1
|
Bystrov DA, Volegova DD, Korsakova SA, Salmina AB, Yurchenko SO. Electric Field-Induced Effects in Eukaryotic Cells: Current Progress and Limitations. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40279199 DOI: 10.1089/ten.teb.2025.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Electric fields (EFs) offer a powerful tool for manipulating cells and modulating their behavior, holding significant promise for regenerative medicine and cell biology. We provide a comprehensive overview of the effects of different types of EF on eukaryotic cells with the special focus on physical mechanisms and signaling pathways involved. Direct current EF induces electrophoresis and electroosmosis, influencing cell migration, proliferation, and differentiation. Alternating current EF, through dielectric polarization and dielectrophoresis, enables cell manipulation, trapping, and sorting. Pulsed EF, particularly high-intensity, short-duration pulses, induces reversible and irreversible electroporation, facilitating drug and gene delivery. The review covers some technological aspects of EF generation, emphasizing the importance of experimental setups, and integration with microfluidic platforms for high-throughput analysis and precise manipulations. Furthermore, the synergistic potential of combining EFs with optical tweezers is highlighted, enabling fine-tuned control of cell positioning, intercellular interactions, and measurement of biophysical properties. Finally, the review addresses limitations of EF application, such as field heterogeneity and potential side effects, and outlines the directions for future studies, including developing the minimally invasive delivery methods.
Collapse
Affiliation(s)
- Daniil A Bystrov
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| | - Daria D Volegova
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| | - Sofia A Korsakova
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| | - Alla B Salmina
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
| | - Stanislav O Yurchenko
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| |
Collapse
|
2
|
Li C, Meng X, Li S, Wang C. Therapeutic Advances in Peripheral Nerve Injuries: Nerve-Guided Conduit and Beyond. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40195945 DOI: 10.1089/ten.teb.2024.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Peripheral nerve injury (PNI), a challenging neurosurgery issue, often leads to partial or complete loss of neuronal functions and even neuropathic pain. Thus far, the gold standard for treating peripheral nerve deficit remains autografts. While numerous reviews have explored PNI and regeneration, this work distinctively synthesizes recent advancements in tissue engineering-particularly four-dimensional (4D) bioprinting and exosome therapies-with an emphasis on their clinical translation. By consolidating findings spanning molecular mechanisms to therapeutic applications, this review proposes an actionable framework for advancing experimental strategies toward clinically viable solutions. Our work critically evaluates emerging innovations such as dynamically adaptive 4D-printed nerve conduits and exosome-based therapies, underscoring their potential to match conventional autografts in achieving functional restoration. Impact Statement Although several previous reviews have been made on describing with great detail the degenerative and regenerative mechanisms of the peripheral nervous systems, as well as the several existing and exploratory treatment strategies, we focus more on the latest advancements of each of those topics.
Collapse
Affiliation(s)
- Changqing Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xianyu Meng
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shengji Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjing Wang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
3
|
Boulingre M, Chodkowski M, Portillo Lara R, Lee A, Goding J, Green RA. Multi-layered electrode constructs for neural tissue engineering. J Mater Chem B 2025; 13:3390-3404. [PMID: 39935279 DOI: 10.1039/d4tb02651a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Although neural tissue engineering holds great therapeutic potential for multiple clinical applications, one important challenge is the development of scaffolds that provide cues required for neural tissue development. To achieve this, biomaterial systems can be leveraged to present appropriate biological, mechanical, topographical and electrical cues that could direct cell fate. In this study, a multi-layered electrode construct was engineered to be used as a platform for 3D cell encapsulation for in vitro applications. The first layer is a conductive hydrogel coating, that improves electrical conductivity from the underlying platinum electrode. The second layer is a biosynthetic hydrogel, specifically tailored to support neural development. This layered electrode construct was electrochemically characterised, and a numerical model was applied to study electrical stimuli reaching the biosynthetic hydrogel layer. The construct was shown to effectively support the growth and proliferation of encapsulated astrocytes within the biosynthetic layer, while the numerical model will enable computational experimentation for benchmarking and study validation. This highly versatile system represents a robust tool to study the influence of electrical stimuli on neural fate, as well as investigating the development of biohybrid interfaces in vitro.
Collapse
Affiliation(s)
- Marjolaine Boulingre
- Department of Bioengineering, Imperial College London, South Kensington, London, UK.
| | - Mateusz Chodkowski
- Department of Bioengineering, Imperial College London, South Kensington, London, UK.
| | - Roberto Portillo Lara
- Department of Bioengineering, Imperial College London, South Kensington, London, UK.
| | - Aaron Lee
- Department of Bioengineering, Imperial College London, South Kensington, London, UK.
| | - Josef Goding
- Department of Bioengineering, Imperial College London, South Kensington, London, UK.
| | - Rylie A Green
- Department of Bioengineering, Imperial College London, South Kensington, London, UK.
| |
Collapse
|
4
|
Grassi A, Rocca MS, Noventa M, Pozzato G, Pozzato A, Scioscia M, Andrisani A, Pontrelli G, Foresta C, De Toni L. In Vitro Gene Expression Profiling of Quantum Molecular Resonance Effects on Human Endometrium Models: A Preliminary Study. Genes (Basel) 2025; 16:290. [PMID: 40149442 PMCID: PMC11942151 DOI: 10.3390/genes16030290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVES The identification of methods to improve the endometrial receptivity (ER) is increasingly of interest. The effect of the electromagnetic field associated with Quantum Molecular Resonance (QMR) on ER was investigated here. METHODS Ishikawa cells were used to evaluate the effects of QMR both on the expression of a group of genes involved in ER, i.e., HOXA10, HOXA11, LIF, ITGB3, and ITGAV, and on cell toxicity. Endometrial samples were obtained from six patients during routine diagnostic procedures, four of which were subsequently used to assess the transcriptional response to QMR through microarray. RESULTS Compared to unexposed controls, a single exposure of Ishikawa cells to QMR for 20 min was associated with a significant and power-dependent up-regulation of all the selected ER-related genes up to 8 power units (PU). Repeated exposure to QMR, up to three consecutive days, showed a significant up-regulation of all the selected genes at power values of 4 PU, from day two onwards. Negligible cytotoxicity was observed. Gene set enrichment analysis, on microarray data of endometrial biopsies stimulated for three consecutive days at 4 PU, showed a significant enrichment of specific gene sets, related to the proteasome system, the cell adhesion, the glucocorticoid receptor, and cell cycle pathways. CONCLUSIONS Our results suggest a possible favorable impact of QMR on ER.
Collapse
Affiliation(s)
- Angela Grassi
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Maria Santa Rocca
- Department of Medicine, University of Padova, 35128 Padova, Italy; (M.S.R.); (C.F.)
| | - Marco Noventa
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padova, 35100 Padova, Italy; (M.N.); (A.A.)
| | | | - Alessandro Pozzato
- Telea Electronic Engineering S.r.l., 36066 Sandrigo, Italy; (G.P.); (A.P.)
| | - Marco Scioscia
- Unit of Gynecological Surgery, Mater Dei Hospital, 70125 Bari, Italy;
| | - Alessandra Andrisani
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padova, 35100 Padova, Italy; (M.N.); (A.A.)
| | - Giovanni Pontrelli
- Department of Obstetrics and Gynecology, Policlinico Hospital, 35031 Abano Terme, Italy;
| | - Carlo Foresta
- Department of Medicine, University of Padova, 35128 Padova, Italy; (M.S.R.); (C.F.)
| | - Luca De Toni
- Department of Medicine, University of Padova, 35128 Padova, Italy; (M.S.R.); (C.F.)
| |
Collapse
|
5
|
Borah R, Diez Clarke D, Upadhyay J, Monaghan MG. From innovation to clinic: Emerging strategies harnessing electrically conductive polymers to enhance electrically stimulated peripheral nerve repair. Mater Today Bio 2025; 30:101415. [PMID: 39816667 PMCID: PMC11733191 DOI: 10.1016/j.mtbio.2024.101415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
Peripheral nerve repair (PNR) is a major healthcare challenge due to the limited regenerative capacity of the nervous system, often leading to severe functional impairments. While nerve autografts are the gold standard, their implications are constrained by issues such as donor site morbidity and limited availability, necessitating innovative alternatives like nerve guidance conduits (NGCs). However, the inherently slow nerve growth rate (∼1 mm/day) and prolonged neuroinflammation, delay recovery even with the use of passive (no-conductive) NGCs, resulting in muscle atrophy and loss of locomotor function. Electrical stimulation (ES) has the ability to enhance nerve regeneration rate by modulating the innate bioelectrical microenvironment of nerve tissue while simultaneously fostering a reparative environment through immunoregulation. In this context, electrically conductive polymer (ECP)-based biomaterials offer unique advantages for nerve repair combining their flexibility, akin to traditional plastics, and mixed ionic-electronic conductivity, similar to ionically conductive nerve tissue, as well as their biocompatibility and ease of fabrication. This review focuses on the progress, challenges, and emerging techniques for integrating ECP based NGCs with ES for functional nerve regeneration. It critically evaluates the various approaches using ECP based scaffolds, identifying gaps that have hindered clinical translation. Key challenges discussed include designing effective 3D NGCs with high electroactivity, optimizing ES modules, and better understanding of immunoregulation during nerve repair. The review also explores innovative strategies in material development and wireless, self-powered ES methods. Furthermore, it emphasizes the need for non-invasive ES delivery methods combined with hybrid ECP based neural scaffolds, highlighting future directions for advancing preclinical and clinical translation. Together, ECP based NGCs combined with ES represent a promising avenue for advancing PNR and improving patient outcomes.
