposure to ultraviolet sterilization treatment. Additionally, it is verified that the incorporation of IL influences the P(VDF-TrFE) microstructure and crystallization process, acting as a defect during its crystallization. Cytotoxicity assays revealed that hybrid films based on [Ch][DHP] alone are not cytotoxic. These films also support H9c2 myoblast cell adhesion and proliferation, demonstrating their suitability for cardiac TE strategies based on electroactive microenvironments.
Collapse
Affiliation(s)
- R M Meira
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, 4710-057 Braga, Portugal. .,LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
| | - D M Correia
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, 4710-057 Braga, Portugal. .,Centre of Chemistry, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - A García Díez
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - S Lanceros-Mendez
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - C Ribeiro
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, 4710-057 Braga, Portugal. .,LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
32
|
Martins L, Ródenas-Rochina J, Salazar D, Cardoso VF, Gómez Ribelles JL, Lanceros-Mendez S. Microfluidic Processing of Piezoelectric and Magnetic Responsive Electroactive Microspheres. ACS APPLIED POLYMER MATERIALS 2022; 4:5368-5379. [PMID: 36824683 PMCID: PMC9940114 DOI: 10.1021/acsapm.2c00380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/05/2022] [Indexed: 06/18/2023]
Abstract
Poly(vinylidene fluoride) (PVDF) combined with cobalt ferrite (CFO) particles is one of the most common and effective polymeric magnetoelectric composites. Processing PVDF into its electroactive phase is a mandatory condition for featuring electroactive behavior and specific (post)processing may be needed to achieve this state, although electroactive phase crystallization is favored at processing temperatures below 60 °C. Different techniques are used to process PVDF-CFO nanocomposite structures into microspheres with high CFO dispersion, with microfluidics adding the advantages of high reproducibility, size tunability, and time and resource efficiency. In this work, magnetoelectric microspheres are produced in a one-step approach. We describe the production of high content electroactive phase PVDF and PVDF-CFO microspheres using microfluidic technology. A flow-focusing polydimethylsiloxane device is fabricated based on a 3D printed polylactic acid master, which enables the production of spherical microspheres with mean diameters ranging from 80 to 330 μm. The microspheres feature internal and external cavernous structures and good CFO distribution with an encapsulation efficacy of 80% and prove to be in the electroactive γ-phase with a mean content of 75%. The microspheres produced using this approach show suitable characteristics as active materials for tissue regeneration strategies and other piezoelectric polymer applications.
Collapse
Affiliation(s)
- Luís
Amaro Martins
- CBIT—Centre
for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia 46022, Spain
| | - Joaquín Ródenas-Rochina
- CBIT—Centre
for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia 46022, Spain
| | - Daniel Salazar
- BCMaterials,
Basque Center for Materials Applications and Nanostructures, UPV/EHU Science Park, Leioa 48940, Spain
| | - Vanessa F. Cardoso
- Department
of Physics, Universidade do Minho, Braga 4710-057, Portugal
- CMEMS-UMinho, Universidade do Minho, Guimarães 4800-058, Portugal
| | - José Luis Gómez Ribelles
- CBIT—Centre
for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia 46022, Spain
- Biomedical
Research Networking Center on Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), Madrid 28029, Spain
| | - Senentxu Lanceros-Mendez
- BCMaterials,
Basque Center for Materials Applications and Nanostructures, UPV/EHU Science Park, Leioa 48940, Spain
- IKERBASQUE,
Basque Foundation for Science, Bilbao 48009, Spain
| |
Collapse
|
33
|
Xia G, Song B, Fang J. Electrical Stimulation Enabled via Electrospun Piezoelectric Polymeric Nanofibers for Tissue Regeneration. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9896274. [PMID: 36061820 PMCID: PMC9394050 DOI: 10.34133/2022/9896274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/08/2022] [Indexed: 11/22/2022]
Abstract
Electrical stimulation has demonstrated great effectiveness in the modulation of cell fate in vitro and regeneration therapy in vivo. Conventionally, the employment of electrical signal comes with the electrodes, battery, and connectors in an invasive fashion. This tedious procedure and possible infection hinder the translation of electrical stimulation technologies in regenerative therapy. Given electromechanical coupling and flexibility, piezoelectric polymers can overcome these limitations as they can serve as a self-powered stimulator via scavenging mechanical force from the organism and external stimuli wirelessly. Wireless electrical cue mediated by electrospun piezoelectric polymeric nanofibers constitutes a promising paradigm allowing the generation of localized electrical stimulation both in a noninvasive manner and at cell level. Recently, numerous studies based on electrospun piezoelectric nanofibers have been carried out in electrically regenerative therapy. In this review, brief introduction of piezoelectric polymer and electrospinning technology is elucidated first. Afterward, we highlight the activating strategies (e.g., cell traction, physiological activity, and ultrasound) of piezoelectric stimulation and the interaction of piezoelectric cue with nonelectrically/electrically excitable cells in regeneration medicine. Then, quantitative comparison of the electrical stimulation effects using various activating strategies on specific cell behavior and various cell types is outlined. Followingly, this review explores the present challenges in electrospun nanofiber-based piezoelectric stimulation for regeneration therapy and summarizes the methodologies which may be contributed to future efforts in this field for the reality of this technology in the clinical scene. In the end, a summary of this review and future perspectives toward electrospun nanofiber-based piezoelectric stimulation in tissue regeneration are elucidated.
