1
|
Li J, Chen M, Cheng S, Gao S, Zhai J, Yu D, Wang J, Zhang J, Cai K. Sensorable zwitterionic antibacterial hydrogel for wound electrostimulation therapy. Biomaterials 2025; 315:122958. [PMID: 39547138 DOI: 10.1016/j.biomaterials.2024.122958] [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: 06/01/2024] [Revised: 10/22/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Wound healing process has always been a focal point of concern, with a plethora of hydrogel dressings available; however, their therapeutic efficacy remains a hindrance to wound closure. This article reports on a dual-network conductive system, PEDOT:PSS-co-PSBMA/XLG (PPSX) hydrogel dressing, Constructed using poly(3,4-ethylenedioxythiophene):poly(styrene sulfonate) (PEDOT: PSS) in combination with zwitterionic N, N-dimethyl-N-(2-methacryloyloxyethyl)-N- (3-sulfopropyl) ammonium betaine (SBMA) and nanoclay-synthesized lithium magnesium silicate (XLG). The hydrogel powder produced from it can absorb interfacial water within 30 s via physical interactions to spontaneously form hydrogels of arbitrary shapes. With a conductivity of 1.8 s/m, it can be utilized for developing flexible sensing bioelectronic devices to monitor human activities (facial expressions, blinking, swallowing, speaking, joint movements), as well as constructing electrodes for monitoring muscle movements and motorial intensity. More importantly, PPSX hydrogel effectively inhibits bacterial growth and promotes cell proliferation, thus facilitating wound healing and presenting extensive application prospects in the medical field.
Collapse
Affiliation(s)
- Jinghua Li
- The 1st Affiliated Hospital, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, 471000, China; Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Wound Repair, The First Affiliated Hospital, College of Emergency and Trauma, Hainan Medical University, Haikou, 570100, China; Key Laboratory of Comprehensive and Highly Efficient Utilization of Salt Lake Resources, Qinghai Institute of Salt Lakes, Chinese Academy of Sciences, Xining, 810008, China.
| | - Meijun Chen
- The 1st Affiliated Hospital, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, 471000, China
| | - Shaowen Cheng
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Wound Repair, The First Affiliated Hospital, College of Emergency and Trauma, Hainan Medical University, Haikou, 570100, China
| | - Shegan Gao
- The 1st Affiliated Hospital, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, 471000, China.
| | - Jingming Zhai
- The 1st Affiliated Hospital, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, 471000, China
| | - Dongmei Yu
- Key Laboratory of Comprehensive and Highly Efficient Utilization of Salt Lake Resources, Qinghai Institute of Salt Lakes, Chinese Academy of Sciences, Xining, 810008, China
| | - Jianping Wang
- Key Laboratory of Comprehensive and Highly Efficient Utilization of Salt Lake Resources, Qinghai Institute of Salt Lakes, Chinese Academy of Sciences, Xining, 810008, China
| | - Jianbo Zhang
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
2
|
James EC, Tomaskovic‐Crook E, Crook JM. Engineering 3D Scaffold-Free Nanoparticle-Laden Stem Cell Constructs for Piezoelectric Enhancement of Human Neural Tissue Formation and Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310010. [PMID: 39049737 PMCID: PMC11516115 DOI: 10.1002/advs.202310010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/04/2024] [Indexed: 07/27/2024]
Abstract
Electrical stimulation (ES) of cellular systems can be utilized for biotechnological applications and electroceuticals (bioelectric medicine). Neural cell stimulation especially has a long history in neuroscience research and is increasingly applied for clinical therapies. Application of ES via conventional electrodes requires external connectors and power sources, hindering scientific and therapeutic applications. Here engineering novel 3D scaffold-free human neural stem cell constructs with integrated piezoelectric nanoparticles for enhanced neural tissue induction and function is described. Tetragonal barium titanate (BaTi03) nanoparticles are employed as piezoelectric stimulators prepared as cytocompatible dispersions, incorporated into 3D self-organizing neural spheroids, and activated wirelessly by ultrasound. Ultrasound delivery (low frequency; 40 kHz) is optimized for cell survival, and nanoparticle activation enabled ES throughout the spheroids during differentiation, tissue formation, and maturation. The resultant human neural tissues represent the first example of direct tissue loading with piezoelectric particles for ensuing 3D ultrasound-mediated piezoelectric enhancement of human neuronal induction from stem cells, including augmented neuritogenesis and synaptogenesis. It is anticipated that the platform described will facilitate advanced tissue engineering and in vitro modeling of human neural (and potentially non-neural) tissues, with modeling including tissue development and pathology, and applicable to preclinical testing and prototyping of both electroceuticals and pharmaceuticals.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials ScienceIntelligent Polymer Research InstituteAIIM FacilityUniversity of WollongongFairy MeadowNSW2519Australia