Collapse
Affiliation(s)
- Rajiv Borah
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Daniel Diez Clarke
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Jnanendra Upadhyay
- Department of Physics, Dakshin Kamrup College, Kamrup, Assam, 781125, India
| | - Michael G. Monaghan
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Research in Medical Devices, University of Galway, H91 W2TY Galway, Ireland
| |
Collapse
|
6
|
Zhou H, Zhang S, Jin X, A C, Gong P, Zhao S. The Electric Field Guided HaCaT Cell Migration Through the EGFR/p38 MAPK/Akt Pathway. Curr Issues Mol Biol 2024; 47:16. [PMID: 39852131 PMCID: PMC11763975 DOI: 10.3390/cimb47010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/28/2024] [Indexed: 01/26/2025] Open
Abstract
Previous studies have shown that the endogenous electric field (EF) is an overriding cure in guiding cell migration toward the wound center to promote wound healing, but the mechanism underlying is unclear. In this study, we investigated the molecular mechanism of electric field-guided cell migration in human keratinocyte HaCaT cells. Our results showed that HaCaT cells migrate toward the anode under EFs. The phosphorylation levels of p38 MAPK and Akt were obviously elevated in the EF. Knocking down p38 MAPK obviously abolished directed migration of HaCaT cells under the EFs. Inhibiting p38 MAPK by SB203580 impaired the EF-guided cell migration. The electric field may guide HaCaT cell migration through the EGFR/p38 MAPK/Akt pathway.
Collapse
Affiliation(s)
| | | | | | | | - Peng Gong
- School of Life Sciences, Yunnan Normal University, Kunming 650500, China; (H.Z.); (S.Z.); (X.J.); (C.A.)
| | - Sanjun Zhao
- School of Life Sciences, Yunnan Normal University, Kunming 650500, China; (H.Z.); (S.Z.); (X.J.); (C.A.)
| |
Collapse
|
7
|
Deng K, Luo R, Chen Y, Liu X, Xi Y, Usman M, Jiang X, Li Z, Zhang J. Electrical Stimulation Therapy - Dedicated to the Perfect Plastic Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2409884. [PMID: 39680745 DOI: 10.1002/advs.202409884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Tissue repair and reconstruction are a clinical difficulty. Bioelectricity has been identified as a critical factor in supporting tissue and cell viability during the repair process, presenting substantial potential for clinical application. This review delves into various sources of electrical stimulation and identifies appropriate electrode materials for clinical use. It also highlights the biological mechanisms of electrical stimulation at both the subcellular and cellular levels, elucidating how these interactions facilitate the repair and regeneration processes across different organs. Moreover, specific electrode materials and stimulation sources are outlined, detailing their impact on cellular activity. The future development trends are projected from two perspectives: the optimization of equipment performance and the fulfillment of clinical demands, focusing on the feasibility, safety, and cost-effectiveness of technologies.
Collapse
Affiliation(s)
- Kexin Deng
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ruizeng Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Chen
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiaoqiang Liu
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuanyin Xi
- A Breast Disease Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Muhammad Usman
- Department of Plastic Surgery and Burn, Central Hospital Affiliated with Chongqing University of Technology, Chongqing, 400054, P.R. China
| | - Xupin Jiang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhou Li
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaping Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
8
|
Zironi I, Cramer T, Fuschi A, Cioni M, Guerra G, Giuliani G, Calienni M, Caramazza L, Liberti M, Apollonio F, Remondini D, Castellani G. Enhancing cell motility via non-contact capacitively coupled electrostatic field. Sci Rep 2024; 14:28085. [PMID: 39543219 PMCID: PMC11564694 DOI: 10.1038/s41598-024-77384-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Cellular motility is essential for making and maintaining multicellular organisms throughout their lifespan. Migrating cells can move either individually or collectively by a crawling movement that links the cytoskeletal activity to the adhesion surface. In vitro stimulation by electric fields can be achieved by direct, capacitive or inductive coupled setups. We tested the effects of electrical stimulation provided by capacitive coupling on glioma cells, using a capacitive-coupled system powered by a potential difference of 35 V between two electrodes placed outside the culture dish. Numerical dosimetry identified two different fields: (i) in the order of 103 V/m at the level of the dielectric substrates, with almost uniform distribution; (ii) in the order of 10-1 V/m at the level of the culture medium, with spatial and material-dependent distribution. The scratch assay and the tracking of single-cell movement showed a boosted motility when crawling occurs on polystyrene surfaces, demonstrating the feasibility of this peculiar exposure system to generate forces capable of influencing cell behavior.
Collapse
Affiliation(s)
- Isabella Zironi
- Department of Physics and Astronomy (DIFA), Alma Mater Studiorum University of Bologna, Viale Berti Pichat 6/2, Bologna, 40127, Italy.
- National Institute for Nuclear Physics (INFN BO), Bologna section, Viale Berti Pichat 6/2, Bologna, 40127, Italy.
| | - Tobias Cramer
- Department of Physics and Astronomy (DIFA), Alma Mater Studiorum University of Bologna, Viale Berti Pichat 6/2, Bologna, 40127, Italy
| | - Alessandro Fuschi
- Department of Physics and Astronomy (DIFA), Alma Mater Studiorum University of Bologna, Viale Berti Pichat 6/2, Bologna, 40127, Italy
| | - Margherita Cioni
- Department of Physics and Astronomy (DIFA), Alma Mater Studiorum University of Bologna, Viale Berti Pichat 6/2, Bologna, 40127, Italy.
| | - Giada Guerra
- Department for Life Quality Studies (QUVI), Alma Mater Studiorum University of Bologna, C.so d'Augusto, 237, Rimini, 47921, Italy
| | - Giacomo Giuliani
- Department of Physics and Astronomy (DIFA), Alma Mater Studiorum University of Bologna, Viale Berti Pichat 6/2, Bologna, 40127, Italy
| | - Maria Calienni
- Centro Laboratori di Didattica Chimica (CILDIC), Alma Mater Studiorum University of Bologna, Via Gobetti 87, Bologna, 40129, Italy
| | - Laura Caramazza
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Via Eudossiana 18, Rome, 00184, Italy
| | - Micaela Liberti
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Via Eudossiana 18, Rome, 00184, Italy
| | - Francesca Apollonio
- BioEM Lab, Department of Information Engineering, Electronics and Telecommunications (DIET), Sapienza University of Rome, Via Eudossiana 18, Rome, 00184, Italy
| | - Daniel Remondini
- Department of Physics and Astronomy (DIFA), Alma Mater Studiorum University of Bologna, Viale Berti Pichat 6/2, Bologna, 40127, Italy
- National Institute for Nuclear Physics (INFN BO), Bologna section, Viale Berti Pichat 6/2, Bologna, 40127, Italy
| | - Gastone Castellani
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Via Massarenti 9, Bologna, 40138, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti, 9, Bologna, 40138, Italy
| |
Collapse
|
9
|
Preetam S, Ghosh A, Mishra R, Pandey A, Roy DS, Rustagi S, Malik S. Electrical stimulation: a novel therapeutic strategy to heal biological wounds. RSC Adv 2024; 14:32142-32173. [PMID: 39399261 PMCID: PMC11467653 DOI: 10.1039/d4ra04258a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Electrical stimulation (ES) has emerged as a powerful therapeutic modality for enhancing biological wound healing. This non-invasive technique utilizes low-level electrical currents to promote tissue regeneration and expedite the wound healing process. ES has been shown to accelerate wound closure, reduce inflammation, enhance angiogenesis, and modulate cell migration and proliferation through various mechanisms. The principle goal of wound management is the rapid recovery of the anatomical continuity of the skin, to prevent infections from the external environment and maintain homeostasis conditions inside. ES at the wound site is a compelling strategy for skin wound repair. Several ES applications are described in medical literature like AC, DC, and PC to improve cutaneous perfusion and accelerate wound healing. This review aimed to evaluate the primary factors and provides an overview of the potential benefits and mechanisms of ES in wound healing, and its ability to stimulate cellular responses, promote tissue regeneration, and improve overall healing outcomes. We also shed light on the application of ES which holds excellent promise as an adjunct therapy for various types of wounds, including chronic wounds, diabetic ulcers, and surgical incisions.