Collapse
Affiliation(s)
- Guangbo Xia
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, China
| | - Beibei Song
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, China
| | - Jian Fang
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, China
| |
Collapse
|
34
|
Samadi A, Salati MA, Safari A, Jouyandeh M, Barani M, Singh Chauhan NP, Golab EG, Zarrintaj P, Kar S, Seidi F, Hejna A, Saeb MR. Comparative review of piezoelectric biomaterials approach for bone tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1555-1594. [PMID: 35604896 DOI: 10.1080/09205063.2022.2065409] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023]
Abstract
Bone as a minerals' reservoir and rigid tissue of the body generating red and white blood cells supports various organs. Although the self-regeneration property of bone, it cannot regenerate spontaneously in severe damages and still remains as a challenging issue. Tissue engineering offers several techniques for regenerating damaged bones, where various biomaterials are examined to fabricate scaffolds for bone repair. Piezoelectric characteristic plays a crucial role in repairing and regenerating damaged bone by mimicking the bone niche behavior. Piezoelectric biomaterials show significant potential for bone tissue engineering. Herein we try to have a comparative review on piezoelectric and non-piezoelectric biomaterials used in bone tissue engineering, classified them, and discussed their effects on implanted cells and manufacturing techniques. Especially, Polyvinylidene fluoride (PVDF) and its composites are the most practically used piezoelectric biomaterials for bone regeneration. PVDF and its composites have been summarized and discussed to repair damaged bone tissues.
Collapse
Affiliation(s)
- Ali Samadi
- Department of Polymer Engineering, Faculty of Engineering, Urmia University, Urmia, Iran
| | | | - Amin Safari
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, Iran
| | - Maryam Jouyandeh
- Center of Excellent in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Narendra Pal Singh Chauhan
- Department of Chemistry, Faculty of Science, Bhupal Nobles' University, Udaipur 313002, Rajasthan, India
| | - Elias Ghaleh Golab
- Department of Petroleum Engineering, Omidiyeh Branch, Islamic Azad University, Iran
| | - Payam Zarrintaj
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Kuwait
| | - Farzad Seidi
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Aleksander Hejna
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12 80-233, Gdańsk, Poland
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12 80-233, Gdańsk, Poland
| |
Collapse
|
35
|
Guillot-Ferriols M, Lanceros-Méndez S, Gómez Ribelles JL, Gallego Ferrer G. Electrical stimulation: Effective cue to direct osteogenic differentiation of mesenchymal stem cells? BIOMATERIALS ADVANCES 2022; 138:212918. [PMID: 35913228 DOI: 10.1016/j.bioadv.2022.212918] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/02/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Mesenchymal stem cells (MSCs) play a major role in bone tissue engineering (BTE) thanks to their capacity for osteogenic differentiation and being easily available. In vivo, MSCs are exposed to an electroactive microenvironment in the bone niche, which has piezoelectric properties. The correlation between the electrically active milieu and bone's ability to adapt to mechanical stress and self-regenerate has led to using electrical stimulation (ES) as physical cue to direct MSCs differentiation towards the osteogenic lineage in BTE. This review summarizes the different techniques to electrically stimulate MSCs to induce their osteoblastogenesis in vitro, including general electrical stimulation and substrate mediated stimulation by means of conductive or piezoelectric cell culture supports. Several aspects are covered, including stimulation parameters, treatment times and cell culture media to summarize the best conditions for inducing MSCs osteogenic commitment by electrical stimulation, from a critical point of view. Electrical stimulation activates different signaling pathways, including bone morphogenetic protein (BMP) Smad-dependent or independent, regulated by mitogen activated protein kinases (MAPK), extracellular signal-regulated kinases (ERK) and p38. The roles of voltage gate calcium channels (VGCC) and integrins are also highlighted according to their application technique and parameters, mainly converging in the expression of RUNX2, the master regulator of the osteogenic differentiation pathway. Despite the evident lack of homogeneity in the approaches used, the ever-increasing scientific evidence confirms ES potential as an osteoinductive cue, mimicking aspects of the in vivo microenvironment and moving one step forward to the translation of this approach into clinic.
