101
|
Bagne L, Oliveira MA, Pereira AT, Caetano GF, Oliveira CA, Aro AA, Chiarotto GB, Santos GMT, Mendonça FAS, Santamaria-Jr M. Electrical therapies act on the Ca 2+ /CaM signaling pathway to enhance bone regeneration with bioactive glass [S53P4] and allogeneic grafts. J Biomed Mater Res B Appl Biomater 2021; 109:2104-2116. [PMID: 34008329 DOI: 10.1002/jbm.b.34858] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/12/2021] [Accepted: 04/24/2021] [Indexed: 12/26/2022]
Abstract
This study aimed to investigate the application of low-intensity electrostimulation (ES) and electromagnetic stimulation (EM) associated with bioactive glass (BG) or allogeneic grafts (BB) in bone regeneration. A cell viability test on osteoblasts (UMR-106) was performed in the presence of BB and BG grafts associated with ES (10 μA/5 min) and EM (500 Hz/2 min). Critical defects (25 mm2 ) in calvaria were generated in male Wistar rats, and bone regeneration was evaluated on the 30th, 60th, and 120th days after surgery. Cell proliferation increased with the application of ES in both grafts and after EM with BG. Bone remodeling was more effective using the allogeneic graft in both therapies, with increased angiogenesis, osteoblast proliferation, and OPN expression in the BB + EM group. A higher number of osteoblasts and osteoclasts, and an increase in bone sialoprotein, Runx-2, and Opn gene expression were found in the BB + ES group. The BG graft associated with EM therapy had an increased proliferation of osteoblasts and increased expression of Runx-2 and Opn. Groups that had BG and ES therapy had increased numbers of osteoblasts, osteoclasts, and increased OPN expression. The expression of voltage-gated calcium channels increased in groups with ES, while calmodulin expression increased in therapies without grafting. ES and EM therapies favored the repair of bone defects upon grafting by improving angiogenesis, osteogenic gene expression, and tissue reorganization. Despite activating different pathways, both therapies increased the intracellular concentrations of calmodulin, leading to cell proliferation and bone regeneration.
Collapse
Affiliation(s)
- Leonardo Bagne
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Maraiara A Oliveira
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Amanda T Pereira
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Guilherme F Caetano
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Camila A Oliveira
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Andréa A Aro
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Gabriela B Chiarotto
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Glaucia M T Santos
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Fernanda A S Mendonça
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| | - Milton Santamaria-Jr
- Graduate Program in Biomedical Sciences, University Center of Hermínio Ometto Foundation - FHO, Araras, Brazil
| |
Collapse
|
102
|
Jiang S, Wang M, He J. A review of biomimetic scaffolds for bone regeneration: Toward a cell-free strategy. Bioeng Transl Med 2021; 6:e10206. [PMID: 34027093 PMCID: PMC8126827 DOI: 10.1002/btm2.10206] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
In clinical terms, bone grafting currently involves the application of autogenous, allogeneic, or xenogeneic bone grafts, as well as natural or artificially synthesized materials, such as polymers, bioceramics, and other composites. Many of these are associated with limitations. The ideal scaffold for bone tissue engineering should provide mechanical support while promoting osteogenesis, osteoconduction, and even osteoinduction. There are various structural complications and engineering difficulties to be considered. Here, we describe the biomimetic possibilities of the modification of natural or synthetic materials through physical and chemical design to facilitate bone tissue repair. This review summarizes recent progresses in the strategies for constructing biomimetic scaffolds, including ion-functionalized scaffolds, decellularized extracellular matrix scaffolds, and micro- and nano-scale biomimetic scaffold structures, as well as reactive scaffolds induced by physical factors, and other acellular scaffolds. The fabrication techniques for these scaffolds, along with current strategies in clinical bone repair, are described. The developments in each category are discussed in terms of the connection between the scaffold materials and tissue repair, as well as the interactions with endogenous cells. As the advances in bone tissue engineering move toward application in the clinical setting, the demonstration of the therapeutic efficacy of these novel scaffold designs is critical.
Collapse
Affiliation(s)
- Sijing Jiang
- Department of Plastic SurgeryFirst Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiChina
| | - Mohan Wang
- Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceHefeiChina
| | - Jiacai He
- Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceHefeiChina
| |
Collapse
|
103
|
Xue G, Zhang Y, Xie T, Zhang Z, Liu Q, Li X, Gou X. Cell Adhesion-Mediated Piezoelectric Self-Stimulation on Polydopamine-Modified Poly(vinylidene fluoride) Membranes. ACS APPLIED MATERIALS & INTERFACES 2021; 13:17361-17371. [PMID: 33823586 DOI: 10.1021/acsami.1c02457] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cell adhesion-mediated piezoelectric stimulation provides a noninvasive method for in situ electrical regulation of cell behavior, offering new opportunities for the design of smart materials for tissue engineering and bioelectronic medicines. In particular, the surface potential is mainly dominated by the inherent piezoelectricity of the biomaterial and the dynamic adhesion state of cells. The development of an efficient and optimized material interface would have important implications in cell regulation. Herein, we modified the surface of poled poly(vinylidene fluoride) (PVDF) membranes through polymerization of dopamine and investigated their influence on cell adhesion and electromechanical self-stimulation. Our results demonstrated that mesenchymal stem cells seeded on the poled PVDF membrane exhibited stronger cell spreading and adhesion. Meanwhile, the surface modification through polydopamine significantly improved the hydrophilicity of the samples and contributed to the formation of cell actin bundles and maturation of focal adhesions, which further positively modulated cell piezoelectric self-stimulation and induced intracellular calcium transients. Combining with theoretical simulations, we found that the self-stimulation was enhanced mainly due to the increase of the adhesion site and adhesion force magnitude. These findings provide new insights for probing the cell regulation mechanism on piezoelectric substrates, offering more opportunities for the rational design of piezoelectric biomaterial interfaces for biomedical engineering.
Collapse
Affiliation(s)
- Guilan Xue
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Yimeng Zhang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Tianpeng Xie
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Zhanlin Zhang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Qingjie Liu
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Xiaohong Li
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Xue Gou
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| |
Collapse
|
104
|
Swain S, Bowen C, Rautray T. Dual response of osteoblast activity and antibacterial properties of polarized strontium substituted hydroxyapatite-Barium strontium titanate composites with controlled strontium substitution. J Biomed Mater Res A 2021; 109:2027-2035. [PMID: 33825314 DOI: 10.1002/jbm.a.37195] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 11/09/2022]
Abstract
To mimic the electrical properties of natural bone, controlled strontium substitution of both hydroxyapatite and ferroelectric barium titanate were achieved by mixing in the ratio 30:70 by weight. The composites were characterized by X-ray diffraction, Fourier transform infrared spectroscopy and scanning electron microscopy to investigate the phase composition and microstructure of the composites. Unpolarized and polarized strontium hydroxyapatite (SrHA)-barium strontium titanate (BST) composites with controlled degree of Sr substitution were examined, including 5SrHA-5BST (5% Sr substitution in both components) and 10SrHA-10BST composites. The 10SrHA-10BST composite showed a higher osteoblast activity, as observed from the cell viability studies performed using CCK-8 assay. The polarized composites showed promise against Staphylococcus aureus bacteria by minimizing the adhesion and growth of bacteria, as compared with their unpolarized counterparts. The polarized 10SrHA-10BST was found to be superior than all other composites. As a result, the approach of polarization of SrHA-BST composites has been found to be an effective bone substitute material in controlled enhancement of osteoblast growth with simultaneous reduction of bacterial infection.
Collapse
Affiliation(s)
- Subhasmita Swain
- Biomaterials and Tissue Regeneration Laboratory, Centre of Excellence in TM Sciences, Siksha 'O' Anusandhan (Deemed to be University), Khandagiri Square, Bhubaneswar, India, 751030, India
| | - Chris Bowen
- Dept of Mechanical Engineering, University of Bath, Bath, UK
| | - Tapash Rautray
- Biomaterials and Tissue Regeneration Laboratory, Centre of Excellence in TM Sciences, Siksha 'O' Anusandhan (Deemed to be University), Khandagiri Square, Bhubaneswar, India, 751030, India
| |
Collapse
|
105
|
Yan L, Zhou T, Han L, Zhu M, Cheng Z, Li D, Ren F, Wang K, Lu X. Conductive Cellulose Bio‐Nanosheets Assembled Biostable Hydrogel for Reliable Bioelectronics. ADVANCED FUNCTIONAL MATERIALS 2021; 31. [DOI: 10.1002/adfm.202010465] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 03/01/2025]
Abstract
AbstractBiostable electronic materials that can maintain their super mechanical and conductive properties, even when exposed to biofluids, are the fundamental basis for designing reliable bioelectronic devices. Herein, cellulose‐derived conductive 2D bio‐nanosheets as electronic base materials are developed and assembled into a conductive hydrogel with ultra‐high biostability, capable of surviving in harsh physiological environments. The bio‐nanosheets are synthesized by guiding the in situ regeneration of cellulose crystal into a 2D planar structure using the polydopamine‐reduced‐graphene oxide as supporting templates. The nanosheet‐assembled hydrogel exhibits stable electrical and mechanical performances after undergoing aqueous immersion and in vivo implantation. Thus, the hydrogel‐based bioelectronic devices are able to conformally integrate with the human body and stably record electrophysiological signals. Owing to its tissue affinity, the hydrogel further serves as an “E‐skin,” which employs electrotherapy to aid in the faster healing of chronic wounds in diabetic mice through transcutaneous electrical stimulation. The nanosheet‐assembled biostable, conductive, flexible, and cell/tissue affinitive hydrogel lays a foundation for designing electronically and mechanically reliable bioelectronic devices.