Collapse
Affiliation(s)
- Subham Preetam
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu 42988 Republic of Korea
| | - Arka Ghosh
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology Bhubaneswar 751003 Odisha India
| | - Richa Mishra
- Department of Computer Engineering, Parul Institute of Engineering and Technology (PIET), Parul University Ta. Waghodia Vadodara Gujarat 391760 India
| | - Arunima Pandey
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology Bhubaneswar 751003 Odisha India
| | - Debanjan Singha Roy
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology Bhubaneswar 751003 Odisha India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University 22 Dehradun Uttarakhand India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand Ranchi Jharkhand 834001 India
- Department of Biotechnology, University Center for Research & Development (UCRD) Chandigarh University Ludhiana Highway Mohali 140413 Punjab India
| |
Collapse
|
10
|
Horrocks MS, Zhurenkov KE, Malmström J. Conducting polymer hydrogels for biomedical application: Current status and outstanding challenges. APL Bioeng 2024; 8:031503. [PMID: 39323539 PMCID: PMC11424142 DOI: 10.1063/5.0218251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
Conducting polymer hydrogels (CPHs) are composite polymeric materials with unique properties that combine the electrical capabilities of conducting polymers (CPs) with the excellent mechanical properties and biocompatibility of traditional hydrogels. This review aims to highlight how the unique properties CPHs have from combining their two constituent materials are utilized within the biomedical field. First, the synthesis approaches and applications of non-CPH conductive hydrogels are discussed briefly, contrasting CPH-based systems. The synthesis routes of hydrogels, CPs, and CPHs are then discussed. This review also provides a comprehensive overview of the recent advancements and applications of CPHs in the biomedical field, encompassing their applications as biosensors, drug delivery scaffolds (DDSs), and tissue engineering platforms. Regarding their applications within tissue engineering, a comprehensive discussion of the usage of CPHs for skeletal muscle prosthetics and regeneration, cardiac regeneration, epithelial regeneration and wound healing, bone and cartilage regeneration, and neural prosthetics and regeneration is provided. Finally, critical challenges and future perspectives are also addressed, emphasizing the need for continued research; however, this fascinating class of materials holds promise within the vastly evolving field of biomedicine.
Collapse
|
11
|
Shlapakova LE, Surmeneva MA, Kholkin AL, Surmenev RA. Revealing an important role of piezoelectric polymers in nervous-tissue regeneration: A review. Mater Today Bio 2024; 25:100950. [PMID: 38318479 PMCID: PMC10840125 DOI: 10.1016/j.mtbio.2024.100950] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Nerve injuries pose a drastic threat to nerve mobility and sensitivity and lead to permanent dysfunction due to low regenerative capacity of mature neurons. The electrical stimuli that can be provided by electroactive materials are some of the most effective tools for the formation of soft tissues, including nerves. Electric output can provide a distinctly favorable bioelectrical microenvironment, which is especially relevant for the nervous system. Piezoelectric biomaterials have attracted attention in the field of neural tissue engineering owing to their biocompatibility and ability to generate piezoelectric surface charges. In this review, an outlook of the most recent achievements in the field of piezoelectric biomaterials is described with an emphasis on piezoelectric polymers for neural tissue engineering. First, general recommendations for the design of an optimal nerve scaffold are discussed. Then, specific mechanisms determining nerve regeneration via piezoelectric stimulation are considered. Activation of piezoelectric responses via natural body movements, ultrasound, and magnetic fillers is also examined. The use of magnetoelectric materials in combination with alternating magnetic fields is thought to be the most promising due to controllable reproducible cyclic deformations and deep tissue permeation by magnetic fields without tissue heating. In vitro and in vivo applications of nerve guidance scaffolds and conduits made of various piezopolymers are reviewed too. Finally, challenges and prospective research directions regarding piezoelectric biomaterials promoting nerve regeneration are discussed. Thus, the most relevant scientific findings and strategies in neural tissue engineering are described here, and this review may serve as a guideline both for researchers and clinicians.
Collapse
Affiliation(s)
- Lada E. Shlapakova
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
| | - Maria A. Surmeneva
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| | - Andrei L. Kholkin
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
- Department of Physics & CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Roman A. Surmenev
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| |
Collapse
|
12
|
Ranjbar N, Bakhshandeh B, Pennisi CP. Electroconductive Nanofibrous Scaffolds Enable Neuronal Differentiation in Response to Electrical Stimulation without Exogenous Inducing Factors. Bioengineering (Basel) 2023; 10:1438. [PMID: 38136029 PMCID: PMC10740536 DOI: 10.3390/bioengineering10121438] [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: 11/08/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Among the various biochemical and biophysical inducers for neural regeneration, electrical stimulation (ES) has recently attracted considerable attention as an efficient means to induce neuronal differentiation in tissue engineering approaches. The aim of this in vitro study was to develop a nanofibrous scaffold that enables ES-mediated neuronal differentiation in the absence of exogenous soluble inducers. A nanofibrous scaffold composed of polycaprolactone (PCL), poly-L-lactic acid (PLLA), and single-walled nanotubes (SWNTs) was fabricated via electrospinning and its physicochemical properties were investigated. The cytocompatibility of the electrospun composite with the PC12 cell line and bone marrow-derived mesenchymal stem cells (BMSCs) was investigated. The results showed that the PCL/PLLA/SWNT nanofibrous scaffold did not exhibit cytotoxicity and supported cell attachment, spreading, and proliferation. ES was applied to cells cultured on the nanofibrous scaffolds at different intensities and the expression of the three neural markers (Nestin, Microtubule-associated protein 2, and β tubulin-3) was evaluated using RT-qPCR analysis. The results showed that the highest expression of neural markers could be achieved at an electric field intensity of 200 mV/cm, suggesting that the scaffold in combination with ES can be an efficient tool to accelerate neural differentiation in the absence of exogenous soluble inducers. This has important implications for the regeneration of nerve injuries and may provide insights for further investigations of the mechanisms underlying ES-mediated neuronal commitment.
Collapse
Affiliation(s)
- Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran 14155-6455, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran 14155-6455, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, DK-9260 Gistrup, Denmark
| |
Collapse
|
13
|
Nwogbaga I, Camley BA. Coupling cell shape and velocity leads to oscillation and circling in keratocyte galvanotaxis. Biophys J 2023; 122:130-142. [PMID: 36397670 PMCID: PMC9822803 DOI: 10.1016/j.bpj.2022.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/03/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
During wound healing, fish keratocyte cells undergo galvanotaxis where they follow a wound-induced electric field. In addition to their stereotypical persistent motion, keratocytes can develop circular motion without a field or oscillate while crawling in the field direction. We developed a coarse-grained phenomenological model that captures these keratocyte behaviors. We fit this model to experimental data on keratocyte response to an electric field being turned on. A critical element of our model is a tendency for cells to turn toward their long axis, arising from a coupling between cell shape and velocity, which gives rise to oscillatory and circular motion. Galvanotaxis is influenced not only by the field-dependent responses, but also cell speed and cell shape relaxation rate. When the cell reacts to an electric field being turned on, our model predicts that stiff, slow cells react slowly but follow the signal reliably. Cells that polarize and align to the field at a faster rate react more quickly and follow the signal more reliably. When cells are exposed to a field that switches direction rapidly, cells follow the average of field directions, while if the field is switched more slowly, cells follow a "staircase" pattern. Our study indicated that a simple phenomenological model coupling cell speed and shape is sufficient to reproduce a broad variety of different keratocyte behaviors, ranging from circling to oscillation to galvanotactic response, by only varying a few parameters.
Collapse
Affiliation(s)
- Ifunanya Nwogbaga
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland
| | - Brian A Camley
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland; William H. Miller III Department of Physics & Astronomy, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
14
|
Yang Y, Luo R, Chao S, Xue J, Jiang D, Feng YH, Guo XD, Luo D, Zhang J, Li Z, Wang ZL. Improved pharmacodynamics of epidermal growth factor via microneedles-based self-powered transcutaneous electrical stimulation. Nat Commun 2022; 13:6908. [PMID: 36376334 PMCID: PMC9663450 DOI: 10.1038/s41467-022-34716-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022] Open
Abstract
Epidermal growth factor is an excellent drug for promoting wound healing; however, its conventional administration strategies are associated with pharmacodynamic challenges, such as low transdermal permeability, reduction, and receptor desensitization. Here, we develop a microneedle-based self-powered transcutaneous electrical stimulation system (mn-STESS) by integrating a sliding free-standing triboelectric nanogenerator with a microneedle patch to achieve improved epidermal growth factor pharmacodynamics. We show that the mn-STESS facilitates drug penetration and utilization by using microneedles to pierce the stratum corneum. More importantly, we find that it converts the mechanical energy of finger sliding into electricity and mediates transcutaneous electrical stimulation through microneedles. We demonstrate that the electrical stimulation applied by mn-STESS acts as an "adjuvant" that suppresses the reduction of epidermal growth factor by glutathione and upregulates its receptor expression in keratinocyte cells, successfully compensating for receptor desensitization. Collectively, this work highlights the promise of self-powered electrical adjuvants in improving drug pharmacodynamics, creating combinatorial therapeutic strategies for traditional drugs.
Collapse
Affiliation(s)
- Yuan Yang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruizeng Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengyu Chao
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiangtao Xue
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, China
| | - Dongjie Jiang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yun Hao Feng
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xin Dong Guo
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Dan Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China.
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Center of Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning, 530004, China.
| | - Jiaping Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China.
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Center of Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning, 530004, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zhong Lin Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- Georgia Institute of Technology, Atlanta, GA, 30332 0245, USA
| |
Collapse
|
15
|
Bierman-Duquette RD, Safarians G, Huang J, Rajput B, Chen JY, Wang ZZ, Seidlits SK. Engineering Tissues of the Central Nervous System: Interfacing Conductive Biomaterials with Neural Stem/Progenitor Cells. Adv Healthc Mater 2022; 11:e2101577. [PMID: 34808031 PMCID: PMC8986557 DOI: 10.1002/adhm.202101577] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/31/2021] [Indexed: 12/19/2022]
Abstract
Conductive biomaterials provide an important control for engineering neural tissues, where electrical stimulation can potentially direct neural stem/progenitor cell (NS/PC) maturation into functional neuronal networks. It is anticipated that stem cell-based therapies to repair damaged central nervous system (CNS) tissues and ex vivo, "tissue chip" models of the CNS and its pathologies will each benefit from the development of biocompatible, biodegradable, and conductive biomaterials. Here, technological advances in conductive biomaterials are reviewed over the past two decades that may facilitate the development of engineered tissues with integrated physiological and electrical functionalities. First, one briefly introduces NS/PCs of the CNS. Then, the significance of incorporating microenvironmental cues, to which NS/PCs are naturally programmed to respond, into biomaterial scaffolds is discussed with a focus on electrical cues. Next, practical design considerations for conductive biomaterials are discussed followed by a review of studies evaluating how conductive biomaterials can be engineered to control NS/PC behavior by mimicking specific functionalities in the CNS microenvironment. Finally, steps researchers can take to move NS/PC-interfacing, conductive materials closer to clinical translation are discussed.