Collapse
Affiliation(s)
- M Guillot-Ferriols
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain.
| | - S Lanceros-Méndez
- Centre of Physics of Minho and Porto Universities, Universidade do Minho, 4710-058 Braga, Portugal; BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - J L Gómez Ribelles
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - G Gallego Ferrer
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain; Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| |
Collapse
|
36
|
Silva CA, Fernandes MM, Ribeiro C, Lanceros-Mendez S. Two- and three-dimensional piezoelectric scaffolds for bone tissue engineering. Colloids Surf B Biointerfaces 2022; 218:112708. [DOI: 10.1016/j.colsurfb.2022.112708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/17/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022]
|
37
|
Garello F, Svenskaya Y, Parakhonskiy B, Filippi M. Micro/Nanosystems for Magnetic Targeted Delivery of Bioagents. Pharmaceutics 2022; 14:pharmaceutics14061132. [PMID: 35745705 PMCID: PMC9230665 DOI: 10.3390/pharmaceutics14061132] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 01/09/2023] Open
Abstract
Targeted delivery of pharmaceuticals is promising for efficient disease treatment and reduction in adverse effects. Nano or microstructured magnetic materials with strong magnetic momentum can be noninvasively controlled via magnetic forces within living beings. These magnetic carriers open perspectives in controlling the delivery of different types of bioagents in humans, including small molecules, nucleic acids, and cells. In the present review, we describe different types of magnetic carriers that can serve as drug delivery platforms, and we show different ways to apply them to magnetic targeted delivery of bioagents. We discuss the magnetic guidance of nano/microsystems or labeled cells upon injection into the systemic circulation or in the tissue; we then highlight emergent applications in tissue engineering, and finally, we show how magnetic targeting can integrate with imaging technologies that serve to assist drug delivery.
Collapse
Affiliation(s)
- Francesca Garello
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy;
| | - Yulia Svenskaya
- Science Medical Center, Saratov State University, 410012 Saratov, Russia;
| | - Bogdan Parakhonskiy
- Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium;
| | - Miriam Filippi
- Soft Robotics Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
- Correspondence:
| |
Collapse
|
38
|
Hu X, Liu W, Sun L, Xu S, Wang T, Meng J, Wen T, Liu Q, Liu J, Xu H. Magnetic Nanofibrous Scaffolds Accelerate the Regeneration of Muscle Tissue in Combination with Extra Magnetic Fields. Int J Mol Sci 2022; 23:ijms23084440. [PMID: 35457258 PMCID: PMC9025939 DOI: 10.3390/ijms23084440] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 12/27/2022] Open
Abstract
The reversal of loss of the critical size of skeletal muscle is urgently required using biomaterial scaffolds to guide tissue regeneration. In this work, coaxial electrospun magnetic nanofibrous scaffolds were fabricated, with gelatin (Gel) as the shell of the fiber and polyurethane (PU) as the core. Iron oxide nanoparticles (Mag) of 10 nm diameter were added to the shell and core layer. Myoblast cells (C2C12) were cultured on the magnetic scaffolds and exposed to the applied magnetic fields. A mouse model of skeletal muscle injury was used to evaluate the repair guided by the scaffolds under the magnetic fields. It was shown that VEGF secretion and MyoG expression for the myoblast cells grown on the magnetic scaffolds under the magnetic fields were significantly increased, while, the gene expression of Myh4 was up-regulated. Results from an in vivo study indicated that the process of skeletal muscle regeneration in the mouse muscle injury model was accelerated by using the magnetic actuated strategy, which was verified by histochemical analysis, immunofluorescence staining of CD31, electrophysiological measurement and ultrasound imaging. In conclusion, the integration of a magnetic scaffold combined with the extra magnetic fields enhanced myoblast differentiation and VEGF secretion and accelerated the defect repair of skeletal muscle in situ.
Collapse
Affiliation(s)
- Xuechun Hu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
| | - Wenhao Liu
- Peking Union Medical College, Beijing 100073, China;
| | - Lihong Sun
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
| | - Shilin Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
| | - Tao Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
| | - Jie Meng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
| | - Tao Wen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
| | - Qingqiao Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
| | - Jian Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
- Correspondence: (J.L.); (H.X.); Tel.: +86-10-6915-6437 (H.X.)
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (X.H.); (L.S.); (S.X.); (T.W.); (J.M.); (T.W.); (Q.L.)
- Correspondence: (J.L.); (H.X.); Tel.: +86-10-6915-6437 (H.X.)
| |
Collapse
|
39
|
Sadeghzadeh H, Mehdipour A, Dianat-Moghadam H, Salehi R, Khoshfetrat AB, Hassani A, Mohammadnejad D. PCL/Col I-based magnetic nanocomposite scaffold provides an osteoinductive environment for ADSCs in osteogenic cues-free media conditions. Stem Cell Res Ther 2022; 13:143. [PMID: 35379318 PMCID: PMC8981929 DOI: 10.1186/s13287-022-02816-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/19/2022] [Indexed: 12/20/2022] Open
Abstract
Background The bone tissue engineering (BTE) approach has been introduced as an alternative to conventional treatments for large non-healing bone defects. Magnetism promotes stem cells' adherence to biocompatible scaffolds toward osteoblast differentiation. Furthermore, osteogenic differentiation media are expensive and any changes in its composition affect stem cells differentiation. Moreover, media growth factors possess a short half-life resulting in the rapid loss of their functions in vivo. With the above in mind, we fabricated a multilayered nanocomposite scaffold containing the wild type of Type I collagen (Col I) with endogenous magnetic property to promote osteogenesis in rat ADSCs with the minimum requirement of osteogenic differentiation medium.