Collapse
Affiliation(s)
- Liwei Yan
- Key Lab of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu Sichuan 610031 China
| | - Ting Zhou
- Key Lab of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu Sichuan 610031 China
| | - Lu Han
- School of Medicine and Pharmaceutics Ocean University of China Qingdao Shandong 266003 China
| | - Mingyu Zhu
- Department of Materials Science and Engineering Southern University of Science and Technology Shenzhen Guangdong 518055 China
| | - Zhuo Cheng
- Department of Materials Science and Engineering Southern University of Science and Technology Shenzhen Guangdong 518055 China
| | - Da Li
- Key Lab of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu Sichuan 610031 China
| | - Fuzeng Ren
- Department of Materials Science and Engineering Southern University of Science and Technology Shenzhen Guangdong 518055 China
| | - Kefeng Wang
- National Engineering Research Center for Biomaterials Genome Research Center for Biomaterials Sichuan University Chengdu Sichuan 610064 China
| | - Xiong Lu
- Key Lab of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu Sichuan 610031 China
| |
Collapse
|
106
|
James EC, Tomaskovic-Crook E, Crook JM. Bioengineering Clinically Relevant Cardiomyocytes and Cardiac Tissues from Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22063005. [PMID: 33809429 PMCID: PMC8001925 DOI: 10.3390/ijms22063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The regenerative capacity of cardiomyocytes is insufficient to functionally recover damaged tissue, and as such, ischaemic heart disease forms the largest proportion of cardiovascular associated deaths. Human-induced pluripotent stem cells (hiPSCs) have enormous potential for developing patient specific cardiomyocytes for modelling heart disease, patient-based cardiac toxicity testing and potentially replacement therapy. However, traditional protocols for hiPSC-derived cardiomyocytes yield mixed populations of atrial, ventricular and nodal-like cells with immature cardiac properties. New insights gleaned from embryonic heart development have progressed the precise production of subtype-specific hiPSC-derived cardiomyocytes; however, their physiological immaturity severely limits their utility as model systems and their use for drug screening and cell therapy. The long-entrenched challenges in this field are being addressed by innovative bioengingeering technologies that incorporate biophysical, biochemical and more recently biomimetic electrical cues, with the latter having the potential to be used to both direct hiPSC differentiation and augment maturation and the function of derived cardiomyocytes and cardiac tissues by mimicking endogenous electric fields.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy 3065, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| |
Collapse
|
107
|
Casella A, Panitch A, Leach JK. Endogenous Electric Signaling as a Blueprint for Conductive Materials in Tissue Engineering. Bioelectricity 2021; 3:27-41. [PMID: 34476376 PMCID: PMC8370482 DOI: 10.1089/bioe.2020.0027] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bioelectricity plays an important role in cell behavior and tissue modulation, but is understudied in tissue engineering research. Endogenous electrical signaling arises from the transmembrane potential inherent to all cells and contributes to many cell behaviors, including migration, adhesion, proliferation, and differentiation. Electrical signals are also involved in tissue development and repair. Synthetic and natural conductive materials are under investigation for leveraging endogenous electrical signaling cues in tissue engineering applications due to their ability to direct cell differentiation, aid in maturing electroactive cell types, and promote tissue functionality. In this review, we provide a brief overview of bioelectricity and its impact on cell behavior, report recent literature using conductive materials for tissue engineering, and discuss opportunities within the field to improve experimental design when using conductive substrates.
Collapse
Affiliation(s)
- Alena Casella
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
- Department of Surgery and UC Davis Health, Sacramento, California, USA
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| |
Collapse
|
108
|
Gómez IJ, Vázquez Sulleiro M, Mantione D, Alegret N. Carbon Nanomaterials Embedded in Conductive Polymers: A State of the Art. Polymers (Basel) 2021; 13:745. [PMID: 33673680 PMCID: PMC7957790 DOI: 10.3390/polym13050745] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Carbon nanomaterials are at the forefront of the newest technologies of the third millennium, and together with conductive polymers, represent a vast area of indispensable knowledge for developing the devices of tomorrow. This review focusses on the most recent advances in the field of conductive nanotechnology, which combines the properties of carbon nanomaterials with conjugated polymers. Hybrid materials resulting from the embedding of carbon nanotubes, carbon dots and graphene derivatives are taken into consideration and fully explored, with discussion of the most recent literature. An introduction into the three most widely used conductive polymers and a final section about the most recent biological results obtained using carbon nanotube hybrids will complete this overview of these innovative and beyond belief materials.
Collapse
Affiliation(s)
- I. Jénnifer Gómez
- Department of Condensed Matter Physics, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic;
| | | | - Daniele Mantione
- Laboratoire de Chimie des Polymères Organiques (LCPO-UMR 5629), Université de Bordeaux, Bordeaux INP, CNRS F, 33607 Pessac, France
| | - Nuria Alegret
- POLYMAT and Departamento de Química Aplicada, University of the Basque Country, UPV/EHU, 20018 Donostia-San Sebastián, Spain
| |
Collapse
|
109
|
Karbowniczek JE, Kaniuk Ł, Berniak K, Gruszczyński A, Stachewicz U. Enhanced Cells Anchoring to Electrospun Hybrid Scaffolds With PHBV and HA Particles for Bone Tissue Regeneration. Front Bioeng Biotechnol 2021; 9:632029. [PMID: 33681169 PMCID: PMC7928304 DOI: 10.3389/fbioe.2021.632029] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/21/2021] [Indexed: 12/18/2022] Open
Abstract
Hybrid materials combining organic and inorganic compounds used as scaffolds are highly beneficial in bone regeneration. In this study, we successfully produced by blend electrospinning poly(3-hydroxybutyric acid-co-3-hydrovaleric acid) (PHBV) scaffolds enriched with hydroxyapatite (HA) particles to biomimic bone tissue for improved and faster regeneration processes. The morphology, fiber diameters, and composition of the scaffolds were investigated by scanning electron microscopy (SEM) techniques followed by focused ion beam (FIB) sectioning to verify HA particles integration with PHBV fibers. In vitro cell culture was performed for 7 days and followed with the cell proliferation test (CellTiter-Blue® Assay). Additionally, cell integration with the scaffold was visualized by confocal and SEM imaging. We developed a simple way of obtaining hybrid scaffolds by electrospinning PHBV solution with HA particles without any post-processing. The PHBV + HA scaffold enhanced cell proliferation and filopodia formation responsible for cell anchoring within the created 3D environment. The obtained results show the great potential in the development of hybrid scaffolds stimulating bone tissue regeneration.
Collapse
Affiliation(s)
- Joanna E Karbowniczek
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, Kraków, Poland
| | - Łukasz Kaniuk
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, Kraków, Poland
| | - Krzysztof Berniak
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, Kraków, Poland
| | - Adam Gruszczyński
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, Kraków, Poland
| | - Urszula Stachewicz
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, Kraków, Poland
| |
Collapse
|
110
|
Srirussamee K, Xue R, Mobini S, Cassidy NJ, Cartmell SH. Changes in the extracellular microenvironment and osteogenic responses of mesenchymal stem/stromal cells induced by in vitro direct electrical stimulation. J Tissue Eng 2021; 12:2041731420974147. [PMID: 33643602 PMCID: PMC7894594 DOI: 10.1177/2041731420974147] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022] Open
Abstract
Electrical stimulation (ES) has potential to be an effective tool for bone injury treatment in clinics. However, the therapeutic mechanism associated with ES is still being discussed. This study aims to investigate the initial mechanism of action by characterising the physical and chemical changes in the extracellular environment during ES and correlate them with the responses of mesenchymal stem/stromal cells (MSCs). Computational modelling was used to estimate the electrical potentials relative to the cathode and the current density across the cell monolayer. We showed expression of phosphorylated ERK1/2, c-FOS, c-JUN, and SPP1 mRNAs, as well as the increased metabolic activities of MSCs at different time points. Moreover, the average of 2.5 μM of H2O2 and 34 μg/L of dissolved Pt were measured from the electrically stimulated media (ES media), which also corresponded with the increases in SPP1 mRNA expression and cell metabolic activities. The addition of sodium pyruvate to the ES media as an antioxidant did not alter the SPP1 mRNA expression, but eliminated an increase in cell metabolic activities induced by ES media treatment. These findings suggest that H2O2 was influencing cell metabolic activity, whereas SPP1 mRNA expression was regulated by other faradic by-products. This study reveals how different electrical stimulation regime alters cellular regenerative responses and the roles of faradic by-products, that might be used as a physical tool to guide and control cell behaviour.
Collapse
Affiliation(s)
- Kasama Srirussamee
- Department of Materials, The University of Manchester, Manchester, UK.,Department of Biomedical Engineering, Faculty of Engineering, King Mongkut's Institute of Technology Ladkrabang (KMITL), Bangkok, Thailand
| | - Ruikang Xue
- Department of Materials, The University of Manchester, Manchester, UK
| | - Sahba Mobini
- Department of Materials, The University of Manchester, Manchester, UK.,Instituto de Micro y Nanotecnología IMN-CNM, The Spanish National Research Council (CSIC), Madrid, Comunidad de Madrid, Spain.,Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Nigel J Cassidy
- Department of Civil Engineering, University of Birmingham, Birmingham, UK
| | - Sarah H Cartmell
- Department of Materials, The University of Manchester, Manchester, UK
| |
Collapse
|
111
|
Sabri E, Brosseau C. Proximity-induced electrodeformation and membrane capacitance coupling between cells. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 50:713-720. [PMID: 33538871 DOI: 10.1007/s00249-021-01504-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 01/19/2021] [Indexed: 12/01/2022]
Abstract
Membrane capacitance and transmembrane potential are sensitive to the proximity of neighboring biological cells which eventually induces anisotropic perturbation of the local electric field distribution in a cell assembly and/or a tissue. The development of robust and reliable multiphysics approaches is essential to solve the challenge of analyzing proximity-induced capacitance coupling (CC) between cells. In this study, we ask to what extent this CC is a minor perturbation on the individual cells or whether it can fundamentally affect bio-electromechanical cues. A key component of our continuum electromechanical analysis is the consideration of elastic models of cells under steady state electric field excitation to characterize electrodeformation (ED). Analyzing the difference between the ED force for a pair of cells and its counterpart for a single reference cell allows us to determine a separation distance-orientation angle diagram providing evidence of a separation distance beyond which the electrostatic interactions between a pair of biological cells become inconsequential for the ED. An attenuation-amplification transition of ED force in this diagram suggests that anisotropy induced by the orientation angle of the cell pair relative to the applied electric field direction has a significant influence on ED and CC. We furthermore observe that the shape of this diagram changes when extracellular conductivity is varied. The results obtained are then contrasted with the corresponding diagrams of similar cell configurations under an oscillating electric field excitation below and above the α-dispersion frequency. This investigation may provide new opportunities for further assessment of electromechanical properties of engineered tissues.