- Arto Hardy Family Biomedical Innovation HubChris O'Brien LifehouseCamperdownNSW2050Australia
| | - Eva Tomaskovic‐Crook
- ARC Centre of Excellence for Electromaterials ScienceIntelligent Polymer Research InstituteAIIM FacilityUniversity of WollongongFairy MeadowNSW2519Australia
- Arto Hardy Family Biomedical Innovation HubChris O'Brien LifehouseCamperdownNSW2050Australia
- School of Medical SciencesFaculty of Medicine and HealthThe University of SydneyCamperdownNSW2006Australia
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials ScienceIntelligent Polymer Research InstituteAIIM FacilityUniversity of WollongongFairy MeadowNSW2519Australia
- Arto Hardy Family Biomedical Innovation HubChris O'Brien LifehouseCamperdownNSW2050Australia
- School of Medical SciencesFaculty of Medicine and HealthThe University of SydneyCamperdownNSW2006Australia
- Institute of Innovative MaterialsAIIM FacilityInnovation CampusFaculty of Engineering and Information SystemsUniversity of WollongongFairy MeadowNSW2519Australia
| |
Collapse
|
3
|
Poling HM, Singh A, Krutko M, Reza AA, Srivastava K, Wells JM, Helmrath MA, Esfandiari L. Promoting Human Intestinal Organoid Formation and Stimulation Using Piezoelectric Nanofiber Matrices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598673. [PMID: 38915647 PMCID: PMC11195230 DOI: 10.1101/2024.06.12.598673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Human organoid model systems have changed the landscape of developmental biology and basic science. They serve as a great tool for human specific interrogation. In order to advance our organoid technology, we aimed to test the compatibility of a piezoelectric material with organoid generation, because it will create a new platform with the potential for sensing and actuating organoids in physiologically relevant ways. We differentiated human pluripotent stem cells into spheroids following the traditional human intestinal organoid (HIO) protocol atop a piezoelectric nanofiber scaffold. We observed that exposure to the biocompatible piezoelectric nanofibers promoted spheroid morphology three days sooner than with the conventional methodology. At day 28 of culture, HIOs grown on the scaffold appeared similar. Both groups were readily transplantable and developed well-organized laminated structures. Graft sizes between groups were similar. Upon characterizing the tissue further, we found no detrimental effects of the piezoelectric nanofibers on intestinal patterning or maturation. Furthermore, to test the practical feasibility of the material, HIOs were also matured on the nanofiber scaffolds and treated with ultrasound, which lead to increased cellular proliferation which is critical for organoid development and tissue maintenance. This study establishes a proof of concept for integrating piezoelectric materials as a customizable platform for on-demand electrical stimulation of cells using remote ultrasonic waveforms in regenerative medicine.
Collapse
|
4
|
Sun Y, Xiao Z, Chen B, Zhao Y, Dai J. Advances in Material-Assisted Electromagnetic Neural Stimulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400346. [PMID: 38594598 DOI: 10.1002/adma.202400346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/26/2024] [Indexed: 04/11/2024]
Abstract
Bioelectricity plays a crucial role in organisms, being closely connected to neural activity and physiological processes. Disruptions in the nervous system can lead to chaotic ionic currents at the injured site, causing disturbances in the local cellular microenvironment, impairing biological pathways, and resulting in a loss of neural functions. Electromagnetic stimulation has the ability to generate internal currents, which can be utilized to counter tissue damage and aid in the restoration of movement in paralyzed limbs. By incorporating implanted materials, electromagnetic stimulation can be targeted more accurately, thereby significantly improving the effectiveness and safety of such interventions. Currently, there have been significant advancements in the development of numerous promising electromagnetic stimulation strategies with diverse materials. This review provides a comprehensive summary of the fundamental theories, neural stimulation modulating materials, material application strategies, and pre-clinical therapeutic effects associated with electromagnetic stimulation for neural repair. It offers a thorough analysis of current techniques that employ materials to enhance electromagnetic stimulation, as well as potential therapeutic strategies for future applications.