Methods Fe3O4 NPs were synthesized by co-precipitation method and characterized using SEM, VSM, and FTIR. Then, a PCL/Col I nanocomposite scaffold entrapping Fe3O4 NPs was fabricated by electrospinning and characterized using SEM, TEM, AFM, VSM, Contact Angle, tensile stretching, and FTIR. ADSCs were isolated from rat adipose tissue and identified by flow cytometry. ADSCs were loaded onto PCL/Col I and PCL/Col I/Fe3O4-scaffolds for 1–3 weeks with/without osteogenic media conditions. The cell viability, cell adhesion, and osteogenic differentiation were evaluated using MTT assay, SEM, DAPI staining, ALP/ARS staining, RT-PCR, and western blotting, respectively. Results SEM, VSM, and FTIR results indicated that Fe3O4 was synthesized in nano-sized (15–30 nm) particles with spherical-shaped morphology and superparamagnetic properties with approved chemical structure as FTIR revealed. According to SEM images, the fabricated magnetic scaffolds consisted of nanofiber (500–700 nm). TEM images have shown the Fe3O4 NPs entrapped in the scaffold's fiber without bead formation. FTIR spectra analysis confirmed the maintenance of the natural structure of Col I, PCL, and Fe3O4 upon electrospinning. AFM data have shown that MNPs incorporation introduced stripe-like topography to nanofibers, while the depth of the grooves has decreased from 800 to 500 nm. Flow cytometry confirmed the phenotype of ADSCs according to their surface markers (i.e., CD29 and CD105). Additionally, Fe3O4 NP improved nanocomposite scaffold strength, wettability, porosity, biocompatibility and also facilitates the ALP activity, calcium-mineralization. Finally, magnetic nanocomposite scaffolds upregulated osteogenic-related genes or proteins’ expression (e.g., Col I, Runx2, OCN, ON, BMP2) in seeded ADSCs with/without osteo-differentiation media conditions. Conclusions Together, these results indicate that Fe3O4 NPs within the natural structure of Col I increase osteogenic differentiation in osteogenic cues-free media conditions. This effect could be translated in vivo toward bone defects healing. These findings support the use of natural ECM materials alongside magnetic particles as composite scaffolds to achieve their full therapeutic potential in BTE treatments. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Hadi Sadeghzadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mehdipour
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Ayla Hassani
- Chemical Engineering Faculty, Sahand University of Technology, 51335-1996, Tabriz, Iran
| | - Daryush Mohammadnejad
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
40
|
Moreira J, Fernandes MM, Carvalho EO, Nicolau A, Lazic V, Nedeljković JM, Lanceros-Mendez S. Exploring electroactive microenvironments in polymer-based nanocomposites to sensitize bacterial cells to low-dose embedded silver nanoparticles. Acta Biomater 2022; 139:237-248. [PMID: 34358697 DOI: 10.1016/j.actbio.2021.07.067] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 01/15/2023]
Abstract
The search for alternative antimicrobial strategies capable of avoiding resistance mechanisms in bacteria are highly needed due to the alarming emergence of antimicrobial resistance. The application of physical stimuli as a mean of sensitizing bacteria for the action of antimicrobials on otherwise resistant bacteria or by allowing the action of low quantity of antimicrobials may be seen as a breakthrough for such purpose. This work proposes the development of antibacterial nanocomposites using the synergy between the electrically active microenvironments, created by a piezoelectric polymer (poly(vinylidene fluoride-co-trifluoroethylene) (PVDF-TrFE)), with green-synthesized silver nanoparticles (AgNPs). The electrical microenvironment is generated via mechanical stimulation of piezoelectric PVDF-TrFE/AgNPs films using a lab-made mechanical bioreactor. The generated material's electrical response further translates to bacterial cells, namely Escherichia coli and Staphylococcus epidermidis which in combination with AgNPs and the specific morphological features of the material induce important antibacterial and antibiofilm activity. Both porous and non-porous PVDF composites have shown antibacterial characteristics when stimulated at a mechanical frequency of 4 Hz being the effect boosted when AgNPs were incorporated in the nanocomposite, reducing in more than 80% the S. epidermidis bacterial growth in planktonic and biofilm form. The electroactive environments sensitize the bacteria allowing the action of a low dose of AgNPs (1.69% (w/w)). Importantly, the material did not compromise the viability of mammalian cells, thus being considered biocompatible. The piezoelectric stimulation of PVDF-based polymeric films may represent a breakthrough in the development of antibacterial coatings for devices used at hospital setting, taking advantage on the use of mechanical stimuli (pressure/touch) to exert antibacterial and antibiofilm activity. STATEMENT OF SIGNIFICANCE: The application of physical methods in alternative to the common chemical ones is seen as a breakthrough for avoiding the emergence of antimicrobial resistance. Antimicrobial strategies that take advantage on the capability of bacteria to sense physical stimuli such as mechanical and electrical cues are scarce. Electroactive nanocomposites comprised of poly(vinylidene fluoride-co-trifluoroethylene (PVDF-TrFE) and green-synthesized silver nanoparticles (AgNPs) were developed to obtain material able to inhibit the colonization of microorganisms. By applying a mechanical stimuli to the nanocomposite, which ultimately mimics movements such as walking or touching, an antimicrobial effect is obtained, resulting from the synergy between the electroactive microenvironments created on the surface of the material and the AgNPs. Such environments sensitize the bacteria to low doses of antimicrobials.