Collapse
Affiliation(s)
| | - Gevick Safarians
- Department of Bioengineering, University of California Los Angeles, USA
| | - Joyce Huang
- Department of Bioengineering, University of California Los Angeles, USA
| | - Bushra Rajput
- Department of Bioengineering, University of California Los Angeles, USA
| | - Jessica Y. Chen
- Department of Bioengineering, University of California Los Angeles, USA
- David Geffen School of Medicine, University of California Los Angeles, USA
| | - Ze Zhong Wang
- Department of Bioengineering, University of California Los Angeles, USA
| | | |
Collapse
|
16
|
Oviedo-Rouco S, Spedalieri C, Scocozza MF, Tomasina F, Tórtora V, Radi R, Murgida DH. Correlated electric field modulation of electron transfer parameters and the access to alternative conformations of multifunctional cytochrome c. Bioelectrochemistry 2022; 143:107956. [PMID: 34624727 DOI: 10.1016/j.bioelechem.2021.107956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 11/21/2022]
Abstract
Cytochrome c (Cytc) is a multifunctional protein that, in its native conformation, shuttles electrons in the mitochondrial respiratory chain. Conformational transitions that involve replacement of the heme distal ligand lead to the gain of alternative peroxidase activity, which is crucial for membrane permeabilization during apoptosis. Using a time-resolved SERR spectroelectrochemical approach, we found that the key physicochemical parameters that characterize the electron transfer (ET) canonic function and those that determine the transition to alternative conformations are strongly correlated and are modulated by local electric fields (LEF) of biologically meaningful magnitude. The electron shuttling function is optimized at moderate LEFs of around 1 V nm-1. A decrease of the LEF is detrimental for ET as it rises the reorganization energy. Moreover, LEF values below and above the optimal for ET favor alternative conformations with peroxidase activity and downshifted reduction potentials. The underlying proposed mechanism is the LEF modulation of the flexibility of crucial protein segments, which produces a differential effect on the kinetic ET and conformational parameters of Cytc. These findings might be related to variations in the mitochondrial membrane potential during apoptosis, as the basis for the switch between canonic and alternative functions of Cytc. Moreover, they highlight the possible role of variable LEFs in determining the function of other moonlighting proteins through modulation of the protein dynamics.
Collapse
Affiliation(s)
- Santiago Oviedo-Rouco
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cecilia Spedalieri
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Magalí F Scocozza
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Florencia Tomasina
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Verónica Tórtora
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Daniel H Murgida
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
17
|
Farber PL, Isoldi FC, Ferreira LM. Electric Factors in Wound Healing. Adv Wound Care (New Rochelle) 2021; 10:461-476. [PMID: 32870772 DOI: 10.1089/wound.2019.1114] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Significance: Electric factors such as electric charges, electrodynamic field, skin battery, and interstitial exclusion permeate wound healing physiology and physiopathology from injury to re-epithelialization. The understanding of how electric factors contribute to wound healing and how treatments may interfere with them is fundamental for the development of better strategies for the management of pathological scarring and chronic wounds. Recent Advances: Angiogenesis, cell migration, macrophage activation hemorheology, and microcirculation can interfere and be interfered with electric factors. New treatments with various types of electric currents, laser, light emitting diode, acupuncture, and weak electric fields applied directly on the wound have been developed to improve wound healing. Critical Issues: Despite the basic and clinical development, pathological scars such as keloids and chronic wounds are still a challenge. Future Directions: New treatments can be developed to improve skin wound healing taking into account the influence of electrical charges. Monitoring electrical activity during skin healing and the influence of treatments on hemorheology and microcirculation are examples of how to use knowledge of electrical factors to increase their effectiveness.
Collapse
Affiliation(s)
| | - Felipe Contoli Isoldi
- Surgery Department, Plastic Surgery Division, Postgraduated Program in Translational Surgery, Universidade Federal de São Paulo (Unifesp), São Paulo, Brazil
| | - Lydia Masako Ferreira
- Surgery Department, Plastic Surgery Division, Postgraduated Program in Translational Surgery, Universidade Federal de São Paulo (Unifesp), São Paulo, Brazil
| |
Collapse
|
18
|
Kosevic D, Wiedemann D, Vukovic P, Ristic V, Riebandt J, Radak U, Brandes K, Goettel P, Duengen H, Tahirovic E, Kottmann T, Voss HW, Zdravkovic M, Putnik S, Schmitto JD, Mueller J, Rame JE, Peric M. Cardio-microcurrent device for chronic heart failure: first-in-human clinical study. ESC Heart Fail 2021; 8:962-970. [PMID: 33559358 PMCID: PMC8006737 DOI: 10.1002/ehf2.13242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/05/2021] [Accepted: 01/19/2021] [Indexed: 12/29/2022] Open
Abstract
AIMS Most devices for treating ambulatory Class II and III heart failure are linked to electrical pulses. However, a steady electric potential gradient is also necessary for appropriate myocardial performance and may be disturbed by structural heart diseases. We investigated whether chronic application of electrical microcurrent to the heart is feasible and safe and improves cardiac performance. The results of this study should provide guidance for the design of a two-arm, randomized, controlled Phase II trial. METHODS AND RESULTS This single-arm, non-randomized pilot study involved 10 patients (9 men; mean age, 62 ± 12 years) at two sites with 6 month follow-up. All patients had New York Heart Association (NYHA) Class III heart failure and non-ischaemic dilated cardiomyopathy, with left ventricular ejection fraction (LVEF) <35%. A device was surgically placed to deliver a constant microcurrent to the heart. The following tests were performed at baseline, at hospital discharge, and at six time points during follow-up: determination of LVEF and left ventricular end-diastolic/end-systolic diameter by echocardiography; the 6 min walk test; and assessment of NYHA classification and quality of life (36-Item Short-Form Health Survey questionnaire). Microcurrent application was feasible and safe; no device-related or treatment-related adverse events occurred. During follow-up, rapid and significant signal of efficacy (P < 0.005) was present with improvements in LVEF, left ventricular end-diastolic diameter, left ventricular end-systolic diameter, and distance walked. For eight patients, NYHA classification improved from Class III to Class I (for seven, as early as 14 days post-operatively); for one, to Class II; and for one, to Class II/III. 36-Item Short-Form Health Survey questionnaire scores also improved highly significantly. CONCLUSIONS Chronic application of microcurrent to the heart is feasible and safe and leads to a rapid and lasting improvement in heart function and a near normalization of heart size within days. The NYHA classification and quality of life improve just as rapidly.
Collapse
Affiliation(s)
| | | | - Petar Vukovic
- Cardiovascular Institute Dedinje BelgradeBelgradeSerbia
| | | | | | - Una Radak
- Cardiovascular Institute Dedinje BelgradeBelgradeSerbia
| | | | | | | | | | | | | | - Marija Zdravkovic
- University Hospital Medical Center Bezanijska Kosa BelgradeBelgradeSerbia
| | | | | | | | - Jesus Eduardo Rame
- Advanced Cardiac and Pulmonary Vascular Disease ProgramsJefferson Heart InstitutePhiladelphiaPAUSA
| | - Miodrag Peric
- Cardiovascular Institute Dedinje BelgradeBelgradeSerbia
| |
Collapse
|
19
|
Jia N, Liu J, Zhu G, Liang Y, Wang Y, Wang W, Chen Y, Yang J, Zhang W, Zhang J. Polarization of ADAM17-driven EGFR signalling in electric field-guided collective migration of epidermal sheets. J Cell Mol Med 2020; 24:14073-14085. [PMID: 33164313 PMCID: PMC7753989 DOI: 10.1111/jcmm.16019] [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: 06/05/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 12/22/2022] Open
Abstract
Endogenous electric field is considered to play an important role in promoting collective migration of epidermis to the wound centre. However, most studies are focused on the effect of bioelectric field on the movement and migration of single epithelial cell; the molecular mechanisms about collective migration of epidermal monolayers remain unclear. Here, we found that EFs dramatically promoted the collective migration of HaCaT cells towards the anode, activated the sheddase activity of ADAM17 and increased the phosphorylation level of EGFR. Moreover, EGFR phosphorylation and HB-EGF shedding level were significantly decreased by the ADAM17 inhibitor TAPI-2 or siADAM17 under EFs, which subsequently attenuated the directed migration of HaCaT sheets. Notably, the inhibition of EF-regulated collective migration by siADAM17 was rescued by addition of recombinant HB-EGF. Furthermore, we observed that F-actin was dynamically polarized along the leading edge of the migrated sheets under EFs and that this polarization was regulated by ADAM17/HB-EGF/EGFR signalling. In conclusion, our study indicated that ADAM17 contributed to the collective directional movement of the epidermal monolayer by driving HB-EGF release and activating EGFR under EFs, and this pathway also mediated the polarization of F-actin in migrating sheets, which is essential in directional migration.