Collapse
Affiliation(s)
- E Sabri
- Univ Brest, CNRS, Lab-STICC, 6 avenue Le Gorgeu, 29238, Brest Cedex 3, France
| | - C Brosseau
- Univ Brest, CNRS, Lab-STICC, 6 avenue Le Gorgeu, 29238, Brest Cedex 3, France.
| |
Collapse
|
112
|
Saghati S, Nasrabadi HT, Khoshfetrat AB, Moharamzadeh K, Hassani A, Mohammadi SM, Rahbarghazi R, Fathi Karkan S. Tissue Engineering Strategies to Increase Osteochondral Regeneration of Stem Cells; a Close Look at Different Modalities. Stem Cell Rev Rep 2021; 17:1294-1311. [PMID: 33547591 DOI: 10.1007/s12015-021-10130-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
The homeostasis of osteochondral tissue is tightly controlled by articular cartilage chondrocytes and underlying subchondral bone osteoblasts via different internal and external clues. As a correlate, the osteochondral region is frequently exposed to physical forces and mechanical pressure. On this basis, distinct sets of substrates and physicochemical properties of the surrounding matrix affect the regeneration capacity of chondrocytes and osteoblasts. Stem cells are touted as an alternative cell source for the alleviation of osteochondral diseases. These cells appropriately respond to the physicochemical properties of different biomaterials. This review aimed to address some of the essential factors which participate in the chondrogenic and osteogenic capacity of stem cells. Elements consisted of biomechanical forces, electrical fields, and biochemical and physical properties of the extracellular matrix are the major determinant of stem cell differentiation capacity. It is suggested that an additional certain mechanism related to signal-transduction pathways could also mediate the chondro-osteogenic differentiation of stem cells. The discovery of these clues can enable us to modulate the regeneration capacity of stem cells in osteochondral injuries and lead to the improvement of more operative approaches using tissue engineering modalities.
Collapse
Affiliation(s)
- Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Baradar Khoshfetrat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine (HBMCDM), Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Ayla Hassani
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Seyedeh Momeneh Mohammadi
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sonia Fathi Karkan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
113
|
Moysidou CM, Barberio C, Owens RM. Advances in Engineering Human Tissue Models. Front Bioeng Biotechnol 2021; 8:620962. [PMID: 33585419 PMCID: PMC7877542 DOI: 10.3389/fbioe.2020.620962] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Research in cell biology greatly relies on cell-based in vitro assays and models that facilitate the investigation and understanding of specific biological events and processes under different conditions. The quality of such experimental models and particularly the level at which they represent cell behavior in the native tissue, is of critical importance for our understanding of cell interactions within tissues and organs. Conventionally, in vitro models are based on experimental manipulation of mammalian cells, grown as monolayers on flat, two-dimensional (2D) substrates. Despite the amazing progress and discoveries achieved with flat biology models, our ability to translate biological insights has been limited, since the 2D environment does not reflect the physiological behavior of cells in real tissues. Advances in 3D cell biology and engineering have led to the development of a new generation of cell culture formats that can better recapitulate the in vivo microenvironment, allowing us to examine cells and their interactions in a more biomimetic context. Modern biomedical research has at its disposal novel technological approaches that promote development of more sophisticated and robust tissue engineering in vitro models, including scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips. Even though such systems are necessarily simplified to capture a particular range of physiology, their ability to model specific processes of human biology is greatly valued for their potential to close the gap between conventional animal studies and human (patho-) physiology. Here, we review recent advances in 3D biomimetic cultures, focusing on the technological bricks available to develop more physiologically relevant in vitro models of human tissues. By highlighting applications and examples of several physiological and disease models, we identify the limitations and challenges which the field needs to address in order to more effectively incorporate synthetic biomimetic culture platforms into biomedical research.
Collapse
Affiliation(s)
| | | | - Róisín Meabh Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
114
|
Choi KH, Yoon JW, Kim M, Lee HJ, Jeong J, Ryu M, Jo C, Lee CK. Muscle stem cell isolation and in vitro culture for meat production: A methodological review. Compr Rev Food Sci Food Saf 2021; 20:429-457. [PMID: 33443788 DOI: 10.1111/1541-4337.12661] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Cultured muscle tissue-based protein products, also known as cultured meat, are produced through in vitro myogenesis involving muscle stem cell culture and differentiation, and mature muscle cell processing for flavor and texture. This review focuses on the in vitro myogenesis for cultured meat production. The muscle stem cell-based in vitro muscle tissue production consists of a sequential process: (1) muscle sampling for stem cell collection, (2) muscle tissue dissociation and muscle stem cell isolation, (3) primary cell culture, (4) upscaled cell culture, (5) muscle differentiation and maturation, and (6) muscle tissue harvest. Although muscle stem cell research is a well-established field, the majority of these steps remain to be underoptimized to enable the in vitro creation of edible muscle-derived meat products. The profound understanding of the process would help not only cultured meat production but also business sectors that have been seeking new biomaterials for the food industry. In this review, we discuss comprehensively and in detail each step of cutting-edge methods for cultured meat production. This would be meaningful for both academia and industry to prepare for the new era of cellular agriculture.
Collapse
Affiliation(s)
- Kwang-Hwan Choi
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Ji Won Yoon
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Minsu Kim
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Minkyung Ryu
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
| |
Collapse
|
115
|
Guette-Marquet S, Roques C, Bergel A. Theoretical analysis of the electrochemical systems used for the application of direct current/voltage stimuli on cell cultures. Bioelectrochemistry 2021; 139:107737. [PMID: 33494030 DOI: 10.1016/j.bioelechem.2020.107737] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 12/31/2022]
Abstract
Endogenous electric fields drive many essential functions relating to cell proliferation, motion, differentiation and tissue development. They are usually mimicked in vitro by using electrochemical systems to apply direct current or voltage stimuli to cell cultures. The many studies devoted to this topic have given rise to a wide variety of experimental systems, whose results are often difficult to compare. Here, these systems are analysed from an electrochemical standpoint to help harmonize protocols and facilitate optimal understanding of the data produced. The theoretical analysis of single-electrode systems shows the necessity of measuring the Nernst potential of the electrode and of discussing the results on this basis rather than using the value of the potential gradient. The paper then emphasizes the great complexity that can arise when high cell voltage is applied to a single electrode, because of the possible occurrence of anode and cathode sites. An analysis of two-electrode systems leads to the advice to change experimental practices by applying current instead of voltage. It also suggests that the values of electric fields reported so far may have been considerably overestimated in macro-sized devices. It would consequently be wise to revisit this area by testing considerably lower electric field values.
Collapse
Affiliation(s)
- Simon Guette-Marquet
- Laboratoire de Génie Chimique, Université de Toulouse, CNRS, INPT, UPS, Toulouse, France
| | - Christine Roques
- Laboratoire de Génie Chimique, Université de Toulouse, CNRS, INPT, UPS, Toulouse, France
| | - Alain Bergel
- Laboratoire de Génie Chimique, Université de Toulouse, CNRS, INPT, UPS, Toulouse, France.
| |
Collapse
|
116
|
Cui S, Mao J, Rouabhia M, Elkoun S, Zhang Z. A biocompatible polypyrrole membrane for biomedical applications. RSC Adv 2021; 11:16996-17006. [PMID: 35479716 PMCID: PMC9031619 DOI: 10.1039/d1ra01338f] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/26/2021] [Indexed: 01/22/2023] Open
Abstract
Polypyrrole (PPy) is the most widely investigated electrically conductive biomaterial. However, because of its intrinsic rigidity, PPy has only been used either in the form of a composite or a thin coating. This work presents a pure and soft PPy membrane that is synergically reinforced with the electrospun polyurethane (PU) and poly-l-lactic acid (PLLA) fibers. This particular reinforcement not only renders the originally rather fragile PPy membrane easy to manipulate, it also prevents the membrane from deformation in an aqueous environment. Peel and mechanical tests confirmed the strong adhesion of the fibers and the significantly increased tensile strength of the reinforced membrane. Surface electrical conductivity and long-term electrical stability were tested, showing that these properties were not affected by the reinforcement. Surface morphology and chemistry were analyzed with scanning electron spectroscopy (SEM), X-ray photoelectron spectroscopy (XPS), and Fourier transform infrared spectroscopy (FTIR). Material thermal stability was investigated with thermogravimetric analysis (TGA). Finally, the adhesion and proliferation of human skin keratinocytes on the membrane were assessed by Hoechst staining and the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. In conclusion, this membrane proves to be the first PPy-based soft conductive biomaterial that can be practically used. Its electrical conductivity and cytocompatibility promise a wide range of biomedical applications. A reinforced soft polypyrrole membrane.![]()
Collapse
Affiliation(s)
- Shujun Cui
- Research Group on Oral Ecology
- Faculty of Dentistry
- Université Laval
- Québec (QC)
- Canada
| | - Jifu Mao
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles
- Donghua University
- Shanghai
- China
| | - Mahmoud Rouabhia
- Research Group on Oral Ecology
- Faculty of Dentistry
- Université Laval
- Québec (QC)
- Canada
| | - Saïd Elkoun
- Department of Mechanical Engineering
- Université de Sherbrooke
- Sherbrooke (QC)
- Canada
| | - Ze Zhang
- Department of Surgery
- Faculty of Medicine
- Université Laval
- Québec (QC)
- Canada
| |
Collapse
|
117
|
Krueger S, Riess A, Jonitz-Heincke A, Weizel A, Seyfarth A, Seitz H, Bader R. Establishment of a New Device for Electrical Stimulation of Non-Degenerative Cartilage Cells In Vitro. Int J Mol Sci 2021; 22:ijms22010394. [PMID: 33401406 PMCID: PMC7794805 DOI: 10.3390/ijms22010394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
In cell-based therapies for cartilage lesions, the main problem is still the formation of fibrous cartilage, caused by underlying de-differentiation processes ex vivo. Biophysical stimulation is a promising approach to optimize cell-based procedures and to adapt them more closely to physiological conditions. The occurrence of mechano-electrical transduction phenomena within cartilage tissue is physiological and based on streaming and diffusion potentials. The application of exogenous electric fields can be used to mimic endogenous fields and, thus, support the differentiation of chondrocytes in vitro. For this purpose, we have developed a new device for electrical stimulation of chondrocytes, which operates on the basis of capacitive coupling of alternating electric fields. The reusable and sterilizable stimulation device allows the simultaneous use of 12 cavities with independently applicable fields using only one main supply. The first parameter settings for the stimulation of human non-degenerative chondrocytes, seeded on collagen type I elastin-based scaffolds, were derived from numerical electric field simulations. Our first results suggest that applied alternating electric fields induce chondrogenic re-differentiation at the gene and especially at the protein level of human de-differentiated chondrocytes in a frequency-dependent manner. In future studies, further parameter optimizations will be performed to improve the differentiation capacity of human cartilage cells.