Collapse
Affiliation(s)
- Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
5
|
Lim PLK, Balakrishnan Y, Goh G, Tham KC, Ng YZ, Lunny DP, Leavesley DI, Bonnard C. Automated Electrical Stimulation Therapy Accelerates Re-Epithelialization in a Three-Dimensional In Vitro Human Skin Wound Model. Adv Wound Care (New Rochelle) 2024; 13:217-234. [PMID: 38062745 DOI: 10.1089/wound.2023.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024] Open
Affiliation(s)
- Priscilla L K Lim
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Yamini Balakrishnan
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Gracia Goh
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Khek-Chian Tham
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Yi Zhen Ng
- Tissue Technologies, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| | - Declan P Lunny
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Asian Skin Biobank, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| | - David I Leavesley
- Tissue Technologies, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| | - Carine Bonnard
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Asian Skin Biobank, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| |
Collapse
|
6
|
Suarato G, Pressi S, Menna E, Ruben M, Petrini EM, Barberis A, Miele D, Sandri G, Salerno M, Schirato A, Alabastri A, Athanassiou A, Proietti Zaccaria R, Papadopoulou EL. Modified Carbon Nanotubes Favor Fibroblast Growth by Tuning the Cell Membrane Potential. ACS APPLIED MATERIALS & INTERFACES 2024; 16:3093-3105. [PMID: 38206310 PMCID: PMC10811621 DOI: 10.1021/acsami.3c14527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024]
Abstract
As is known, carbon nanotubes favor cell growth in vitro, although the underlying mechanisms are not yet fully elucidated. In this study, we explore the hypothesis that electrostatic fields generated at the interface between nonexcitable cells and appropriate scaffold might favor cell growth by tuning their membrane potential. We focused on primary human fibroblasts grown on electrospun polymer fibers (poly(lactic acid)─PLA) with embedded multiwall carbon nanotubes (MWCNTs). The MWCNTs were functionalized with either the p-methoxyphenyl (PhOME) or the p-acetylphenyl (PhCOMe) moiety, both of which allowed uniform dispersion in a solvent, good mixing with PLA and the consequent smooth and homogeneous electrospinning process. The inclusion of the electrically conductive MWCNTs in the insulating PLA matrix resulted in differences in the surface potential of the fibers. Both PLA and PLA/MWCNT fiber samples were found to be biocompatible. The main features of fibroblasts cultured on different substrates were characterized by scanning electron microscopy, immunocytochemistry, Rt-qPCR, and electrophysiology revealing that fibroblasts grown on PLA/MWCNT reached a healthier state as compared to pure PLA. In particular, we observed physiological spreading, attachment, and Vmem of fibroblasts on PLA/MWCNT. Interestingly, the electrical functionalization of the scaffold resulted in a more suitable extracellular environment for the correct biofunctionality of these nonexcitable cells. Finally, numerical simulations were also performed in order to understand the mechanism behind the different cell behavior when grown either on PLA or PLA/MWCNT samples. The results show a clear effect on the cell membrane potential, depending on the underlying substrate.