Collapse
Affiliation(s)
- Naixin Jia
- Key Laboratory of Freshwater Fish Reproduction and DevelopmentMinistry of EducationLaboratory of Molecular Developmental BiologySchool of Life SciencesSouthwest UniversityChongqingChina
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| | - Jie Liu
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| | - Guoqin Zhu
- Key Laboratory of Freshwater Fish Reproduction and DevelopmentMinistry of EducationLaboratory of Molecular Developmental BiologySchool of Life SciencesSouthwest UniversityChongqingChina
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| | - Yi Liang
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| | - Yuan Wang
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| | - Weiyi Wang
- Dalian Rehabilitation Recuperation Center of PLA Joint Logistics Support ForceDalianChina
| | - Ying Chen
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| | - Jinrui Yang
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| | - Wangjun Zhang
- Department of Pancreatic and Biliary SurgeryThe First Affiliated Hospital of Harbin Medical UniversityHeilongjiangChina
| | - Jiaping Zhang
- Department of Plastic and Aesthetic SurgeryState Key Laboratory of Trauma, Burns and Combined InjurySouthwest HospitalThe Third Military Medical University(Army Medical University)ChongqingChina
| |
Collapse
|
20
|
Li L, Hu C, Lu C, Zhang K, Han R, Lin C, Zhao S, A C, Cheng C, Zhao M, He Y. Applied electric fields suppress osimertinib-induced cytotoxicity via inhibiting FOXO3a nuclear translocation through AKT activation. Carcinogenesis 2020; 41:600-610. [PMID: 31504249 DOI: 10.1093/carcin/bgz150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/16/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
Abstract
Osimertinib is a third-generation epidermal growth factor receptor tyrosine kinase inhibitor against T790M-mutant non-small cell lung cancer (NSCLC). Acquired resistance to osimertinib is a growing clinical challenge that is not fully understood. Endogenous electric fields (EFs), components of the tumor microenvironment, are associated with cancer cell migration and proliferation. However, the impact of EFs on drug efficiency has not been studied. In this study, we observed that EFs counteracted the effects of osimertinib. EFs of 100 mV/mm suppressed osimertinib-induced cell death and promoted cell proliferation. Transcriptional analysis revealed that the expression pattern induced by osimertinib was altered by EFs stimulation. KEGG analysis showed that differential expression genes were mostly enriched in PI3K-AKT pathway. Then, we found that osimertinib inhibited AKT phosphorylation, while EFs stimulation resulted in significant activation of AKT, which could override the effects generated by osimertinib. Importantly, pharmacological inhibition of PI3K/AKT by LY294002 diminished EF-induced activation of AKT and restored the cytotoxicity of osimertinib suppressed by EFs, which proved that AKT activation was essential for EFs to attenuate the efficacy of osimertinib. Furthermore, activation of AKT by EFs led to phosphorylation of forkhead box O3a (FOXO3a), and reduction in nuclear translocation of FOXO3a induced by osimertinib, resulting in decreased expression of Bim and attenuated cytotoxicity of osimertinib. Taken together, we demonstrated that EFs suppressed the antitumor activity of osimertinib through AKT/FOXO3a/Bim pathway, and combination of PI3K/AKT inhibitor with osimertinib counteracted the effects of EFs. Our findings provided preliminary data for therapeutic strategies to enhance osimertinib efficacy in NSCLC patients.
Collapse
Affiliation(s)
- Li Li
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Chen Hu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Kejun Zhang
- Department of Clinical Laboratory, Daping Hospital, Army Medical University, Chongqing, China
| | - Rui Han
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Sanjun Zhao
- School of Life Sciences, Yunnan Normal University, Kunming, China
| | - Chunxian A
- School of Life Sciences, Yunnan Normal University, Kunming, China
| | | | - Min Zhao
- Department of Dermatology, Department of Ophthalmology, Institute for Regenerative Cures, University of California, Davis, CA, USA
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
21
|
Zarrintaj P, Zangene E, Manouchehri S, Amirabad LM, Baheiraei N, Hadjighasem MR, Farokhi M, Ganjali MR, Walker BW, Saeb MR, Mozafari M, Thomas S, Annabi N. Conductive biomaterials as nerve conduits: Recent advances and future challenges. APPLIED MATERIALS TODAY 2020; 20:100784. [DOI: 10.1016/j.apmt.2020.100784] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
|
22
|
All Roads Lead to Directional Cell Migration. Trends Cell Biol 2020; 30:852-868. [PMID: 32873438 DOI: 10.1016/j.tcb.2020.08.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 01/17/2023]
Abstract
Directional cell migration normally relies on a variety of external signals, such as chemical, mechanical, or electrical, which instruct cells in which direction to move. Many of the major molecular and physical effects derived from these cues are now understood, leading to questions about whether directional cell migration is alike or distinct under these different signals, and how cells might be directed by multiple simultaneous cues, which would be expected in complex in vivo environments. In this review, we compare how different stimuli are spatially distributed, often as gradients, to direct cell movement and the mechanisms by which they steer cells. A comparison of the downstream effectors of directional cues suggests that different external signals regulate a common set of components: small GTPases and the actin cytoskeleton, which implies that the mechanisms downstream of different signals are likely to be closely related and underlies the idea that cell migration operates by a common set of physical principles, irrespective of the input.
Collapse
|
23
|
Cellular processes involved in lung cancer cells exposed to direct current electric field. Sci Rep 2020; 10:5289. [PMID: 32210363 PMCID: PMC7093422 DOI: 10.1038/s41598-020-62332-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/06/2020] [Indexed: 11/08/2022] Open
Abstract
With the rapid breakthrough of electrochemical treatment of tumors, electric field (EF)-sensitive genes, previously rarely exploited, have become an emerging field recently. Here, we reported our work for the identification of EF-sensitive genes in lung cancer cells. The gene expression profile (GSE33845), in which the human lung cancer CL1-0 cells were treated with a direct current electric field (dcEF) (300 mV/mm) for 2 h, was retrieved from GEO database. Differentially expressed genes (DEGs) were acquired, followed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) and protein-protein interaction (PPI) analysis. Hub genes were acquired and analyzed by various tools including the Human Protein Atlas, Kaplan-Meier analysis, Cytoscape, FunRich, Oncomine and cBioPortal. Subsequently, three-dimensional protein models of hub genes were modeled by Modeller 9.20 and Rosetta 3.9. Finally, a 100 ns molecular dynamics simulation for each hub protein was performed with GROMACS 2018.2. A total of 257 DEGs were acquired and analyzed by GO, KEGG and PPI. Then, 10 hub genes were obtained, and the signal pathway analysis showed that two inflammatory pathways were activated: the FoxO signaling pathway and the AGE-RAGE signaling pathway. The molecular dynamic analysis including RMSD and the radius of gyration hinted that the 3D structures of hub proteins were built. Overall, our work identified EF-sensitive genes in lung cancer cells and identified that the inflammatory state of tumor cells may be involved in the feedback mechanism of lung cancer cells in response to electric field stimulation. In addition, qualified three-dimensional protein models of hub genes were also constructed, which will be helpful in understanding the complex effects of dcEF on human lung cancer CL1-0 cells.
Collapse
|
24
|
Chen C, Bai X, Ding Y, Lee IS. Electrical stimulation as a novel tool for regulating cell behavior in tissue engineering. Biomater Res 2019; 23:25. [PMID: 31844552 PMCID: PMC6896676 DOI: 10.1186/s40824-019-0176-8] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
Recently, electrical stimulation as a physical stimulus draws lots of attention. It shows great potential in disease treatment, wound healing, and mechanism study because of significant experimental performance. Electrical stimulation can activate many intracellular signaling pathways, and influence intracellular microenvironment, as a result, affect cell migration, cell proliferation, and cell differentiation. Electrical stimulation is using in tissue engineering as a novel type of tool in regeneration medicine. Besides, with the advantages of biocompatible conductive materials coming into view, the combination of electrical stimulation with suitable tissue engineered scaffolds can well combine the benefits of both and is ideal for the field of regenerative medicine. In this review, we summarize the various materials and latest technologies to deliver electrical stimulation. The influences of electrical stimulation on cell alignment, migration and its underlying mechanisms are discussed. Then the effect of electrical stimulation on cell proliferation and differentiation are also discussed.
Collapse
Affiliation(s)
- Cen Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018 People’s Republic of China
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, 310018 People’s Republic of China
| | - Xue Bai
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018 People’s Republic of China
| | - Yahui Ding
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, 310014 People’s Republic of China
- People’s Hospital of Hangzhou Medical College, Hangzhou, 310014 People’s Republic of China
| | - In-Seop Lee
- Institute of Natural Sciences, Yonsei University, 134 Shinchon-dong, Seodaemoon-gu, Seoul, 03722 Republic of Korea
| |
Collapse
|
25
|
Jeon TJ, Gao R, Kim H, Lee A, Jeon P, Devreotes PN, Zhao M. Cell migration directionality and speed are independently regulated by RasG and Gβ in Dictyostelium cells in electrotaxis. Biol Open 2019; 8:bio.042457. [PMID: 31221628 PMCID: PMC6679393 DOI: 10.1242/bio.042457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Motile cells manifest increased migration speed and directionality in gradients of stimuli, including chemoattractants, electrical potential and substratum stiffness. Here, we demonstrate that Dictyostelium cells move directionally in response to an electric field (EF) with specific acceleration/deceleration kinetics of directionality and migration speed. Detailed analyses of the migration kinetics suggest that migration speed and directionality are separately regulated by Gβ and RasG, respectively, in EF-directed cell migration. Cells lacking Gβ, which is essential for all chemotactic responses in Dictyostelium, showed EF-directed cell migration with the same increase in directionality in an EF as wild-type cells. However, these cells failed to show induction of the migration speed upon EF stimulation as much as wild-type cells. Loss of RasG, a key regulator of chemoattractant-directed cell migration, resulted in almost complete loss of directionality, but similar acceleration/deceleration kinetics of migration speed as wild-type cells. These results indicate that Gβ and RasG are required for the induction of migration speed and directionality, respectively, in response to an EF, suggesting separation of migration speed and directionality even with intact feedback loops between mechanical and signaling networks. Summary: Cell migration directionality and speed are independently regulated by RasG and Gβ, respectively, in electric field-directed cell migration in Dictyostelium, suggesting the points of molecular divergence of the two characteristics.