Collapse
Affiliation(s)
- Simone Krueger
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
- Correspondence: (S.K.); (A.R.)
| | - Alexander Riess
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
- Correspondence: (S.K.); (A.R.)
| | - Anika Jonitz-Heincke
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
| | - Alina Weizel
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
| | - Anika Seyfarth
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
| | - Hermann Seitz
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
| | - Rainer Bader
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
| |
Collapse
|
118
|
Wang W, Hou Y, Martinez D, Kurniawan D, Chiang WH, Bartolo P. Carbon Nanomaterials for Electro-Active Structures: A Review. Polymers (Basel) 2020; 12:E2946. [PMID: 33317211 PMCID: PMC7764097 DOI: 10.3390/polym12122946] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 11/18/2022] Open
Abstract
The use of electrically conductive materials to impart electrical properties to substrates for cell attachment proliferation and differentiation represents an important strategy in the field of tissue engineering. This paper discusses the concept of electro-active structures and their roles in tissue engineering, accelerating cell proliferation and differentiation, consequently leading to tissue regeneration. The most relevant carbon-based materials used to produce electro-active structures are presented, and their main advantages and limitations are discussed in detail. Particular emphasis is put on the electrically conductive property, material synthesis and their applications on tissue engineering. Different technologies, allowing the fabrication of two-dimensional and three-dimensional structures in a controlled way, are also presented. Finally, challenges for future research are highlighted. This review shows that electrical stimulation plays an important role in modulating the growth of different types of cells. As highlighted, carbon nanomaterials, especially graphene and carbon nanotubes, have great potential for fabricating electro-active structures due to their exceptional electrical and surface properties, opening new routes for more efficient tissue engineering approaches.
Collapse
Affiliation(s)
- Weiguang Wang
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK; (Y.H.); (P.B.)
| | - Yanhao Hou
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK; (Y.H.); (P.B.)
| | - Dean Martinez
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei E2-514, Taiwan; (D.M.); (D.K.); (W.-H.C.)
| | - Darwin Kurniawan
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei E2-514, Taiwan; (D.M.); (D.K.); (W.-H.C.)
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei E2-514, Taiwan; (D.M.); (D.K.); (W.-H.C.)
| | - Paulo Bartolo
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK; (Y.H.); (P.B.)
| |
Collapse
|
119
|
Farr AC, Hogan KJ, Mikos AG. Nanomaterial Additives for Fabrication of Stimuli-Responsive Skeletal Muscle Tissue Engineering Constructs. Adv Healthc Mater 2020; 9:e2000730. [PMID: 32691983 DOI: 10.1002/adhm.202000730] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/13/2020] [Indexed: 12/12/2022]
Abstract
Volumetric muscle loss necessitates novel tissue engineering strategies for skeletal muscle repair, which have traditionally involved cells and extracellular matrix-mimicking scaffolds and have thus far been unable to successfully restore physiologically relevant function. However, the incorporation of various nanomaterial additives with unique physicochemical properties into scaffolds has recently been explored as a means of fabricating constructs that are responsive to electrical, magnetic, and photothermal stimulation. Herein, several classes of nanomaterials that are used to mediate external stimulation to tissue engineered skeletal muscle are reviewed and the impact of these stimuli-responsive biomaterials on cell growth and differentiation and in vivo muscle repair is discussed. The degradation kinetics and biocompatibilities of these nanomaterial additives are also briefly examined and their potential for incorporation into clinically translatable skeletal muscle tissue engineering strategies is considered. Overall, these nanomaterial additives have proven efficacious and incorporation in tissue engineering scaffolds has resulted in enhanced functional skeletal muscle regeneration.
Collapse
Affiliation(s)
- Amy Corbin Farr
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
- Center for Engineering Complex Tissues, USA
| | - Katie J Hogan
- Center for Engineering Complex Tissues, USA
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Antonios G Mikos
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
- Center for Engineering Complex Tissues, USA
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| |
Collapse
|
120
|
Farooqi AR, Zimmermann J, Bader R, van Rienen U. Computational study on electromechanics of electroactive hydrogels for cartilage-tissue repair. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 197:105739. [PMID: 32950923 DOI: 10.1016/j.cmpb.2020.105739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/31/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND AND OBJECTIVE The self-repair capability of articular cartilage is limited because of non-vascularization and low turnover of its extracellular matrix. Regenerating hyaline cartilage remains a significant clinical challenge as most non-surgical and surgical treatments provide only mid-term relief. Eventually, further pain and mobility loss occur for many patients in the long run due to further joint deterioration. Repair of articular cartilage tissue using electroactive scaffolds and biophysical stimuli like electrical and osmotic stimulation may have the potential to heal cartilage defects occurring due to trauma, osteoarthritis, or sport-related injuries. Therefore, the focus of the current study is to present a computational model of electroactive hydrogels for the cartilage-tissue repair as a first step towards an optimized experimental design. METHODS The multiphysics transport model that mainly includes the Poisson-Nernst-Planck equations and the mechanical equation is used to find the electrical stimulation response of the polyelectrolyte hydrogels. Based upon this, a numerical model on electromechanics of electroactive hydrogels seeded with chondrocytes is presented employing the open-source software FEniCS, which is a Python library for finite-element analysis. RESULTS We analyzed the ionic concentrations and electric potential in a hydrogel sample and the cell culture medium, the osmotic pressure created due to ionic concentration variations and the resulting hydrogel displacement. The proposed mathematical model was validated with examples from literature. CONCLUSIONS The presented model for the electrical and osmotic stimulation of a hydrogel sample can serve as a useful tool for the development and analysis of a cartilaginous scaffold employing electrical stimulation. By analyzing various parameters, we pave the way for future research on a finer scale using open-source software.
Collapse
Affiliation(s)
- Abdul Razzaq Farooqi
- Institute of General Electrical Engineering, Faculty of Computer Science and Electrical Engineering, University of Rostock, Albert Einstein Str. 2, Rostock 18059, Germany; Department of Electronic Engineering, Faculty of Engineering, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Julius Zimmermann
- Institute of General Electrical Engineering, Faculty of Computer Science and Electrical Engineering, University of Rostock, Albert Einstein Str. 2, Rostock 18059, Germany
| | - Rainer Bader
- Department of Orthopaedics, University Medical Center Rostock, Rostock 18057, Germany; Department Life, Light & Matter, University of Rostock, Rostock 18051, Germany
| | - Ursula van Rienen
- Institute of General Electrical Engineering, Faculty of Computer Science and Electrical Engineering, University of Rostock, Albert Einstein Str. 2, Rostock 18059, Germany; Department Life, Light & Matter, University of Rostock, Rostock 18051, Germany
| |
Collapse
|
121
|
Changing Expression Profiles and Inclination to Competing Endogenous RNA Networks on MAPK Signaling Pathways of Human Adipose-Derived Stem Cells in a Direct Current Electric Field. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7134719. [PMID: 33204710 PMCID: PMC7666630 DOI: 10.1155/2020/7134719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/11/2020] [Accepted: 08/20/2020] [Indexed: 11/17/2022]
Abstract
Adipose-derived stem cells (ADSCs) are an abundant cell source and provide an easy way to harvest mesenchymal stem cells, which are the focus of considerable attention in regenerative medicine. Electric fields (EF) play roles in many biological events and have been reported to promote cell proliferation, migration, and differentiation. In this study, ADSCs were treated with a direct current electric field (DCEF) of either 0 (control group) or 300 mV/mm (EF group) for six hours. RNA screening and analysis revealed that 66, 164, 26, and 1310 circRNAs, lncRNAs, miRNAs, and mRNAs, respectively, were differentially expressed in the DCEF-treated ADSCs compared with untreated ADSCs. Differentially expressed mRNAs were enriched in the MAPK signaling pathway, TNF signaling pathway, and some other pathways. ANXA1, ERRFI1, JAG1, EPHA2, PRR9, and H2AFY2 were related to the keratinocyte differentiation process. Competing endogenous RNA (ceRNA) networks were constructed on the basis of genes in the MAPK signaling pathway. Twenty-one RNAs in the above networks were randomly chosen, and their expression was validated using qRT-PCR, which showed the same expression trends as the RNA sequencing analysis. The MAPK signaling pathway is of great importance in the ADSC processes that occur in a DCEF, including keratinocyte differentiation. Several ceRNAs may participate in the regulation of MAPK signaling. This study may give new insight into the proliferation, migration, and differentiation of ADSCs, which will be valuable for tissue engineering and regenerative medicine.
Collapse
|
122
|
Yadav V, Chong N, Ellis B, Ren X, Senapati S, Chang HC, Zorlutuna P. Constant-potential environment for activating and synchronizing cardiomyocyte colonies with on-chip ion-depleting perm-selective membranes. LAB ON A CHIP 2020; 20:4273-4284. [PMID: 33090162 DOI: 10.1039/d0lc00809e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this study, an ion depleted zone created by an ion-selective membrane was used to impose a high and uniform constant extracellular potential over an entire ∼1000 cell rat cardiomyocyte (rCM) colony on-a-chip to trigger synchronized voltage-gated ion channel activities while preserving cell viability, thus extending single-cell voltage-clamp ion channel studies to an entire normalized colony. Image analysis indicated that rCM beating was strengthened and accelerated (by a factor of ∼2) within minutes of ion depletion and the duration of contraction and relaxation phases was significantly reduced. After the initial synchronization, the entire colony responds collectively to external potential changes such that beating over the entire colony can be activated or deactivated within 0.1 s. These newly observed collective dynamic responses, due to simultaneous ion channel activation/deactivation by a uniform constant-potential extracellular environment, suggest that perm-selective membrane modules on cell culture chips can facilitate studies of extracellular cardiac cell electrical communication and how ion-channel related pathologies affect cardiac cell synchronization. The future applications of this new technology can lead to better drug screening platforms for cardiotoxicity as well as platforms that can facilitate synchronized maturation of pluripotent stem cells into colonies with high electrical connectivity.
Collapse
Affiliation(s)
- Vivek Yadav
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Nicholas Chong
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Bradley Ellis
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Center for Microfluidics and Medical Diagnostics, University of Notre Dame, Notre Dame, IN 46556, USA and Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Pinar Zorlutuna
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA. and Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA and Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
123
|
Cartmell SH. Regenerative Technologies: Future Grand Challenges and Emerging Strategies. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:603580. [PMID: 35047885 PMCID: PMC8757714 DOI: 10.3389/fmedt.2020.603580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/06/2020] [Indexed: 11/13/2022] Open
|
124
|
Fu X, Cheong YH, Ahamed A, Zhou C, Robert C, Krikstolaityte V, Gordon KC, Lisak G. Diagnostics of skin features through 3D skin mapping based on electro-controlled deposition of conducting polymers onto metal-sebum modified surfaces and their possible applications in skin treatment. Anal Chim Acta 2020; 1142:84-98. [PMID: 33280707 DOI: 10.1016/j.aca.2020.10.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/02/2020] [Accepted: 10/27/2020] [Indexed: 11/26/2022]
Abstract
Analytical diagnostics of skin features was developed through application of portable and fast skin mapping based on electro-controlled deposition of conducting polymers onto metal-sebum modified surfaces. In this analytical diagnostic technique, the development of skin pattern is based on electropolymerization of conducting polymers within insulating barriers in skin stamp provided by natural sebum to monitor the 3D nature of various skin features. The recorded skin maps reach a μm-level resolution and are proved to be capable of recognition, enhancement, and reproduction of surface outlines of various skin topographies, subsequently assisting dermatological diagnosis. The technique can precisely record skin surface morphology and reflect the vertical dimension information within 10 min and is aimed to assist dermatologists working with patients suffering from skin diseases via recording or monitoring the skin surface conditions. Additionally, successful trials of loading and electro-controlled release of Cu2+ into/from the developed skin patterns reveals its potential to be also utilized for treatment of pathological skin conditions. Based on the developed analytical diagnostic technique, a well-designed 3D printed portable prototype device based on electrosynthesis of the conducting polymer powered by an ordinary battery (1.5 V) was tested and was found to have excellent performance in onsite 3D skin pattern reproduction from live human skin.