Collapse
Affiliation(s)
- Giulia Suarato
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Samuel Pressi
- Department
of Chemical Sciences, University of Padua, via Marzolo 1, 35131 Padova, Italy
- Interdepartmental
Centre Giorgio Levi Cases for Energy Economics and Technology, University of Padua, via Marzolo 9, 35131 Padova, Italy
| | - Enzo Menna
- Department
of Chemical Sciences, University of Padua, via Marzolo 1, 35131 Padova, Italy
- Interdepartmental
Centre Giorgio Levi Cases for Energy Economics and Technology, University of Padua, via Marzolo 9, 35131 Padova, Italy
| | - Massimo Ruben
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | | | - Andrea Barberis
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Dalila Miele
- Department
of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department
of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Marco Salerno
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Schirato
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
- Dipartimento
di Fisica, Politecnico di Milano, Pizza Leonardo da Vinci 32, Milan 20133, Italy
- Department
of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Alessandro Alabastri
- Department
of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | | | | | | |
Collapse
|
7
|
Agam O, Braun E. Universal calcium fluctuations in Hydramorphogenesis. Phys Biol 2023; 20:066002. [PMID: 37696269 DOI: 10.1088/1478-3975/acf8a4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/11/2023] [Indexed: 09/13/2023]
Abstract
Understanding the collective physical processes that drive robust morphological transitions in animal development necessitates the characterization of the relevant fields involved in morphogenesis. Calcium (Ca2+) is recognized as one such field. In this study, we demonstrate that the spatial fluctuations of Ca2+duringHydraregeneration exhibit universal characteristics. To investigate this phenomenon, we employ two distinct controls, an external electric field andheptanol, a gap junction-blocking drug. Both lead to the modulation of the Ca2+activity and a reversible halting of the regeneration process. The application of an electric field enhances Ca2+activity in theHydra's tissue and increases its spatial correlations, while the administration ofheptanolinhibits its activity and diminishes the spatial correlations. Remarkably, the statistical characteristics of Ca2+spatial fluctuations, including the coefficient of variation and skewness, manifest universal shape distributions across tissue samples and conditions. We introduce a field-theoretic model, describing fluctuations in a tilted double-well potential, which successfully captures these universal properties. Moreover, our analysis reveals that the Ca2+activity is spatially localized, and theHydra's tissue operates near the onset of bistability, where the local Ca2+activity fluctuates between low and high excited states in distinct regions. These findings highlight the prominent role of the Ca2+field inHydramorphogenesis and provide insights into the underlying mechanisms governing robust morphological transitions.
Collapse
Affiliation(s)
- Oded Agam
- The Racah Institute of Physics, Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Erez Braun
- Department of Physics and Network Biology Research Laboratories, Technion-Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
8
|
Petsakou A, Liu Y, Liu Y, Comjean A, Hu Y, Perrimon N. Epithelial Ca 2+ waves triggered by enteric neurons heal the gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553227. [PMID: 37645990 PMCID: PMC10461974 DOI: 10.1101/2023.08.14.553227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
A fundamental and unresolved question in regenerative biology is how tissues return to homeostasis after injury. Answering this question is essential for understanding the etiology of chronic disorders such as inflammatory bowel diseases and cancer. We used the Drosophila midgut to investigate this question and discovered that during regeneration a subpopulation of cholinergic enteric neurons triggers Ca2+ currents among enterocytes to promote return of the epithelium to homeostasis. Specifically, we found that down-regulation of the cholinergic enzyme Acetylcholinesterase in the epithelium enables acetylcholine from defined enteric neurons, referred as ARCENs, to activate nicotinic receptors in enterocytes found near ARCEN-innervations. This activation triggers high Ca2+ influx that spreads in the epithelium through Inx2/Inx7 gap junctions promoting enterocyte maturation followed by reduction of proliferation and inflammation. Disrupting this process causes chronic injury consisting of ion imbalance, Yki activation and increase of inflammatory cytokines together with hyperplasia, reminiscent of inflammatory bowel diseases. Altogether, we found that during gut regeneration the conserved cholinergic pathway facilitates epithelial Ca2+ waves that heal the intestinal epithelium. Our findings demonstrate nerve- and bioelectric-dependent intestinal regeneration which advance the current understanding of how a tissue returns to its homeostatic state after injury and could ultimately help existing therapeutics.