Collapse
Affiliation(s)
- Taeck J Jeon
- Department of Biology & BK21-Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 61452, Republic of Korea
| | - Runchi Gao
- School of life science, Yunnan Normal University, Kunming, Yunnan 650500, China
| | - Hyeseon Kim
- Department of Biology & BK21-Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 61452, Republic of Korea
| | - Ara Lee
- Department of Biology & BK21-Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 61452, Republic of Korea
| | - Pyeonghwa Jeon
- Department of Biology & BK21-Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 61452, Republic of Korea
| | - Peter N Devreotes
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Min Zhao
- Departments of Dermatology and Ophthalmology, Institute for Regenerative Cures, School of Medicine, University of California at Davis, CA 95817, USA
| |
Collapse
|
26
|
Electrotaxis of Glioblastoma and Medulloblastoma Spheroidal Aggregates. Sci Rep 2019; 9:5309. [PMID: 30926929 PMCID: PMC6441013 DOI: 10.1038/s41598-019-41505-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 02/26/2019] [Indexed: 11/08/2022] Open
Abstract
Treatment of neuroepithelial cancers remains a daunting clinical challenge, particularly due to an inability to address rampant invasion deep into eloquent regions of the brain. Given the lack of access, and the dispersed nature of brain tumor cells, we explore the possibility of electric fields inducing directed tumor cell migration. In this study we investigate the properties of populations of brain cancer undergoing electrotaxis, a phenomenon whereby cells are directed to migrate under control of an electrical field. We investigate two cell lines for glioblastoma and medulloblastoma (U87mg & DAOY, respectively), plated as spheroidal aggregates in Matrigel-filled electrotaxis channels, and report opposing electrotactic responses. To further understand electrotactic migration of tumor cells, we performed RNA-sequencing for pathway discovery to identify signaling that is differentially affected by the exposure of direct-current electrical fields. Further, using selective pharmacological inhibition assays, focused on the PI3K/mTOR/AKT signaling axis, we validate whether there is a causal relationship to electrotaxis and these mechanisms of action. We find that U87 mg electrotaxis is abolished under pharmacological inhibition of PI3Kγ, mTOR, AKT and ErbB2 signaling, whereas DAOY cell electrotaxis was not attenuated by these or other pathways evaluated.
Collapse
|
27
|
Dong ZY, Pei Z, Wang YL, Li Z, Khan A, Meng XT. Ascl1 Regulates Electric Field-Induced Neuronal Differentiation Through PI3K/Akt Pathway. Neuroscience 2019; 404:141-152. [PMID: 30771509 DOI: 10.1016/j.neuroscience.2019.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/14/2022]
Abstract
Directing differentiation of neural stem/progenitor cells (NSCs/NPCs) to produce functional neurons is one of the greatest challenges in regenerative medicine. Our previous paper has confirmed that electrical stimulation has a high efficiency of triggering neuronal differentiation by using isolated filum terminale (FT)-derived NPCs. To further clarify the intrinsic molecular mechanisms, protein-protein interaction (PPI) network analysis was applied to pinpoints novel hubs in electric field (EF)-induced neuronal differentiation. In this study, siRNA transfection of Achaete-scute homolog 1 (Ascl1) in NPCs or NPCs was followed by direct current stimulation at 150 mV/mm. Neuronal differentiation rate and protein expression level were analyzed after 7 or 14 days of electrical stimulation. The data showed that the expression level of Ascl1 was enhanced by electrical stimulation and positively correlated to EF strength. Moreover, we identified that the expression of Ascl1 positively regulated neuronal differentiation of NPCs and can be up-regulated by EF-stimulation through the activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway. Therefore, this study provides new insights into the role of Ascl1 and its relevant PI3K/Akt pathway in regulating of EF-induced neuronal differentiation and pointed out that continuous expression of Ascl1 in NPCs is required for EF-induced neuronal differentiation.
Collapse
Affiliation(s)
- Zhi-Yong Dong
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Pei
- Department of Neuroscience and Pediatric, GSRB1 Duke University, Durham 27710, USA
| | - Yan-Ling Wang
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Li
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Amber Khan
- The Graduate Center and CUNY School of Medicine, CUNY, 85 St Nicholas Terrace, New York, NY 10027, USA.
| | - Xiao-Ting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
28
|
Electric field-responsive nanoparticles and electric fields: physical, chemical, biological mechanisms and therapeutic prospects. Adv Drug Deliv Rev 2019; 138:56-67. [PMID: 30414494 DOI: 10.1016/j.addr.2018.10.017] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/05/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022]
Abstract
Electric fields are among physical stimuli that have revolutionized therapy. Occurring endogenously or exogenously, the electric field can be used as a trigger for controlled drug release from electroresponsive drug delivery systems, can stimulate wound healing and cell proliferation, may enhance endocytosis or guide stem cell differentiation. Electric field pulses may be applied to induce cell fusion, can increase the penetration of therapeutic agents into cells, or can be applied as a standalone therapy to ablate tumors. This review describes the main therapeutic trends and overviews the main physical, chemical and biological mechanisms underlying the actions of electric fields. Overall, the electric field can be used in therapeutic approaches in several ways. The electric field can act on drug carriers, cells and tissues. Understanding the multiple effects of this powerful tool will help harnessing its full therapeutic potential in an efficient and safe way.
Collapse
|
29
|
Yan T, Jiang X, Lin G, Tang D, Zhang J, Guo X, Zhang D, Zhang Q, Jia J, Huang Y. Autophagy is required for the directed motility of keratinocytes driven by electric fields. FASEB J 2018; 33:3922-3935. [PMID: 30509146 DOI: 10.1096/fj.201801294r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Endogenous wound electric fields (EFs), an important and fundamental occurrence of wound healing, profoundly influence the directed migration of keratinocytes. Although numerous studies have unveiled the signals responsible for EF-biased direction, the mechanisms by which EFs promote keratinocyte motility remains to be elucidated. In our study, EFs enhanced the directed migratory speed of keratinocytes by inducing autophagic activity, thereby facilitating skin barrier restoration. Initially, we found that electrical signals directed keratinocytes to the cathode with enhanced motility parameters [ i.e., trajectory distance, trajectory speed, displacement distance, and displacement speed ( Td/ t)] and more efficient migration (directionality and Td/ t along the x axis, among others). Meanwhile, EFs induced a time-dependent increase in autophagic activity in keratinocytes, with constant autophagic flux, accompanied by increased transcription of numerous autophagy-related genes. Deficiency in Atg5, a key protein necessary for autophagosome formation, led to significant reduction of autophagy, which was accompanied by a substantial reduction in EF-stimulated directed motility. These results demonstrated a causal relationship between autophagy and EF-directed migratory speed. In addition, both cell migration under normal conditions and EF-biased directionality were autophagy independent. Thus, our findings define autophagy as an important functional regulator of electrically enhanced directed motility, adding to a growing understanding of EFs.-Yan, T., Jiang, X., Lin, G., Tang, D., Zhang, J., Guo, X., Zhang, D., Zhang, Q., Jia, J., Huang, Y. Autophagy is required for the directed motility of keratinocytes driven by electric fields.
Collapse
Affiliation(s)
- Tiantian Yan
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Military Burn Center, the 990th (159th) Hospital of the People's Liberation Army, Zhumadian, China
| | - Xupin Jiang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guoan Lin
- Military Burn Center, the 990th (159th) Hospital of the People's Liberation Army, Zhumadian, China
| | - Di Tang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junhui Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaowei Guo
- Department of Burns and Plastic Surgery, the 205th Hospital of the People's Liberation Army, Jinzhou, China
| | - Dongxia Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiezhi Jia
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
30
|
Yang C, Wang L, Weng W, Wang S, Ma Y, Mao Q, Gao G, Chen R, Feng J. Steered migration and changed morphology of human astrocytes by an applied electric field. Exp Cell Res 2018; 374:282-289. [PMID: 30508512 DOI: 10.1016/j.yexcr.2018.11.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/01/2023]
Abstract
Direct current electric field (DC EF) plays a role in influencing the biological behaviors and functions of cells. We hypothesize that human astrocytes (HAs) could also be influenced in EF. Astrocytes, an important type of nerve cells with a high proportion quantitatively, are generally activated and largely decide the brain repair results after brain injury. So far, no electrotaxis study on HAs has been performed. We here obtained HAs derived from brain trauma patients. After purification and identification, HAs were seeded in the EF chamber and recorded in a time-lapse image system. LY294002 and U0126 were then used to probe the role of PI3K or ERK signaling pathway on cellular behaviors. The results showed that HAs could be guided to migrate to the anode in DC EFs, in a voltage-dependent manner. The HAs displayed elongated cell bodies and reoriented perpendicularly to the EF in morphology. When treated with LY294002 or U0126, alternation of parameters such as cellular verticality, track speed, displacement speed, long axis, vertical length and circularity were inhibited partly as expected, while the EF-induced directedness was not terminated even at a high drug dosage which was not consistent with previous electrotaxis studies. In conclusion, applied EFs steered the patient-derived HAs directional migration and changed morphology, in which PI3K and ERK pathways at least partially participate. The characteristics of HAs to EF stimulation may be involved in wound healing and neural regeneration, which could be utilized as a novel treatment strategy in brain injury.