Collapse
Affiliation(s)
- Xiaoxu Fu
- School of Civil and Environmental Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore; Nanyang Environment and Water Research Institute, Residues and Resource Reclamation Center, 1 Cleantech Loop, CleanTech, Singapore, 637141, Singapore
| | - Yi-Heng Cheong
- School of Civil and Environmental Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore; Nanyang Environment and Water Research Institute, Residues and Resource Reclamation Center, 1 Cleantech Loop, CleanTech, Singapore, 637141, Singapore
| | - Ashiq Ahamed
- Nanyang Environment and Water Research Institute, Residues and Resource Reclamation Center, 1 Cleantech Loop, CleanTech, Singapore, 637141, Singapore; Åbo Akademi University, Johan Gadolin Process Chemistry Centre, Laboratory of Molecular Science and Engineering, Biskopsgatan 8, FI-20500, Turku/Åbo, Finland
| | - Chao Zhou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Chima Robert
- Department of Chemistry, University of Otago, 70 Union Street, West Dunedin, 9016, New Zealand
| | - Vida Krikstolaityte
- School of Civil and Environmental Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore; Nanyang Environment and Water Research Institute, Residues and Resource Reclamation Center, 1 Cleantech Loop, CleanTech, Singapore, 637141, Singapore
| | - Keith C Gordon
- Department of Chemistry, University of Otago, 70 Union Street, West Dunedin, 9016, New Zealand
| | - Grzegorz Lisak
- School of Civil and Environmental Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore; Nanyang Environment and Water Research Institute, Residues and Resource Reclamation Center, 1 Cleantech Loop, CleanTech, Singapore, 637141, Singapore.
| |
Collapse
|
125
|
Zheng T, Huang Y, Zhang X, Cai Q, Deng X, Yang X. Mimicking the electrophysiological microenvironment of bone tissue using electroactive materials to promote its regeneration. J Mater Chem B 2020; 8:10221-10256. [PMID: 33084727 DOI: 10.1039/d0tb01601b] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The process of bone tissue repair and regeneration is complex and requires a variety of physiological signals, including biochemical, electrical and mechanical signals, which collaborate to ensure functional recovery. The inherent piezoelectric properties of bone tissues can convert mechanical stimulation into electrical effects, which play significant roles in bone maturation, remodeling and reconstruction. Electroactive materials, including conductive materials, piezoelectric materials and electret materials, can simulate the physiological and electrical microenvironment of bone tissue, thereby promoting bone regeneration and reconstruction. In this paper, the structures and performances of different types of electroactive materials and their applications in the field of bone repair and regeneration are reviewed, particularly by providing the results from in vivo evaluations using various animal models. Their advantages and disadvantages as bone repair materials are discussed, and the methods for tuning their performances are also described, with the aim of providing an up-to-date account of the proposed topics.
Collapse
Affiliation(s)
- Tianyi Zheng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Yiqian Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, P. R. China
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P. R. China
| | - Xiaoping Yang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| |
Collapse
|
126
|
Reid JA, McDonald A, Callanan A. Modulating electrospun polycaprolactone scaffold morphology and composition to alter endothelial cell proliferation and angiogenic gene response. PLoS One 2020; 15:e0240332. [PMID: 33031435 PMCID: PMC7544109 DOI: 10.1371/journal.pone.0240332] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to look at how the composition and morphology of polymer scaffolds could be altered to create an optimized environment for endothelial cells. Four polycaprolactone (PCL) scaffolds were electrospun with increasing fibre diameters ranging from 1.64 μm to 4.83 μm. The scaffolds were seeded with human umbilical vein endothelial cells (HUVEC) and cultured for 12 days. PCL scaffolds were then electrospun incorporating decellularized bovine aorta ECM and cultured in a hypoxic environment. We noted deeper cell infiltration on the largest fibre diameter compared to the other three scaffolds which resulted in an increase in the gene expression of CD31; a key angiogenic marker. Increased cell viability and cell proliferation were also noted on the largest fibre. Furthermore, we noted that the incorporation of extracellular matrix (ECM) had minimal effect on cell viability, both in normoxic and hypoxic culture conditions. Our results showed that these environments had limited influences on hypoxic gene expression. Interestingly, the major findings from this study was the production of excretory ECM components as shown in the scanning electron microscopy (SEM) images. The results from this study suggest that fibre diameter had a bigger impact on the seeded HUVECs than the incorporation of ECM or the culture conditions. The largest fibre dimeter (4.83 μm) is more suitable for seeding of HUVECs.
Collapse
Affiliation(s)
- James Alexander Reid
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alison McDonald
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
| | - Anthony Callanan
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
127
|
Parandeh S, Kharaziha M, Karimzadeh F, Hosseinabadi F. Triboelectric nanogenerators based on graphene oxide coated nanocomposite fibers for biomedical applications. NANOTECHNOLOGY 2020; 31:385402. [PMID: 32498060 DOI: 10.1088/1361-6528/ab9972] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A high demand for green and eco-friendly triboelectric nanogenerators (TENGs) has multiplied the importance of their degradability for biomedical applications. However, the charge generation of current eco-friendly TENGs is generally limited. In this research, a flexible TENG based on a silk fibroin (SF) fibrous layer and a polycaprolactone (PCL)/graphene oxide (GO) fibrous layer was developed. Moreover, the PCL/GO layer was surface modified using various concentrations of GO (0, 1.5, 3, 6, and 9 wt%). We demonstrated that surface modification using GO nanosheets significantly improved the output of the TENG. Notably, the optimized GO modified layer resulted in a voltage of 100 V, a current of 3.15 mA [Formula: see text], and a power density of 72 mW[Formula: see text]. Moreover, a thin PCL layer applied as an encapsulation layer did not significantly modulate the performance of the TENG. Furthermore, during 28 d of soaking in a phosphate buffer solution, the proposed TENG was able to successfully generate electricity. The TENG was also proposed to be used for the electrical stimulation of PC12 cells. The results confirmed that this self-powered electrical stimulator could promote the attachment and proliferation of PC12 cells. Therefore, we have shown the potential for an eco-friendly and cost-effective TENG based on GO modified PCl/GO and silk fibrous layers to be used as a power source for biomedical applications.
Collapse
Affiliation(s)
- S Parandeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | | | | | | |
Collapse
|
128
|
Zhang Z, Zheng T, Zhu R. Microchip with Single-Cell Impedance Measurements for Monitoring Osteogenic Differentiation of Mesenchymal Stem Cells under Electrical Stimulation. Anal Chem 2020; 92:12579-12587. [PMID: 32859132 DOI: 10.1021/acs.analchem.0c02556] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective induction methods and in situ monitoring are essential for studying the mechanism of biological responses in stem cell differentiation. This article proposes an induction method incorporating electrical stimulation under an inhomogeneous field with single-cell impedance monitoring for studying osteogenic differentiation of mesenchymal stem cells (MSCs) using a microchip. The microchip contains an array of sextupole-electrode units for implementing a combination of controllable electrical stimulation and single-cell impedance measurements. MSCs are inducted to osteogenic differentiation under electrical stimulation using quadrupole electrodes and single-cell impedances are monitored in situ using a pair of microelectrodes at each unit center. The proposed microchip adopts an array design to monitor a number of MSCs in parallel, which improves measurement throughput and facilitates to carry out statistic tests. We perform osteogenic differentiation of MSCs on the microchip with and without electrical stimulation meanwhile monitoring single-cell impedance in real time for 21 days. The recorded impedance results show the detailed characteristic change of MSCs at the single-cell level during osteogenic differentiation, which demonstrates a significant difference between the conditions with and without electrical stimulation. The cell morphology and various staining analyses are also used to validate osteogenesis and correlate with the impedance expression. Correlation analysis of the impedance measurement, cell morphology, and various staining assays proves the great acceleration effect of the proposed electrical stimulation on osteogenic differentiation of MSCs. The proposed impedance method can monitor the dynamic process of cell development and study heterogeneity of stem cell differentiation at the single-cell level.
Collapse
Affiliation(s)
- Zhizhong Zhang
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Tianyang Zheng
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Rong Zhu
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| |
Collapse
|
129
|
Ferrigno B, Bordett R, Duraisamy N, Moskow J, Arul MR, Rudraiah S, Nukavarapu SP, Vella AT, Kumbar SG. Bioactive polymeric materials and electrical stimulation strategies for musculoskeletal tissue repair and regeneration. Bioact Mater 2020; 5:468-485. [PMID: 32280836 PMCID: PMC7139146 DOI: 10.1016/j.bioactmat.2020.03.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/15/2020] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
Electrical stimulation (ES) is predominantly used as a physical therapy modality to promote tissue healing and functional recovery. Research efforts in both laboratory and clinical settings have shown the beneficial effects of this technique for the repair and regeneration of damaged tissues, which include muscle, bone, skin, nerve, tendons, and ligaments. The collective findings of these studies suggest ES enhances cell proliferation, extracellular matrix (ECM) production, secretion of several cytokines, and vasculature development leading to better tissue regeneration in multiple tissues. However, there is still a gap in the clinical relevance for ES to better repair tissue interfaces, as ES applied clinically is ineffective on deeper tissue. The use of a conducting material can transmit the stimulation applied from skin electrodes to the desired tissue and lead to an increased function on the repair of that tissue. Ionically conductive (IC) polymeric scaffolds in conjunction with ES may provide solutions to utilize this approach effectively. Injectable IC formulations and their scaffolds may provide solutions for applying ES into difficult to reach tissue types to enable tissue repair and regeneration. A better understanding of ES-mediated cell differentiation and associated molecular mechanisms including the immune response will allow standardization of procedures applicable for the next generation of regenerative medicine. ES, along with the use of IC scaffolds is more than sufficient for use as a treatment option for single tissue healing and may fulfill a role in interfacing multiple tissue types during the repair process.