Collapse
Affiliation(s)
- Joana Moreira
- Centre of Physics, University of Minho, Braga 4710-057, Portugal; Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Margarida M Fernandes
- Centre of Physics, University of Minho, Braga 4710-057, Portugal; Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| | - Estela O Carvalho
- Centre of Physics, University of Minho, Braga 4710-057, Portugal; Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Ana Nicolau
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Vesna Lazic
- Vinča Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Jovan M Nedeljković
- Vinča Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Senentxu Lanceros-Mendez
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
41
|
Pryadko AS, Botvin VV, Mukhortova YR, Pariy I, Wagner DV, Laktionov PP, Chernonosova VS, Chelobanov BP, Chernozem RV, Surmeneva MA, Kholkin AL, Surmenev RA. Core-Shell Magnetoactive PHB/Gelatin/Magnetite Composite Electrospun Scaffolds for Biomedical Applications. Polymers (Basel) 2022; 14:polym14030529. [PMID: 35160518 PMCID: PMC8839593 DOI: 10.3390/polym14030529] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/21/2022] Open
Abstract
Novel hybrid magnetoactive composite scaffolds based on poly(3-hydroxybutyrate) (PHB), gelatin, and magnetite (Fe3O4) were fabricated by electrospinning. The morphology, structure, phase composition, and magnetic properties of composite scaffolds were studied. Fabrication procedures of PHB/gelatin and PHB/gelatin/Fe3O4 scaffolds resulted in the formation of both core-shell and ribbon-shaped structure of the fibers. In case of hybrid PHB/gelatin/Fe3O4 scaffolds submicron-sized Fe3O4 particles were observed in the surface layers of the fibers. The X-ray photoelectron spectroscopy results allowed the presence of gelatin on the fiber surface (N/C ratio–0.11) to be revealed. Incubation of the composite scaffolds in saline for 3 h decreased the amount of gelatin on the surface by more than ~75%. The differential scanning calorimetry results obtained for pure PHB scaffolds revealed a characteristic melting peak at 177.5 °C. The presence of gelatin in PHB/gelatin and PHB/gelatin/Fe3O4 scaffolds resulted in the decrease in melting temperature to 168–169 °C in comparison with pure PHB scaffolds due to the core-shell structure of the fibers. Hybrid scaffolds also demonstrated a decrease in crystallinity from 52.3% (PHB) to 16.9% (PHB/gelatin) and 9.2% (PHB/gelatin/Fe3O4). All the prepared scaffolds were non-toxic and saturation magnetization of the composite scaffolds with magnetite was 3.27 ± 0.22 emu/g, which makes them prospective candidates for usage in biomedical applications.
Collapse
Affiliation(s)
- Artyom S. Pryadko
- Physical Materials Science and Composite Materials Center, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.P.); (Y.R.M.); (I.P.); (R.V.C.); (M.A.S.)
| | - Vladimir V. Botvin
- International Research and Development Center “Piezo- and Magnetoelectric Materials”, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Yulia R. Mukhortova
- Physical Materials Science and Composite Materials Center, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.P.); (Y.R.M.); (I.P.); (R.V.C.); (M.A.S.)
- International Research and Development Center “Piezo- and Magnetoelectric Materials”, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Igor Pariy
- Physical Materials Science and Composite Materials Center, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.P.); (Y.R.M.); (I.P.); (R.V.C.); (M.A.S.)
| | - Dmitriy V. Wagner
- Faculty of Radiophysics, National Research Tomsk State University, 634050 Tomsk, Russia;
| | - Pavel P. Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (P.P.L.); (V.S.C.); (B.P.C.)
| | - Vera S. Chernonosova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (P.P.L.); (V.S.C.); (B.P.C.)
| | - Boris P. Chelobanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (P.P.L.); (V.S.C.); (B.P.C.)
- Laboratory of Molecular Medicine, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Roman V. Chernozem
- Physical Materials Science and Composite Materials Center, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.P.); (Y.R.M.); (I.P.); (R.V.C.); (M.A.S.)
- International Research and Development Center “Piezo- and Magnetoelectric Materials”, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Maria A. Surmeneva
- Physical Materials Science and Composite Materials Center, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.P.); (Y.R.M.); (I.P.); (R.V.C.); (M.A.S.)