Collapse
Affiliation(s)
- Chun Yang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China; Shanghai Institute of Head Trauma, Shanghai 200127, People's Republic of China
| | - Lei Wang
- Department of Neurosurgery, the Yuhuangding Hospital, Yantai 264000, People's Republic of China
| | - Weiji Weng
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China; Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, People's Republic of China
| | - Shen Wang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China; Shanghai Institute of Head Trauma, Shanghai 200127, People's Republic of China
| | - Yuxiao Ma
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China; Shanghai Institute of Head Trauma, Shanghai 200127, People's Republic of China
| | - Qing Mao
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China; Shanghai Institute of Head Trauma, Shanghai 200127, People's Republic of China
| | - Guoyi Gao
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China
| | - Rui Chen
- Department of Plastic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China.
| | - Junfeng Feng
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China; Shanghai Institute of Head Trauma, Shanghai 200127, People's Republic of China.
| |
Collapse
|
31
|
Electric Pulses Can Influence Galvanotaxis of Dictyostelium discoideum. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2534625. [PMID: 30186854 PMCID: PMC6112078 DOI: 10.1155/2018/2534625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/02/2018] [Accepted: 07/31/2018] [Indexed: 01/14/2023]
Abstract
Galvanotaxis, or electrotaxis, plays an essential role in wound healing, embryogenesis, and nerve regeneration. Up until now great efforts have been made to identify the underlying mechanism related to galvanotaxis in various cells under direct current electric field (DCEF) in laboratory studies. However, abundant clinical research shows that non-DCEFs including monopolar or bipolar electric field may also contribute to wound healing and regeneration, although the mechanism remains elusive. Here, we designed a novel electric stimulator and applied DCEF, pulsed DCEF (pDCEF), and bipolar pulse electric field (bpEF) to the cells of Dictyostelium discoideum. The cells had better directional performance under asymmetric 90% duty cycle pDCEF and 80% duty cycle bpEF compared to DCEF, with 10 Hz frequency electric fields eliciting a better cell response than 5 Hz. Interestingly, electrically neutral 50% duty cycle bpEF triggered the highest migration speed, albeit in random directions. The results suggest that electric pulses are vital to galvanotaxis and non-DCEF is promising in both basic and clinical researches.
Collapse
|
32
|
Wang X, Ren Y, Liu J. Liquid Metal Enabled Electrobiology: A New Frontier to Tackle Disease Challenges. MICROMACHINES 2018; 9:E360. [PMID: 30424293 PMCID: PMC6082282 DOI: 10.3390/mi9070360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/09/2018] [Accepted: 07/18/2018] [Indexed: 01/06/2023]
Abstract
In this article, a new conceptual biomedical engineering strategy to tackle modern disease challenges, called liquid metal (LM) enabled electrobiology, is proposed. This generalized and simple method is based on the physiological fact that specially administrated electricity induces a series of subsequent desired biological effects, either shortly, transitionally, or permanently. Due to high compliance within biological tissues, LM would help mold a pervasive method for treating physiological or psychological diseases. As highly conductive and non-toxic multifunctional flexible materials, such LMs can generate any requested electric treating fields (ETFields), which can adapt to various sites inside the human body. The basic mechanisms of electrobiology in delivering electricity to the target tissues and then inducing expected outputs for disease treatment are interpreted. The methods for realizing soft and conformable electronics based on LM are illustrated. Furthermore, a group of typical disease challenges are observed to illustrate the basic strategies for performing LM electrobiology therapy, which include but are not limited to: tissue electronics, brain disorder, immunotherapy, neural functional recovery, muscle stimulation, skin rejuvenation, cosmetology and dieting, artificial organs, cardiac pacing, cancer therapy, etc. Some practical issues regarding electrobiology for future disease therapy are discussed. Perspectives in this direction for incubating a simple biomedical tool for health care are pointed out.
Collapse
Affiliation(s)
- Xuelin Wang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Yi Ren
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Jing Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China.
- Beijing Key Lab of CryoBiomedical Engineering and Key Lab of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
33
|
Sella S, Adami V, Amati E, Bernardi M, Chieregato K, Gatto P, Menarin M, Pozzato A, Pozzato G, Astori G. In-vitro analysis of Quantum Molecular Resonance effects on human mesenchymal stromal cells. PLoS One 2018; 13:e0190082. [PMID: 29293552 PMCID: PMC5749755 DOI: 10.1371/journal.pone.0190082] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 12/07/2017] [Indexed: 01/08/2023] Open
Abstract
Electromagnetic fields play an essential role in cellular functions interfering with cellular pathways and tissue physiology. In this context, Quantum Molecular Resonance (QMR) produces waves with a specific form at high-frequencies (4–64 MHz) and low intensity through electric fields. We evaluated the effects of QMR stimulation on bone marrow derived mesenchymal stromal cells (MSC). MSC were treated with QMR for 10 minutes for 4 consecutive days for 2 weeks at different nominal powers. Cell morphology, phenotype, multilineage differentiation, viability and proliferation were investigated. QMR effects were further investigated by cDNA microarray validated by real-time PCR. After 1 and 2 weeks of QMR treatment morphology, phenotype and multilineage differentiation were maintained and no alteration of cellular viability and proliferation were observed between treated MSC samples and controls. cDNA microarray analysis evidenced more transcriptional changes on cells treated at 40 nominal power than 80 ones. The main enrichment lists belonged to development processes, regulation of phosphorylation, regulation of cellular pathways including metabolism, kinase activity and cellular organization. Real-time PCR confirmed significant increased expression of MMP1, PLAT and ARHGAP22 genes while A2M gene showed decreased expression in treated cells compared to controls. Interestingly, differentially regulated MMP1, PLAT and A2M genes are involved in the extracellular matrix (ECM) remodelling through the fibrinolytic system that is also implicated in embryogenesis, wound healing and angiogenesis. In our model QMR-treated MSC maintained unaltered cell phenotype, viability, proliferation and the ability to differentiate into bone, cartilage and adipose tissue. Microarray analysis may suggest an involvement of QMR treatment in angiogenesis and in tissue regeneration probably through ECM remodelling.
Collapse
Affiliation(s)
- Sabrina Sella
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Valentina Adami
- High Throughput Screening Core Facility, Center for Integrative Biology, University of Trento, Trento, Italy
| | - Eliana Amati
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Martina Bernardi
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
- Hematology Project Foundation, Vicenza, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
- Hematology Project Foundation, Vicenza, Italy
| | - Pamela Gatto
- High Throughput Screening Core Facility, Center for Integrative Biology, University of Trento, Trento, Italy
| | - Martina Menarin
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | | | | | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
- * E-mail:
| |
Collapse
|
34
|
Jeong GJ, Oh JY, Kim YJ, Bhang SH, Jang HK, Han J, Yoon JK, Kwon SM, Lee TI, Kim BS. Therapeutic Angiogenesis via Solar Cell-Facilitated Electrical Stimulation. ACS APPLIED MATERIALS & INTERFACES 2017; 9:38344-38355. [PMID: 29043772 DOI: 10.1021/acsami.7b13322] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cell therapy has been suggested as a treatment modality for ischemic diseases, but the poor survival and engraftment of implanted cells limit its therapeutic efficacy. To overcome such limitation, we used electrical stimulation (ES) derived from a wearable solar cell for inducing angiogenesis in ischemic tissue. ES enhanced the secretion of angiogenic growth factors and the migration of mesenchymal stem cells (MSCs), myoblasts, endothelial progenitor cells, and endothelial cells in vitro. In a mouse ischemic hindlimb model, ES generated by a solar cell and applied to the ischemic region promoted migration of MSCs toward the ischemic site and upregulated expression of angiogenic paracrine factors (vascular endothelial, basic fibroblast, and hepatocyte growth factors; and stromal cell-derived factor-1α). Importantly, solar cell-generated ES promoted the formation of capillaries and arterioles at the ischemic region, attenuated muscle necrosis and fibrosis, and eventually prevented loss of the ischemic limb. Solar cell ES therapy showed higher angiogenic efficacy than conventional MSC therapy. This study shows the feasibility of using solar cell ES as a novel treatment for therapeutic angiogenesis.