Collapse
Affiliation(s)
- Bryan Ferrigno
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Rosalie Bordett
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Nithyadevi Duraisamy
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Joshua Moskow
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Michael R. Arul
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Swetha Rudraiah
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Pharmaceutical Sciences, University of Saint Joseph, Hartford, CT, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Anthony T. Vella
- Department of Department of Immunology, University of Connecticut Health, Farmington, CT, USA
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
130
|
Lyons JG, Plantz MA, Hsu WK, Hsu EL, Minardi S. Nanostructured Biomaterials for Bone Regeneration. Front Bioeng Biotechnol 2020; 8:922. [PMID: 32974298 PMCID: PMC7471872 DOI: 10.3389/fbioe.2020.00922] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
This review article addresses the various aspects of nano-biomaterials used in or being pursued for the purpose of promoting bone regeneration. In the last decade, significant growth in the fields of polymer sciences, nanotechnology, and biotechnology has resulted in the development of new nano-biomaterials. These are extensively explored as drug delivery carriers and as implantable devices. At the interface of nanomaterials and biological systems, the organic and synthetic worlds have merged over the past two decades, forming a new scientific field incorporating nano-material design for biological applications. For this field to evolve, there is a need to understand the dynamic forces and molecular components that shape these interactions and influence function, while also considering safety. While there is still much to learn about the bio-physicochemical interactions at the interface, we are at a point where pockets of accumulated knowledge can provide a conceptual framework to guide further exploration and inform future product development. This review is intended as a resource for academics, scientists, and physicians working in the field of orthopedics and bone repair.
Collapse
Affiliation(s)
- Joseph G. Lyons
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute, Northwestern University, Chicago, IL, United States
| | - Mark A. Plantz
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute, Northwestern University, Chicago, IL, United States
| | - Wellington K. Hsu
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute, Northwestern University, Chicago, IL, United States
| | - Erin L. Hsu
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute, Northwestern University, Chicago, IL, United States
| | - Silvia Minardi
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute, Northwestern University, Chicago, IL, United States
| |
Collapse
|
131
|
Effect of electrical stimulation on chondrogenic differentiation of mesenchymal stem cells cultured in hyaluronic acid – Gelatin injectable hydrogels. Bioelectrochemistry 2020; 134:107536. [DOI: 10.1016/j.bioelechem.2020.107536] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
|
132
|
Zhang W, Chu G, Wang H, Chen S, Li B, Han F. Effects of Matrix Stiffness on the Differentiation of Multipotent Stem Cells. Curr Stem Cell Res Ther 2020; 15:449-461. [PMID: 32268870 DOI: 10.2174/1574888x15666200408114632] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 11/22/2022]
Abstract
Differentiation of stem cells, a crucial step in the process of tissue development, repair and regeneration, can be regulated by a variety of mechanical factors such as the stiffness of extracellular matrix. In this review article, the effects of stiffness on the differentiation of stem cells, including bone marrow-derived stem cells, adipose-derived stem cells and neural stem cells, are briefly summarized. Compared to two-dimensional (2D) surfaces, three-dimensional (3D) hydrogel systems better resemble the native environment in the body. Hence, the studies which explore the effects of stiffness on stem cell differentiation in 3D environments are specifically introduced. Integrin is a well-known transmembrane molecule, which plays an important role in the mechanotransduction process. In this review, several integrin-associated signaling molecules, including caveolin, piezo and Yes-associated protein (YAP), are also introduced. In addition, as stiffness-mediated cell differentiation may be affected by other factors, the combined effects of matrix stiffness and viscoelasticity, surface topography, chemical composition, and external mechanical stimuli on cell differentiation are also summarized.
Collapse
Affiliation(s)
- Weidong Zhang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Genglei Chu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Song Chen
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Fengxuan Han
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
133
|
Chen J, Huang Y, Yang J, Li K, Jiang Y, Heng BC, Cai Q, Zhang J, Ge Z. Multiple nanosecond pulsed electric fields stimulation with conductive poly(
l
‐lactic acid)/carbon nanotubes films maintains the multipotency of mesenchymal stem cells during prolonged in vitro culture. J Tissue Eng Regen Med 2020; 14:1136-1148. [DOI: 10.1002/term.3088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Jiaqing Chen
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| | - Yiqian Huang
- State Key Laboratory of Organic‐Inorganic Composites, Beijing Laboratory of Biomedical MaterialsBeijing University of Chemical Technology Beijing China
| | - Jiabei Yang
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| | - Kejia Li
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong Kong Hong Kong China
| | - Boon Chin Heng
- Central LaboratoryPeking University School of Stomatology Beijing Beijing China
| | - Qing Cai
- State Key Laboratory of Organic‐Inorganic Composites, Beijing Laboratory of Biomedical MaterialsBeijing University of Chemical Technology Beijing China
| | - Jue Zhang
- Academy for Advanced Interdisciplinary StudiesPeking University Beijing China
| | - Zigang Ge
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| |
Collapse
|
134
|
Zhao S, Mehta AS, Zhao M. Biomedical applications of electrical stimulation. Cell Mol Life Sci 2020; 77:2681-2699. [PMID: 31974658 PMCID: PMC7954539 DOI: 10.1007/s00018-019-03446-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/12/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
This review provides a comprehensive overview on the biomedical applications of electrical stimulation (EStim). EStim has a wide range of direct effects on both biomolecules and cells. These effects have been exploited to facilitate proliferation and functional development of engineered tissue constructs for regenerative medicine applications. They have also been tested or used in clinics for pain mitigation, muscle rehabilitation, the treatment of motor/consciousness disorders, wound healing, and drug delivery. However, the research on fundamental mechanism of cellular response to EStim has fell behind its applications, which has hindered the full exploitation of the clinical potential of EStim. Moreover, despite the positive outcome from the in vitro and animal studies testing the efficacy of EStim, existing clinical trials failed to establish strong, conclusive supports for the therapeutic efficacy of EStim for most of the clinical applications mentioned above. Two potential directions of future research to improve the clinical utility of EStim are presented, including the optimization and standardization of the stimulation protocol and the development of more tissue-matching devices.
Collapse
Affiliation(s)
- Siwei Zhao
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, 985965 Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Surgery, University of Nebraska Medical Center, Nebraska Medical Center 985965, Omaha, NE, 68198, USA.
| | - Abijeet Singh Mehta
- Department of Dermatology, University of California, Davis, CA, USA
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| | - Min Zhao
- Department of Dermatology, University of California, Davis, CA, USA
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| |
Collapse
|
135
|
Zhao Y, Liang Y, Ding S, Zhang K, Mao HQ, Yang Y. Application of conductive PPy/SF composite scaffold and electrical stimulation for neural tissue engineering. Biomaterials 2020; 255:120164. [PMID: 32554132 DOI: 10.1016/j.biomaterials.2020.120164] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/24/2020] [Accepted: 05/30/2020] [Indexed: 01/31/2023]
Abstract
Electrical stimulation (ES) with conductive polymers can dramatically enhance neurite outgrowth and promote neural regeneration. However, besides ES, the practical applications of neural repair is also highly dependent on the nerve cell functionality and response to substrate conductivity. Therefore, the combination of the ES and suitable materials, such as tissue scaffolds, has been applied to facilitate treatment of neural injuries and demonstrated great potential in peripheral nerve regeneration. In this study, polypyrrole/silk fibroin (PPy/SF) conductive composite scaffold was fabricated by 3D bioprinting and electrospinning. Schwann cells seeded on these scaffolds were electrically stimulated and hence demonstrated enhanced viability, proliferation and migration, as well as upregulated expression of neurotrophic factors. Furthermore, the constructed PPy/SF conductive nerve guidance conduits accompanying with ES could effectively promote axonal regeneration and remyelination in vivo. Moreover, we found that the MAPKs signal transduction pathway was activated by ES at the conductive conduit. Our findings demonstrate that the PPy/SF conductive composite scaffolds with longitudinal guidance exhibit favorable properties for clinical use and promotes nerve regeneration and functional recovery.
Collapse
Affiliation(s)
- Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, PR China; Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, 226001, PR China; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yunyun Liang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, PR China; Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, 226001, PR China
| | - Supeng Ding
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kunyu Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, PR China; Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, 226001, PR China.
| |
Collapse
|
136
|
Hu M, Hong L, He S, Huang G, Cheng Y, Chen Q. Effects of electrical stimulation on cell activity, cell cycle, cell apoptosis and β‑catenin pathway in the injured dorsal root ganglion cell. Mol Med Rep 2020; 21:2385-2394. [PMID: 32323840 PMCID: PMC7185288 DOI: 10.3892/mmr.2020.11058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/11/2020] [Indexed: 12/02/2022] Open
Abstract
The present study aimed to investigate the effects of electrical stimulation (ES) on cell activity, cell cycle and apoptosis in injured rat dorsal root ganglion (DRG) cells induced by cyclic mechanical stretching (CMS). The present study also investigated whether the Wnt/β‑catenin pathway is involved in this process. Injury and ES models were established in DRG cells. Then, cell activity was detected using a Cell Counting Kit‑8 and 5‑ethynyl‑2'‑deoxyuridine‑594 cell proliferation assay kit. Cell cycle distribution was detected using a cell cycle detection kit. Apoptosis was detected using an Annexin V‑FITC apoptosis detection kit, and Wnt/β‑catenin pathway‑associated proteins were detected using western blotting. The present study demonstrated that CMS decreased DRG cell activity, increased the number of cells in the S phase, promoted cell apoptosis and inhibited the Wnt/β‑catenin pathway. In addition, ES significantly increased the proliferation activity of DRG cells, increased the number of cells in the G2 phase, decreased the apoptotic rate and activated the Wnt/β‑catenin pathway, ultimately reversing the injury caused by CMS. Following inhibition of the Wnt/β‑catenin signaling pathway using XAV939, the effects of ES were weakened. In conclusion, the present study demonstrated that ES may reverse CMS‑induced injury in DRG cells, and that the Wnt signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Ming Hu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Songming He
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Guotao Huang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yanxiang Cheng
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
137
|
Oliveira KMC, Leppik L, Keswani K, Rajeev S, Bhavsar MB, Henrich D, Barker JH. Electrical Stimulation Decreases Dental Pulp Stem Cell Osteo-/Odontogenic Differentiation. Biores Open Access 2020; 9:162-173. [PMID: 32642331 PMCID: PMC7337168 DOI: 10.1089/biores.2020.0002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Dental pulp stem cells (DPSCs) have great potential for use in tissue engineering (TE)-based dental treatments. Electrical stimulation (EStim) has been shown to influence cellular functions that could play an important role in the success of TE treatments. Despite many recent studies focused on DPSCs, few have investigated the effect EStim has on these cells. The aim of this research was to investigate the effects of direct current (DC) EStim on osteo-/odontogenic differentiation of DPSCs. To do so cells were isolated from male Sprague Dawley rats (7-8 weeks old), and phenotype characterization and multilineage differentiation analysis were conducted to verify their "stemness." Different voltages of DC EStim were administrated 1 h/day for 7 days, and the effect of EStim on DPSC osteo-/odontogenic differentiation was assessed by measuring calcium and collagen deposition, alkaline phosphatase (ALP) activity, and expression of osteo- and odontogenic marker genes at days 7 and 14 of culture. We found that while 10 and 50 mV/mm of EStim had no effect on cell number or metabolic activity, 100 mV/mm caused a significant reduction in cell number, and 150 mV/mm resulted in cell death. Despite increased gene expression of osteo-/odontogenic gene markers, Osteocalcin, RunX2, BSP, and DMP1, at day 7 in EStim treated cells, 50 mV/mm of EStim decreased collagen deposition and ALP activity at both time points, and calcium deposition was found to be lower at day 14. In conclusion, under the conditions tested, EStim appears to impair DPSC osteo-/odontogenic differentiation. Additional studies are needed to further characterize and understand the mechanisms involved in DPSC response to EStim, with an eye toward its potential use in TE-based dental treatments.