- International Research and Development Center “Piezo- and Magnetoelectric Materials”, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Andrei L. Kholkin
- International Research and Development Center “Piezo- and Magnetoelectric Materials”, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Department of Physics and CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence: (A.L.K.); (R.A.S.)
| | - Roman A. Surmenev
- Physical Materials Science and Composite Materials Center, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.S.P.); (Y.R.M.); (I.P.); (R.V.C.); (M.A.S.)
- International Research and Development Center “Piezo- and Magnetoelectric Materials”, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Correspondence: (A.L.K.); (R.A.S.)
| |
Collapse
|
42
|
Vukoje I, Lazić V, Sredojević D, Fernandes MM, Lanceros-Mendez S, Ahrenkiel SP, Nedeljković JM. Influence of glucose, sucrose, and dextran coatings on the stability and toxicity of silver nanoparticles. Int J Biol Macromol 2022; 194:461-469. [PMID: 34800522 DOI: 10.1016/j.ijbiomac.2021.11.089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/10/2021] [Accepted: 11/14/2021] [Indexed: 11/30/2022]
Abstract
Aqueous colloids, consisting of 15-30 nm-sized silver nanoparticles (Ag NPs), were prepared using the reducing and stabilizing abilities of glucose, sucrose, and dextran. The long-term stability of coated Ag NPs increases from glucose over sucrose to dextran, i.e., with the increase of the molecular weight of carbohydrate molecules. The density functional theory (DFT) calculations of the partial atomic (Mulliken) charges and adsorption energies are applied to explain the enhanced stability of coated Ag NPs. All coated Ag NPs have a significantly broader concentration range of nontoxic behavior toward pre-osteoblast cells than bare Ag NPs prepared using sodium borohydride. The carbohydrate-coated Ag NPs display the same level of toxic ability against Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria as bare Ag NPs. The differences in toxicity mechanism of the coated and bare Ag NPs are a consequence of the absence and presence of co-occurring Ag+ ions in examined dispersion, respectively.
Collapse
Affiliation(s)
- Ivana Vukoje
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vesna Lazić
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dušan Sredojević
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia; Texas A&M University at Qatar, P.O. Box 23874, Doha, Qatar
| | - Margarida M Fernandes
- Centre of Physics, University of Minho, 4710-057 Braga, Portugal; Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Senentxu Lanceros-Mendez
- Centre of Physics, University of Minho, 4710-057 Braga, Portugal; BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - S Phillip Ahrenkiel
- South Dakota School of Mines and Technology, 501 E. Saint Joseph Street, Rapid City, SD 57701, USA
| | - Jovan M Nedeljković
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
43
|
Yao J, Yao C, Zhang A, Xu X, Wu A, Yang F. Magnetomechanical force: an emerging paradigm for therapeutic applications. J Mater Chem B 2022; 10:7136-7147. [DOI: 10.1039/d2tb00428c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mechanical forces, which play an profound role in cell fate regulation, have prompted the rapid development and popularization of mechanobiology. More recently, magnetic fields in combination with intelligent materials featuring...
Collapse
|
44
|
Hao Z, Xu Z, Wang X, Wang Y, Li H, Chen T, Hu Y, Chen R, Huang K, Chen C, Li J. Biophysical Stimuli as the Fourth Pillar of Bone Tissue Engineering. Front Cell Dev Biol 2021; 9:790050. [PMID: 34858997 PMCID: PMC8630705 DOI: 10.3389/fcell.2021.790050] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 01/12/2023] Open
Abstract
The repair of critical bone defects remains challenging worldwide. Three canonical pillars (biomaterial scaffolds, bioactive molecules, and stem cells) of bone tissue engineering have been widely used for bone regeneration in separate or combined strategies, but the delivery of bioactive molecules has several obvious drawbacks. Biophysical stimuli have great potential to become the fourth pillar of bone tissue engineering, which can be categorized into three groups depending on their physical properties: internal structural stimuli, external mechanical stimuli, and electromagnetic stimuli. In this review, distinctive biophysical stimuli coupled with their osteoinductive windows or parameters are initially presented to induce the osteogenesis of mesenchymal stem cells (MSCs). Then, osteoinductive mechanisms of biophysical transduction (a combination of mechanotransduction and electrocoupling) are reviewed to direct the osteogenic differentiation of MSCs. These mechanisms include biophysical sensing, transmission, and regulation. Furthermore, distinctive application strategies of biophysical stimuli are presented for bone tissue engineering, including predesigned biomaterials, tissue-engineered bone grafts, and postoperative biophysical stimuli loading strategies. Finally, ongoing challenges and future perspectives are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Xu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuan Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Renxin Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kegang Huang
- Wuhan Institute of Proactive Health Management Science, Wuhan, China
| | - Chao Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Orthopedics, Hefeng Central Hospital, Enshi, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