Collapse
Affiliation(s)
| | - Jin Young Oh
- Department of Materials Science and Engineering, Yonsei University , Seoul 03722, Republic of Korea
| | - Yeon-Ju Kim
- Department of Physiology, School of Medicine, Pusan National University , Yangsan, 50612 Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University , Suwon 16419, Republic of Korea
| | | | | | | | - Sang-Mo Kwon
- Department of Physiology, School of Medicine, Pusan National University , Yangsan, 50612 Republic of Korea
| | - Tae Il Lee
- Department of BioNano Technology, Gachon University , Seongnam 13120, Republic of Korea
| | | |
Collapse
|
35
|
Li Y, Xu T, Chen X, Lin S, Cho M, Sun D, Yang M. Effects of direct current electric fields on lung cancer cell electrotaxis in a PMMA-based microfluidic device. Anal Bioanal Chem 2017; 409:2163-2178. [DOI: 10.1007/s00216-016-0162-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 12/14/2016] [Accepted: 12/16/2016] [Indexed: 11/27/2022]
|
36
|
Shiozaki Y, Segawa H, Ohnishi S, Ohi A, Ito M, Kaneko I, Kido S, Tatsumi S, Miyamoto KI. Relationship between sodium-dependent phosphate transporter (NaPi-IIc) function and cellular vacuole formation in opossum kidney cells. THE JOURNAL OF MEDICAL INVESTIGATION 2017; 62:209-18. [PMID: 26399350 DOI: 10.2152/jmi.62.209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
NaPi-IIc/SLC34A3 is a sodium-dependent inorganic phosphate (Pi) transporter in the renal proximal tubules and its mutations cause hereditary hypophosphatemic rickets with hypercalciuria (HHRH). In the present study, we created a specific antibody for opossum SLC34A3, NaPi-IIc (oNaPi-IIc), and analyzed its localization and regulation in opossum kidney cells (a tissue culture model of proximal tubular cells). Immunoreactive oNaPi-IIc protein levels increased during the proliferative phase and decreased during differentiation. Moreover, stimulating cell growth upregulated oNaPi-IIc protein levels, whereas suppressing cell proliferation downregulated oNaPi-IIc protein levels. Immunocytochemistry revealed that endogenous and exogenous oNaPi-IIc proteins localized at the protrusion of the plasma membrane, which is a phosphatidylinositol 4,5-bisphosphate (PIP2) rich-membrane, and at the intracellular vacuolar membrane. Exogenous NaPi-IIc also induced cellular vacuoles and localized in the plasma membrane. The ability to form vacuoles is specific to electroneutral NaPi-IIc, and not electrogenic NaPi-IIa or NaPi-IIb. In addition, mutations of NaPi-IIc (S138F and R468W) in HHRH did not cause cellular PIP2-rich vacuoles. In conclusion, our data anticipate that NaPi-IIc may regulate PIP2 production at the plasma membrane and cellular vesicle formation.
Collapse
Affiliation(s)
- Yuji Shiozaki
- Department of Molecular Nutrition, University of Tokushima Graduate School
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hayashi H, Edin F, Li H, Liu W, Rask-Andersen H. The effect of pulsed electric fields on the electrotactic migration of human neural progenitor cells through the involvement of intracellular calcium signaling. Brain Res 2016; 1652:195-203. [PMID: 27746154 DOI: 10.1016/j.brainres.2016.09.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/07/2016] [Accepted: 09/27/2016] [Indexed: 01/15/2023]
Abstract
Endogenous electric fields (EFs) are required for the physiological control of the central nervous system development. Application of the direct current EFs to neural stem cells has been studied for the possibility of stem cell transplantation as one of the therapies for brain injury. EFs generated within the nervous system are often associated with action potentials and synaptic activity, apparently resulting in a pulsed current in nature. The aim of this study is to investigate the effect of pulsed EF, which can reduce the cytotoxicity, on the migration of human neural progenitor cells (hNPCs). We applied the mono-directional pulsed EF with a strength of 250mV/mm to hNPCs for 6h. The migration distance of the hNPCs exposed to pulsed EF was significantly greater compared with the control not exposed to the EF. Pulsed EFs, however, had less of an effect on the migration of the differentiated hNPCs. There was no significant change in the survival of hNPCs after exposure to the pulsed EF. To investigate the role of Ca2+ signaling in electrotactic migration of hNPCs, pharmacological inhibition of Ca2+ channels in the EF-exposed cells revealed that the electrotactic migration of hNPCs exposed to Ca2+ channel blockers was significantly lower compared to the control group. The findings suggest that the pulsed EF induced migration of hNPCs is partly influenced by intracellular Ca2+ signaling.
Collapse
Affiliation(s)
- Hisamitsu Hayashi
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden; Department of Otolaryngology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan.
| | - Fredrik Edin
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden.
| | - Hao Li
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden.
| | - Wei Liu
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden.
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden.
| |
Collapse
|
38
|
Taghian T, Narmoneva DA, Kogan AB. Modulation of cell function by electric field: a high-resolution analysis. J R Soc Interface 2016; 12:rsif.2015.0153. [PMID: 25994294 DOI: 10.1098/rsif.2015.0153] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Regulation of cell function by a non-thermal, physiological-level electromagnetic field has potential for vascular tissue healing therapies and advancing hybrid bioelectronic technology. We have recently demonstrated that a physiological electric field (EF) applied wirelessly can regulate intracellular signalling and cell function in a frequency-dependent manner. However, the mechanism for such regulation is not well understood. Here, we present a systematic numerical study of a cell-field interaction following cell exposure to the external EF. We use a realistic experimental environment that also recapitulates the absence of a direct electric contact between the field-sourcing electrodes and the cells or the culture medium. We identify characteristic regimes and present their classification with respect to frequency, location, and the electrical properties of the model components. The results show a striking difference in the frequency dependence of EF penetration and cell response between cells suspended in an electrolyte and cells attached to a substrate. The EF structure in the cell is strongly inhomogeneous and is sensitive to the physical properties of the cell and its environment. These findings provide insight into the mechanisms for frequency-dependent cell responses to EF that regulate cell function, which may have important implications for EF-based therapies and biotechnology development.
Collapse
Affiliation(s)
- T Taghian
- Department of Physics, University of Cincinnati, 345 Clifton Court, RM 400 Geo/Physics Building, Cincinnati, OH 45221-0011, USA
| | - D A Narmoneva
- Department of Biomedical, Chemical, and Environmental Engineering, University of Cincinnati, 2901 Woodside Dr., ML 0012, Cincinnati, OH 45221, USA
| | - A B Kogan
- Department of Physics, University of Cincinnati, 345 Clifton Court, RM 400 Geo/Physics Building, Cincinnati, OH 45221-0011, USA
| |
Collapse
|
39
|
Rouabhia M, Park HJ, Zhang Z. Electrically Activated Primary Human Fibroblasts Improve In Vitro and In Vivo Skin Regeneration. J Cell Physiol 2016; 231:1814-21. [PMID: 26661681 DOI: 10.1002/jcp.25289] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 12/10/2015] [Indexed: 12/14/2022]
Abstract
Electrical stimulation (ES) changes cellular behaviors and thus constitutes a potential strategy to promote wound healing. However, well-controlled in vitro findings have yet to be translated to in vivo trials. This study was to demonstrate the feasibility and advantages of transplanting electrically activated cells (E-Cells) to help wound healing. Primary human skin fibroblasts were activated through well defined ES and cultured with keratinocytes to generate engineered human skin (EHS), which were transplanted to nu/nu mice. The electrically activated EHS grafts were analyzed at 20 and 30 days post-grafting, showing faster wound closure, thick epidermis, vasculature, and functional basement membrane containing laminin and type IV collagen that were totally produced by the implanted human cells. Because a variety of cells can be electrically activated, E-Cells may become a new cell source and the transplantation of E-Cells may represent a new strategy in wound healing and tissue engineering. J. Cell. Physiol. 231: 1814-1821, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mahmoud Rouabhia
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, Canada
| | - Hyun Jin Park
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, Canada.,Département de Chirurgie, Faculté de Médecine, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada
| | - Ze Zhang
- Département de Chirurgie, Faculté de Médecine, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada
| |
Collapse
|
40
|
Collective cell migration: guidance principles and hierarchies. Trends Cell Biol 2015; 25:556-66. [DOI: 10.1016/j.tcb.2015.06.003] [Citation(s) in RCA: 227] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/21/2015] [Accepted: 06/08/2015] [Indexed: 12/18/2022]
|
41
|
Tumour cell membrane poration and ablation by pulsed low-intensity electric field with carbon nanotubes. Int J Mol Sci 2015; 16:6890-901. [PMID: 25822874 PMCID: PMC4424994 DOI: 10.3390/ijms16046890] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 01/04/2023] Open
Abstract
Electroporation is a physical method to increase permeabilization of cell membrane by electrical pulses. Carbon nanotubes (CNTs) can potentially act like "lighting rods" or exhibit direct physical force on cell membrane under alternating electromagnetic fields thus reducing the required field strength. A cell poration/ablation system was built for exploring these effects of CNTs in which two-electrode sets were constructed and two perpendicular electric fields could be generated sequentially. By applying this system to breast cancer cells in the presence of multi-walled CNTs (MWCNTs), the effective pulse amplitude was reduced to 50 V/cm (main field)/15 V/cm (alignment field) at the optimized pulse frequency (5 Hz) of 500 pulses. Under these conditions instant cell membrane permeabilization was increased to 38.62%, 2.77-fold higher than that without CNTs. Moreover, we also observed irreversible electroporation occurred under these conditions, such that only 39.23% of the cells were viable 24 h post treatment, in contrast to 87.01% cell viability without presence of CNTs. These results indicate that CNT-enhanced electroporation has the potential for tumour cell ablation by significantly lower electric fields than that in conventional electroporation therapy thus avoiding potential risks associated with the use of high intensity electric pulses.
Collapse
|