Collapse
Affiliation(s)
| | - Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, J.W. Goethe-University, Frankfurt/Main, Germany
| | - Khyati Keswani
- Frankfurt Initiative for Regenerative Medicine, J.W. Goethe-University, Frankfurt/Main, Germany
| | - Sreeraj Rajeev
- Frankfurt Initiative for Regenerative Medicine, J.W. Goethe-University, Frankfurt/Main, Germany
| | - Mit B. Bhavsar
- Frankfurt Initiative for Regenerative Medicine, J.W. Goethe-University, Frankfurt/Main, Germany
| | - Dirk Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, J.W. Goethe-University, Frankfurt/Main, Germany
| | - John H. Barker
- Frankfurt Initiative for Regenerative Medicine, J.W. Goethe-University, Frankfurt/Main, Germany
| |
Collapse
|
138
|
Bordoni M, Scarian E, Rey F, Gagliardi S, Carelli S, Pansarasa O, Cereda C. Biomaterials in Neurodegenerative Disorders: A Promising Therapeutic Approach. Int J Mol Sci 2020; 21:ijms21093243. [PMID: 32375302 PMCID: PMC7247337 DOI: 10.3390/ijms21093243] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative disorders (i.e., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and spinal cord injury) represent a great problem worldwide and are becoming prevalent because of the increasing average age of the population. Despite many studies having focused on their etiopathology, the exact cause of these diseases is still unknown and until now, there are only symptomatic treatments. Biomaterials have become important not only for the study of disease pathogenesis, but also for their application in regenerative medicine. The great advantages provided by biomaterials are their ability to mimic the environment of the extracellular matrix and to allow the growth of different types of cells. Biomaterials can be used as supporting material for cell proliferation to be transplanted and as vectors to deliver many active molecules for the treatments of neurodegenerative disorders. In this review, we aim to report the potentiality of biomaterials (i.e., hydrogels, nanoparticles, self-assembling peptides, nanofibers and carbon-based nanomaterials) by analyzing their use in the regeneration of neural and glial cells their role in axon outgrowth. Although further studies are needed for their use in humans, the promising results obtained by several groups leads us to suppose that biomaterials represent a potential therapeutic approach for the treatments of neurodegenerative disorders.
Collapse
Affiliation(s)
- Matteo Bordoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy;
| | - Eveljn Scarian
- Department of Brain and Behavioural Sciences, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy;
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
| | - Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (S.C.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milan, Via Grassi, 74, 20157 Milan, Italy
| | - Stella Gagliardi
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (S.C.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milan, Via Grassi, 74, 20157 Milan, Italy
| | - Orietta Pansarasa
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
- Correspondence: ; Tel.: +39-0382-380-248
| | - Cristina Cereda
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
| |
Collapse
|
139
|
Altered β-Cell Calcium Dynamics via Electric Field Exposure. Ann Biomed Eng 2020; 49:106-114. [PMID: 32323041 DOI: 10.1007/s10439-020-02517-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Electric field stimulation has long been investigated with results supporting its therapeutic potential; however, its effects on insulin secreting cells has yet to be fully elucidated. Herein we explored the effects of physiological direct current (DC) electric field stimulation on the intracellular calcium dynamics of mouse derived βTC-6 insulinoma cells. This electrical stimulation resulted in an elevation in intracellular calcium along with a rise in calcium spiking activity. Further investigation indicated that the rise in intracellular calcium was mediated by an influx of calcium via L-type voltage gated calcium channels. Additionally, the effects of the electric field stimulation were able to induce insulin secretion in the absence of glucose stimulation. Given these results, DC electric field stimulation could be used as a non-invasive tool to modulate intracellular calcium dynamics and insulin secretion of β-cells for therapeutic application.
Collapse
|
140
|
Vieira D, Angel S, Honjol Y, Gruenheid S, Gbureck U, Harvey E, Merle G. Electroceutical Silk-Silver Gel to Eradicate Bacterial Infection. ACTA ACUST UNITED AC 2020; 4:e1900242. [PMID: 32293155 DOI: 10.1002/adbi.201900242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/24/2020] [Indexed: 01/23/2023]
Abstract
With more than 50% of bacteria resistant to standard antibiotics, new strategies to treat bacterial infection and colonization are needed. Based on the concept of targeting the bacteria synergistically on various fronts, it is hypothesized that an electrical insult associated with antibacterial materials may be a highly effective means of killing bacteria. In this work, an injectable conductive gel based on silk fibroin (SF) and silver nanoparticles (Ag-NPs) is synthesized, capable of coating a zone of injury, allowing the application of a low electrical current to decrease bacterial contamination. With a high conductivity of 1.5 S cm-1 , SF/Ag-NPs gels killed 80% of Escherichia coli in 1 min, no toxicity toward Chinese hamster ovary cells is observed. The mechanism of an electrical composite gel combined with electrical wound therapy is associated with silver ion (Ag+ ) release, and reactive oxygen species (ROS) production. The findings in the present study show a similar Ag+ release for treatment with gels and the combined effect, whereas ROS generation is 50% higher when a small electrical current is applied leading to a broad bactericidal effect.
Collapse
Affiliation(s)
- Daniela Vieira
- Experimental Surgery, Faculty of Medicine, McGill University, Montreal, H3A 2B2, Canada
| | - Samuel Angel
- Experimental Surgery, Faculty of Medicine, McGill University, Montreal, H3A 2B2, Canada
| | - Yazan Honjol
- Experimental Surgery, Faculty of Medicine, McGill University, Montreal, H3A 2B2, Canada
| | | | - Uwe Gbureck
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Würzburg, 97070, Germany
| | | | - Geraldine Merle
- McGill University, Montreal, H3A 0C5, Canada.,Polytechnique Montréal, C.P. 6079, succ. Centre-ville, Montréal, H3C 3A7, Québec, Canada
| |
Collapse
|
141
|
Leppik L, Oliveira KMC, Bhavsar MB, Barker JH. Electrical stimulation in bone tissue engineering treatments. Eur J Trauma Emerg Surg 2020; 46:231-244. [PMID: 32078704 PMCID: PMC7113220 DOI: 10.1007/s00068-020-01324-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
Abstract
Electrical stimulation (EStim) has been shown to promote bone healing and regeneration both in animal experiments and clinical treatments. Therefore, incorporating EStim into promising new bone tissue engineering (BTE) therapies is a logical next step. The goal of current BTE research is to develop combinations of cells, scaffolds, and chemical and physical stimuli that optimize treatment outcomes. Recent studies demonstrating EStim's positive osteogenic effects at the cellular and molecular level provide intriguing clues to the underlying mechanisms by which it promotes bone healing. In this review, we discuss results of recent in vitro and in vivo research focused on using EStim to promote bone healing and regeneration and consider possible strategies for its application to improve outcomes in BTE treatments. Technical aspects of exposing cells and tissues to EStim in in vitro and in vivo model systems are also discussed.
Collapse
Affiliation(s)
- Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany.
| | - Karla Mychellyne Costa Oliveira
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - Mit Balvantray Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - John Howard Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| |
Collapse
|
142
|
Tomaskovic-Crook E, Gu Q, Rahim SNA, Wallace GG, Crook JM. Conducting Polymer Mediated Electrical Stimulation Induces Multilineage Differentiation with Robust Neuronal Fate Determination of Human Induced Pluripotent Stem Cells. Cells 2020; 9:cells9030658. [PMID: 32182797 PMCID: PMC7140718 DOI: 10.3390/cells9030658] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/15/2022] Open
Abstract
Electrical stimulation is increasingly being used to modulate human cell behaviour for biotechnological research and therapeutics. Electrically conductive polymers (CPs) such as polypyrrole (PPy) are amenable to in vitro and in vivo cell stimulation, being easy to synthesise with different counter ions (dopants) to augment biocompatibility and cell-effects. Extending our earlier work, which showed that CP-mediated electrical stimulation promotes human neural stem cell differentiation, here we report using electroactive PPy containing the anionic dopant dodecylbenzenesulfonate (DBS) to modulate the fate determination of human induced pluripotent stem cells (iPSCs). Remarkably, the stimulation without conventional chemical inducers resulted in the iPSCs differentiating to cells of the three germ lineages-endoderm, ectoderm, and mesoderm. The unstimulated iPSC controls remained undifferentiated. Phenotypic characterisation further showed a robust induction to neuronal fate with electrical stimulation, again without customary chemical inducers. Our findings add to the growing body of evidence supporting the use of electrical stimulation to augment stem cell differentiation, more specifically, pluripotent stem cell differentiation, and especially neuronal induction. Moreover, we have shown the versatility of electroactive PPy as a cell-compatible platform for advanced stem cell research and translation, including identifying novel mechanisms of fate regulation, tissue development, electroceuticals, and regenerative medicine.
Collapse
Affiliation(s)
- Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500 Wollongong, Australia; (E.T.-C.); (Q.G.); (S.N.A.R.)
- Illawarra Health and Medical Research Institute, University of Wollongong, 2500 Wollongong, Australia
| | - Qi Gu
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500 Wollongong, Australia; (E.T.-C.); (Q.G.); (S.N.A.R.)