45
|
Jampilek J, Placha D. Advances in Use of Nanomaterials for Musculoskeletal Regeneration. Pharmaceutics 2021; 13:1994. [PMID: 34959276 PMCID: PMC8703496 DOI: 10.3390/pharmaceutics13121994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/24/2022] Open
Abstract
Since the worldwide incidence of bone disorders and cartilage damage has been increasing and traditional therapy has reached its limits, nanomaterials can provide a new strategy in the regeneration of bones and cartilage. The nanoscale modifies the properties of materials, and many of the recently prepared nanocomposites can be used in tissue engineering as scaffolds for the development of biomimetic materials involved in the repair and healing of damaged tissues and organs. In addition, some nanomaterials represent a noteworthy alternative for treatment and alleviating inflammation or infections caused by microbial pathogens. On the other hand, some nanomaterials induce inflammation processes, especially by the generation of reactive oxygen species. Therefore, it is necessary to know and understand their effects in living systems and use surface modifications to prevent these negative effects. This contribution is focused on nanostructured scaffolds, providing a closer structural support approximation to native tissue architecture for cells and regulating cell proliferation, differentiation, and migration, which results in cartilage and bone healing and regeneration.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Daniela Placha
- Nanotechnology Centre, CEET, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 33 Ostrava-Poruba, Czech Republic
- Centre ENET, CEET, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, 708 33 Ostrava-Poruba, Czech Republic
| |
Collapse
|
46
|
Etemadi H, Buchanan JK, Kandile NG, Plieger PG. Iron Oxide Nanoparticles: Physicochemical Characteristics and Historical Developments to Commercialization for Potential Technological Applications. ACS Biomater Sci Eng 2021; 7:5432-5450. [PMID: 34786932 DOI: 10.1021/acsbiomaterials.1c00938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Iron oxide nanoparticles (IONPs) have gained increasing attention in various biomedical and industrial sectors due to their physicochemical and magnetic properties. In the biomedical field, IONPs are being developed for enzyme/protein immobilization, magnetofection, cell labeling, DNA detection, and tissue engineering. However, in some established areas, such as magnetic resonance imaging (MRI), magnetic drug targeting (MDT), magnetic fluid hyperthermia (MFH), immunomagnetic separation (IMS), and magnetic particle imaging (MPI), IONPs have crossed from the research bench, received clinical approval, and have been commercialized. Additionally, in industrial sectors IONP-based fluids (ferrofluids) have been marketed in electronic and mechanical devices for some time. This review explores the historical evolution of IONPs to their current state in biomedical and industrial applications.
Collapse
Affiliation(s)
- Hossein Etemadi
- School of Fundamental Sciences, Massey University, Private Bag 11 222, Palmerston North 4410, New Zealand
| | - Jenna K Buchanan
- School of Fundamental Sciences, Massey University, Private Bag 11 222, Palmerston North 4410, New Zealand
| | - Nadia G Kandile
- Department of Chemistry, Faculty of Women, Ain Shams University, Heliopolis 11757, Cairo, Egypt
| | - Paul G Plieger
- School of Fundamental Sciences, Massey University, Private Bag 11 222, Palmerston North 4410, New Zealand
| |
Collapse
|
47
|
Tai Y, Banerjee A, Goodrich R, Jin L, Nam J. Development and Utilization of Multifunctional Polymeric Scaffolds for the Regulation of Physical Cellular Microenvironments. Polymers (Basel) 2021; 13:3880. [PMID: 34833179 PMCID: PMC8624881 DOI: 10.3390/polym13223880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 10/30/2021] [Accepted: 11/06/2021] [Indexed: 12/11/2022] Open
Abstract
Polymeric biomaterials exhibit excellent physicochemical characteristics as a scaffold for cell and tissue engineering applications. Chemical modification of the polymers has been the primary mode of functionalization to enhance biocompatibility and regulate cellular behaviors such as cell adhesion, proliferation, differentiation, and maturation. Due to the complexity of the in vivo cellular microenvironments, however, chemical functionalization alone is usually insufficient to develop functionally mature cells/tissues. Therefore, the multifunctional polymeric scaffolds that enable electrical, mechanical, and/or magnetic stimulation to the cells, have gained research interest in the past decade. Such multifunctional scaffolds are often combined with exogenous stimuli to further enhance the tissue and cell behaviors by dynamically controlling the microenvironments of the cells. Significantly improved cell proliferation and differentiation, as well as tissue functionalities, are frequently observed by applying extrinsic physical stimuli on functional polymeric scaffold systems. In this regard, the present paper discusses the current state-of-the-art functionalized polymeric scaffolds, with an emphasis on electrospun fibers, that modulate the physical cell niche to direct cellular behaviors and subsequent functional tissue development. We will also highlight the incorporation of the extrinsic stimuli to augment or activate the functionalized polymeric scaffold system to dynamically stimulate the cells.