Collapse
Affiliation(s)
| | - Yifang Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Aram Comjean
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Boston, USA
| |
Collapse
|
9
|
Abasi S, Jain A, Cooke JP, Guiseppi-Elie A. Electrically stimulated gene expression under exogenously applied electric fields. Front Mol Biosci 2023; 10:1161191. [PMID: 37214334 PMCID: PMC10192815 DOI: 10.3389/fmolb.2023.1161191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/14/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction: Electrical stimulation, the application of an electric field to cells and tissues grown in culture to accelerate growth and tight junction formation among endothelial cells, could be impactful in cardiovascular tissue engineering, allotransplantation, and wound healing. Methods: Using Electrical Cell Stimulation And Recording Apparatus (ECSARA), the exploration of the stimulatory influences of electric fields of different magnitude and frequencies on growth and proliferation, trans endothelial electrical resistance (TEER) and gene expression of human endothelia cells (HUVECs) were explored. Results: Within the range of endogenous electrical pulses studied, frequency was found to be more significant (p = 0.05) than voltage in influencing HUVEC gene expression. Localization of Yes Associated Protein (YAP) and expression of CD-144 are shown to be consistent with temporal manifestations of TEER. Discussion: This work introduces the field of electromics, the study of cellular gene expression profiles and their implications under the influence of exogenously applied electric fields. Homology of electrobiology and mechanobiology suggests use of such exogenous cues in tissue and regenerative engineering.
Collapse
Affiliation(s)
- Sara Abasi
- Bioelectronics, Biosensors and Biochips (C3B), Department of Biomedical Engineering, Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Abhishek Jain
- Bioelectronics, Biosensors and Biochips (C3B), Department of Biomedical Engineering, Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, Houston, TX, United States
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, United States
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, Houston, TX, United States
| | - Anthony Guiseppi-Elie
- Bioelectronics, Biosensors and Biochips (C3B), Department of Biomedical Engineering, Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, Houston, TX, United States
- Division of Engineering and Industrial Technology, Tri-County Technical College, Pendleton, SC, United States
- ABTECH Scientific, Inc., Richmond, VA, United States
| |
Collapse
|
10
|
Moreddu R, Boschi A, d’Amora M, Hubarevich A, Dipalo M, De Angelis F. Passive Recording of Bioelectrical Signals from Non-Excitable Cells by Fluorescent Mirroring. NANO LETTERS 2023; 23:3217-3223. [PMID: 37019439 PMCID: PMC10141418 DOI: 10.1021/acs.nanolett.2c05053] [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] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Bioelectrical variations trigger different cell responses, including migration, mitosis, and mutation. At the tissue level, these actions result in phenomena such as wound healing, proliferation, and pathogenesis. Monitoring these mechanisms dynamically is highly desirable in diagnostics and drug testing. However, existing technologies are invasive: either they require physical access to the intracellular compartments, or they imply direct contact with the cellular medium. Here, we present a novel approach for the passive recording of electrical signals from non-excitable cells adhering to 3D microelectrodes, based on optical mirroring. Preliminary results yielded a fluorescence intensity output increase of the 5,8% in the presence of a HEK-293 cell on the electrode compared to bare microelectrodes. At present, this technology may be employed to evaluate cell-substrate adhesion and monitor cell proliferation. Further refinements could allow extrapolating quantitative data on surface charges and resting potential to investigate the electrical phenomena involved in cell migration and cancer progression.
Collapse
Affiliation(s)
- Rosalia Moreddu
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
| | - Alessio Boschi
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Department
of Bioengineering, University of Genoa, 16126 Genoa, Italy
| | - Marta d’Amora
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Department
of Biology, University of Pisa, 56127 Pisa, Italy
| | | | - Michele Dipalo
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Email
| | - Francesco De Angelis
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Email
| |
Collapse
|
11
|
Role of Innate Immune Cells in Chronic Diabetic Wounds. J Indian Inst Sci 2023. [DOI: 10.1007/s41745-022-00355-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
12
|
Li H, Li B, Lv D, Li W, Lu Y, Luo G. Biomaterials releasing drug responsively to promote wound healing via regulation of pathological microenvironment. Adv Drug Deliv Rev 2023; 196:114778. [PMID: 36931347 DOI: 10.1016/j.addr.2023.114778] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/06/2022] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Wound healing is characterized by complex, orchestrated, spatiotemporal dynamic processes. Recent findings demonstrated suitable local microenvironments were necessities for wound healing. Wound microenvironments include various biological, biochemical and physical factors, which are produced and regulated by endogenous biomediators, exogenous drugs, and external environment. Successful drug delivery to wound is complicated, and need to overcome the destroyed blood supply, persistent inflammation and enzymes, spatiotemporal requirements of special supplements, and easy deactivation of drugs. Triggered by various factors from wound microenvironment itself or external elements, stimuli-responsive biomaterials have tremendous advantages of precise drug delivery and release. Here, we discuss recent advances of stimuli-responsive biomaterials to regulate local microenvironments during wound healing, emphasizing on the design and application of different biomaterials which respond to wound biological/biochemical microenvironments (ROS, pH, enzymes, glucose and glutathione), physical microenvironments (mechanical force, temperature, light, ultrasound, magnetic and electric field), and the combination modes. Moreover, several novel promising drug carriers (microbiota, metal-organic frameworks and microneedles) are also discussed.