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100000 Beijing, China
| | - Siti N Abdul Rahim
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500 Wollongong, Australia; (E.T.-C.); (Q.G.); (S.N.A.R.)
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500 Wollongong, Australia; (E.T.-C.); (Q.G.); (S.N.A.R.)
- Correspondence: (G.G.W.); (J.M.C.); Tel.: +61-2-4221-3127 (G.G.W.); +61-2-4221-3011 (J.M.C.)
| | - Jeremy M Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, 2500 Wollongong, Australia; (E.T.-C.); (Q.G.); (S.N.A.R.)
- Illawarra Health and Medical Research Institute, University of Wollongong, 2500 Wollongong, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, 3065 Fitzroy, Australia
- Correspondence: (G.G.W.); (J.M.C.); Tel.: +61-2-4221-3127 (G.G.W.); +61-2-4221-3011 (J.M.C.)
| |
Collapse
|
143
|
Angulo-Pineda C, Srirussamee K, Palma P, Fuenzalida VM, Cartmell SH, Palza H. Electroactive 3D Printed Scaffolds Based on Percolated Composites of Polycaprolactone With Thermally Reduced Graphene Oxide for Antibacterial and Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E428. [PMID: 32121237 PMCID: PMC7152842 DOI: 10.3390/nano10030428] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Applying electrical stimulation (ES) could affect different cellular mechanisms, thereby producing a bactericidal effect and an increase in human cell viability. Despite its relevance, this bioelectric effect has been barely reported in percolated conductive biopolymers. In this context, electroactive polycaprolactone (PCL) scaffolds with conductive Thermally Reduced Graphene Oxide (TrGO) nanoparticles were obtained by a 3D printing method. Under direct current (DC) along the percolated scaffolds, a strong antibacterial effect was observed, which completely eradicated S. aureus on the surface of scaffolds. Notably, the same ES regime also produced a four-fold increase in the viability of human mesenchymal stem cells attached to the 3D conductive PCL/TrGO scaffold compared with the pure PCL scaffold. These results have widened the design of novel electroactive composite polymers that could both eliminate the bacteria adhered to the scaffold and increase human cell viability, which have great potential in tissue engineering applications.
Collapse
Affiliation(s)
- Carolina Angulo-Pineda
- Department of Chemical Engineering and Biotechnology and Materials, University of Chile, Santiago 8370456, Chile
- Millenium Nuclei in Soft Smart Mechanical Metamaterials, Universidad de Chile, Santiago 8370456, Chile
| | - Kasama Srirussamee
- Department of Biomedical Engineering, Faculty of Engineering, King Mongkut’s Institute of Technology Ladkrabang (KMITL), Bangkok 10520, Thailand;
| | - Patricia Palma
- Department of Pathology and Oral Medicine, University of Chile, Santiago 8380492, Chile;
| | | | - Sarah H. Cartmell
- Department of Materials, The University of Manchester, Manchester M13 9PL, UK;
| | - Humberto Palza
- Department of Chemical Engineering and Biotechnology and Materials, University of Chile, Santiago 8370456, Chile
- Millenium Nuclei in Soft Smart Mechanical Metamaterials, Universidad de Chile, Santiago 8370456, Chile
| |
Collapse
|
144
|
Zheng T, Zhang Z, Zhu R, Sun D. A microelectrode array chip for osteogenic differentiation of mesenchymal stem cells under electrical stimulation. LAB ON A CHIP 2020; 20:373-383. [PMID: 31850469 DOI: 10.1039/c9lc01081e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Electrical stimulation (ES) as an easy and effective inducing method has been widely used in induction differentiation of stem cells, e.g. osteogenic differentiation of mesenchymal stem cells (MSCs) for bone healing and bone tissue therapies. However, the micro-effect of an inhomogeneous electric field has rarely been investigated for ES in induction differentiation, and conventionally used ex situ assays may preclude accurate assessment due to variation from cell inoculation and treatments. Here, a novel electrical stimulation method with a microelectrode array chip is proposed for osteogenic differentiation of MSCs. The electric field applied onto the MSCs by the microelectrode array is designed similarly with a natural aggregation distribution of differentiated MSCs. The proposed ES method accelerates osteoblast proliferation and differentiation in the electrode array region and generates a larger amount of mineralized deposits, which are assayed via in situ alizarin red staining and morphology observation as well as immunocytochemistry. In addition, this method allows a direct in situ assessment to compare the osteogenic differentiation of MSCs with and without ES on a single chip to avoid culture environment difference. The method provides a fundamental platform for investigating induced differentiation of stem cells and allows integration with multifunctional cell assays to achieve in situ tracking for the differentiation process of stem cells.
Collapse
Affiliation(s)
- Tianyang Zheng
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Zhizhong Zhang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Rong Zhu
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Dong Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
145
|
Budde K, Zimmermann J, Neuhaus E, Schroder M, Uhrmacher AM, van Rienen U. Requirements for Documenting Electrical Cell Stimulation Experiments for Replicability and Numerical Modeling ∗. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:1082-1088. [PMID: 31946082 DOI: 10.1109/embc.2019.8856863] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Thorough documentation of biological experiments is necessary for their replicability. This becomes even more evident when individual steps of in vitro wet-lab experiments are to be incorporated into computer simulation models. In the highly interdisciplinary field of electrical stimulation of biological cells, not only biological but also physical aspects play a crucial role. Simulations may help to identify parameters that influence cells and thereby reveal new insights into mechanisms of the cell biological system. However, missing or misleading documentation of the electrical stimulation step within wet-lab experiments may lead to discrepancies between reported and simulated electrical quantities. In addition, this threatens the replicability of electrical stimulation experiments. Thus, we argue that a minimal set of information is needed to enable a translation of electrical stimulation experiments of biological cells into computer simulation experiments and to support replicability. This set includes detailed information about the electronic devices and components, their set-up as well as the applied stimulus and shall be integrated into an existing guideline for cell biological experiments. Ideally, the documentation should also contain measured properties of the cellular and experimental environment. Furthermore, a realization of our proposed documentation requirements within electronic lab notebooks may provide a crucial step toward a more seamless integration of wet-lab data into simulations. Based on two exemplary studies, we demonstrate the relevance of our claim.
Collapse
|
146
|
Vandghanooni S, Eskandani M. Natural polypeptides-based electrically conductive biomaterials for tissue engineering. Int J Biol Macromol 2020; 147:706-733. [PMID: 31923500 DOI: 10.1016/j.ijbiomac.2019.12.249] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/28/2019] [Accepted: 12/28/2019] [Indexed: 12/11/2022]
Abstract
Fabrication of an appropriate scaffold is the key fundamental step required for a successful tissue engineering (TE). The artificial scaffold as extracellular matrix in TE has noticeable role in the fate of cells in terms of their attachment, proliferation, differentiation, orientation and movement. In addition, chemical and electrical stimulations affect various behaviors of cells such as polarity and functionality. Therefore, the fabrication approach and materials used for the preparation of scaffold should be more considered. Various synthetic and natural polymers have been used extensively for the preparation of scaffolds. The electrically conductive polymers (ECPs), moreover, have been used in combination with other polymers to apply electric fields (EF) during TE. In this context, composites of natural polypeptides and ECPs can be taken into account as context for the preparation of suitable scaffolds with superior biological and physicochemical features. In this review, we overviewed the simultaneous usage of natural polypeptides and ECPs for the fabrication of scaffolds in TE.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Research Center for Pharmaceutical Nanotechnology, Biomedicine institute, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
147
|
Abasi S, Aggas JR, Venkatesh N, Vallavanatt IG, Guiseppi-Elie A. Design, fabrication and testing of an electrical cell stimulation and recording apparatus (ECSARA) for cells in electroculture. Biosens Bioelectron 2020; 147:111793. [DOI: 10.1016/j.bios.2019.111793] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
|
148
|
Cottrill E, Pennington Z, Ahmed AK, Lubelski D, Goodwin ML, Perdomo-Pantoja A, Westbroek EM, Theodore N, Witham T, Sciubba D. The effect of electrical stimulation therapies on spinal fusion: a cross-disciplinary systematic review and meta-analysis of the preclinical and clinical data. J Neurosurg Spine 2020; 32:106-126. [PMID: 31593923 DOI: 10.3171/2019.5.spine19465] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/17/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Nonunion is a common complication of spinal fusion surgeries. Electrical stimulation technologies (ESTs)-namely, direct current stimulation (DCS), capacitive coupling stimulation (CCS), and inductive coupling stimulation (ICS)-have been suggested to improve fusion rates. However, the evidence to support their use is based solely on small trials. Here, the authors report the results of meta-analyses of the preclinical and clinical data from the literature to provide estimates of the overall effect of these therapies at large and in subgroups. METHODS A systematic review of the English-language literature was performed using PubMed, Embase, and Web of Science databases. The query of these databases was designed to include all preclinical and clinical studies examining ESTs for spinal fusion. The primary endpoint was the fusion rate at the last follow-up. Meta-analyses were performed using a Freeman-Tukey double arcsine transformation followed by random-effects modeling. RESULTS A total of 33 articles (17 preclinical, 16 clinical) were identified, of which 11 preclinical studies (257 animals) and 13 clinical studies (2144 patients) were included in the meta-analysis. Among preclinical studies, the mean fusion rates were higher among EST-treated animals (OR 4.79, p < 0.001). Clinical studies similarly showed ESTs to increase fusion rates (OR 2.26, p < 0.001). Of EST modalities, only DCS improved fusion rates in both preclinical (OR 5.64, p < 0.001) and clinical (OR 2.13, p = 0.03) populations; ICS improved fusion in clinical studies only (OR 2.45, p = 0.014). CCS was not effective at increasing fusion, although only one clinical study was identified. A subanalysis of the clinical studies found that ESTs increased fusion rates in the following populations: patients with difficult-to-fuse spines, those who smoke, and those who underwent multilevel fusions. CONCLUSIONS The authors found that electrical stimulation devices may produce clinically significant increases in arthrodesis rates among patients undergoing spinal fusion. They also found that the pro-arthrodesis effects seen in preclinical studies are also found in clinical populations, suggesting that findings in animal studies are translatable. Additional research is needed to analyze the cost-effectiveness of these devices.
Collapse
|
149
|
Electric Phenomenon: A Disregarded Tool in Tissue Engineering and Regenerative Medicine. Trends Biotechnol 2020; 38:24-49. [DOI: 10.1016/j.tibtech.2019.07.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
|
150
|
Kim S, Cha C. Enhanced mechanical and electrical properties of heteroscaled hydrogels infused with aqueous-dispersible hybrid nanofibers. Biofabrication 2019; 12:015020. [DOI: 10.1088/1758-5090/ab5385] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|