Collapse
Affiliation(s)
| | | | | | | | - Jin Nam
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; (Y.T.); (A.B.); (R.G.); (L.J.)
| |
Collapse
|
48
|
Kush P, Kumar P, Singh R, Kaushik A. Aspects of high-performance and bio-acceptable magnetic nanoparticles for biomedical application. Asian J Pharm Sci 2021; 16:704-737. [PMID: 35027950 PMCID: PMC8737424 DOI: 10.1016/j.ajps.2021.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/01/2021] [Accepted: 05/22/2021] [Indexed: 12/11/2022] Open
Abstract
This review covers extensively the synthesis & surface modification, characterization, and application of magnetic nanoparticles. For biomedical applications, consideration should be given to factors such as design strategies, the synthesis process, coating, and surface passivation. The synthesis method regulates post-synthetic change and specific applications in vitro and in vivo imaging/diagnosis and pharmacotherapy/administration. Special insights have been provided on biodistribution, pharmacokinetics, and toxicity in a living system, which is imperative for their wider application in biology. These nanoparticles can be decorated with multiple contrast agents and thus can also be used as a probe for multi-mode imaging or double/triple imaging, for example, MRI-CT, MRI-PET. Similarly loading with different drug molecules/dye/fluorescent molecules and integration with other carriers have found application not only in locating these particles in vivo but simultaneously target drug delivery/hyperthermia inside the body. Studies are underway to collect the potential of these magnetically driven nanoparticles in various scientific fields such as particle interaction, heat conduction, imaging, and magnetism. Surely, this comprehensive data will help in the further development of advanced techniques for theranostics based on high-performance magnetic nanoparticles and will lead this research area in a new sustainable direction.
Collapse
Affiliation(s)
- Preeti Kush
- School of Pharmacy, Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University Gangoh, Saharanpur, Uttar Pradesh 247341, India
| | - Parveen Kumar
- Nanotechnology Division (H-1), CSIR-Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Ranjit Singh
- School of Pharmacy, Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University Gangoh, Saharanpur, Uttar Pradesh 247341, India
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health System Engineering, Department of Natural Sciences, Florida Polytechnic University, Lakeland, FL 33805-8531, United States
| |
Collapse
|
49
|
Friedrich RP, Cicha I, Alexiou C. Iron Oxide Nanoparticles in Regenerative Medicine and Tissue Engineering. NANOMATERIALS 2021; 11:nano11092337. [PMID: 34578651 PMCID: PMC8466586 DOI: 10.3390/nano11092337] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
In recent years, many promising nanotechnological approaches to biomedical research have been developed in order to increase implementation of regenerative medicine and tissue engineering in clinical practice. In the meantime, the use of nanomaterials for the regeneration of diseased or injured tissues is considered advantageous in most areas of medicine. In particular, for the treatment of cardiovascular, osteochondral and neurological defects, but also for the recovery of functions of other organs such as kidney, liver, pancreas, bladder, urethra and for wound healing, nanomaterials are increasingly being developed that serve as scaffolds, mimic the extracellular matrix and promote adhesion or differentiation of cells. This review focuses on the latest developments in regenerative medicine, in which iron oxide nanoparticles (IONPs) play a crucial role for tissue engineering and cell therapy. IONPs are not only enabling the use of non-invasive observation methods to monitor the therapy, but can also accelerate and enhance regeneration, either thanks to their inherent magnetic properties or by functionalization with bioactive or therapeutic compounds, such as drugs, enzymes and growth factors. In addition, the presence of magnetic fields can direct IONP-labeled cells specifically to the site of action or induce cell differentiation into a specific cell type through mechanotransduction.
Collapse
|
50
|
Kopyl S, Surmenev R, Surmeneva M, Fetisov Y, Kholkin A. Magnetoelectric effect: principles and applications in biology and medicine- a review. Mater Today Bio 2021; 12:100149. [PMID: 34746734 PMCID: PMC8554634 DOI: 10.1016/j.mtbio.2021.100149] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/26/2022] Open
Abstract
Magnetoelectric (ME) effect experimentally discovered about 60 years ago remains one of the promising research fields with the main applications in microelectronics and sensors. However, its applications to biology and medicine are still in their infancy. For the diagnosis and treatment of diseases at the intracellular level, it is necessary to develop a maximally non-invasive way of local stimulation of individual neurons, navigation, and distribution of biomolecules in damaged cells with relatively high efficiency and adequate spatial and temporal resolution. Recently developed ME materials (composites), which combine elastically coupled piezoelectric (PE) and magnetostrictive (MS) phases, have been shown to yield very strong ME effects even at room temperature. This makes them a promising toolbox for solving many problems of modern medicine. The main ME materials, processing technologies, as well as most prospective biomedical applications will be overviewed, and modern trends in using ME materials for future therapies, wireless power transfer, and optogenetics will be considered.
Collapse
Affiliation(s)
- S. Kopyl
- Department of Physics & CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - R. Surmenev
- Physical Materials Science and Composite Materials Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - M. Surmeneva
- Physical Materials Science and Composite Materials Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Y. Fetisov
- Research & Education Centre ‘Magnetoelectric Materials and Devices’, MIREA – Russian Technological University, Moscow, Russia
| | - A. Kholkin
- Department of Physics & CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, Russia
- School of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg, Russia
| |
Collapse
|