Collapse
Affiliation(s)
- Haisheng Li
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Buying Li
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dalun Lv
- Department of Burn and Plastic Surgery, First Affiliated Hospital of Wannan Medical College, Wuhu City, China; Beijing Jayyalife Biological Technology Company, Beijing, China
| | - Wenhong Li
- Beijing Jayyalife Biological Technology Company, Beijing, China
| | - Yifei Lu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
13
|
Funk RHW, Scholkmann F. The significance of bioelectricity on all levels of organization of an organism. Part 1: From the subcellular level to cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:185-201. [PMID: 36481271 DOI: 10.1016/j.pbiomolbio.2022.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/24/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Bioelectricity plays an essential role in the structural and functional organization of biological organisms. In this first article of our three-part series, we summarize the importance of bioelectricity for the basic structural level of biological organization, i.e. from the subcellular level (charges, ion channels, molecules and cell organelles) to cells.
Collapse
Affiliation(s)
- Richard H W Funk
- Institute of Anatomy, Center for Theoretical Medicine, TU-Dresden, 01307, Dresden, Germany; Dresden International University, 01067, Dresden, Germany.
| | - Felix Scholkmann
- Biomedical Optics Research Laboratory, Department of Neonatology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland.
| |
Collapse
|
14
|
Yasmin R, Deaton R. Logical computation with self-assembling electric circuits. PLoS One 2022; 17:e0278033. [PMID: 36477295 PMCID: PMC9728908 DOI: 10.1371/journal.pone.0278033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Inspired by self-assembled biological growth, the Circuit Tile Assembly Model (cTAM) was developed to provide insights into signal propagation, information processing, and computation in bioelectric networks. The cTAM is an abstract model that produces a family of circuits of different sizes that is amenable to exact analysis. Here, the cTAM is extended to the Boolean Circuit Tile Assembly Model (bcTAM) that implements a computationally complete set of Boolean gates through self-assembled and self-controlled growth. The proposed model approximates axonal growth in neural networks and thus, investigates the computational capability of dynamic biological networks, for example, in growing networks of axons. Thus, the bcTAM models the effect of electrical activity on growth and shows how that growth might implement Boolean computations. In this sense, given a set of input voltages, the bcTAM is a system that is able to monitor and make decisions about its own growth.
Collapse
Affiliation(s)
- Rojoba Yasmin
- Department of Electrical Engineering, University of Wisconsin Green Bay, Green Bay, WI, United States of America,* E-mail:
| | - Russell Deaton
- Department of Electrical and Computer Engineering, The University of Memphis, Memphis, TN, United States of America
| |
Collapse
|
15
|
Bioelectric regulation of intestinal stem cells. Trends Cell Biol 2022:S0962-8924(22)00234-3. [PMID: 36396487 PMCID: PMC10183058 DOI: 10.1016/j.tcb.2022.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022]
Abstract
Proper regulation of ion balance across the intestinal epithelium is essential for physiological functions, while ion imbalance causes intestinal disorders with dire health consequences. Ion channels, pumps, and exchangers are vital for regulating ion movements (i.e., bioelectric currents) that control epithelial absorption and secretion. Recent in vivo studies used the Drosophila gut to identify conserved pathways that link regulators of Ca2+, Na+ and Cl- with intestinal stem cell (ISC) proliferation. These studies laid a foundation for using the Drosophila gut to identify conserved proliferative responses triggered by bioelectric regulators. Here, we review these studies, discuss their significance, as well as the advantages of using Drosophila to unravel conserved bioelectrically induced molecular pathways in the intestinal epithelium under physiological, pathophysiological, and regenerative conditions.
Collapse
|
16
|
Trueman RP, Ahlawat AS, Phillips JB. A Shock to the (Nervous) System: Bioelectricity Within Peripheral Nerve Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1137-1150. [PMID: 34806913 DOI: 10.1089/ten.teb.2021.0159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The peripheral nervous system has the remarkable ability to regenerate in response to injury. However, this is only successful over shorter nerve gaps and often provides poor outcomes for patients. Currently, the gold standard of treatment is the surgical intervention of an autograft, whereby patient tissue is harvested and transplanted to bridge the nerve gap. Despite being the gold standard, more than half of patients have dissatisfactory functional recovery after an autograft. Peripheral nerve tissue engineering aims to create biomaterials that can therapeutically surpass the autograft. Current tissue-engineered constructs are designed to deliver a combination of therapeutic benefits to the regenerating nerve, such as supportive cells, alignment, extracellular matrix, soluble factors, immunosuppressants, and other therapies. An emerging therapeutic opportunity in nerve tissue engineering is the use of electrical stimulation (ES) to modify and enhance cell function. ES has been shown to positively affect four key cell types, such as neurons, endothelial cells, macrophages, and Schwann cells, involved in peripheral nerve repair. Changes elicited include faster neurite extension, cellular alignment, and changes in cell phenotype associated with improved regeneration and functional recovery. This review considers the relevant modes of administration and cellular responses that could underpin incorporation of ES into nerve tissue engineering strategies. Impact Statement Tissue engineering is becoming increasingly complex, with multiple therapeutic modalities often included within the final tissue-engineered construct. Electrical stimulation (ES) is emerging as a viable therapeutic intervention to be included within peripheral nerve tissue engineering strategies; however, to date, there have been no review articles that collate the information regarding the effects of ES on key cell within peripheral nerve injury. This review article aims to inform the field on the different therapeutic effects that may be achieved by using ES and how they may become incorporated into existing strategies.
Collapse
Affiliation(s)
- Ryan P Trueman
- Center for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - Ananya S Ahlawat
- Center for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - James B Phillips
- Center for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
17
|
Systems of axon-like circuits for self-assembled and self-controlled growth of bioelectric networks. Sci Rep 2022; 12:13371. [PMID: 35927304 PMCID: PMC9352688 DOI: 10.1038/s41598-022-17103-4] [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: 04/06/2022] [Accepted: 07/20/2022] [Indexed: 11/09/2022] Open
Abstract
By guiding cell and chemical migration and coupling with genetic mechanisms, bioelectric networks of potentials influence biological pattern formation and are known to have profound effects on growth processes. An abstract model that is amenable to exact analysis has been proposed in the circuit tile assembly model (cTAM) to understand self-assembled and self-controlled growth as an emergent phenomenon that is capable of complex behaviors, like self-replication. In the cTAM, a voltage source represents a finite supply of energy that drives growth until it is unable to overcome randomizing factors in the environment, represented by a threshold. Here, the cTAM is extended to the axon or alternating cTAM model (acTAM) to include a circuit similar to signal propagation in axons, exhibiting time-varying electric signals and a dependence on frequency of the input voltage. The acTAM produces systems of circuits whose electrical properties are coupled to their length as growth proceeds through self-assembly. The exact response is derived for increasingly complex circuit systems as the assembly proceeds. The model exhibits complicated behaviors that elucidate the interactive role of energy, environment, and noise with electric signals in axon-like circuits during biological growth of complex patterns and function.
Collapse
|
18
|
Szechyńska-Hebda M, Lewandowska M, Witoń D, Fichman Y, Mittler R, Karpiński SM. Aboveground plant-to-plant electrical signaling mediates network acquired acclimation. THE PLANT CELL 2022; 34:3047-3065. [PMID: 35595231 PMCID: PMC9338792 DOI: 10.1093/plcell/koac150] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/16/2022] [Indexed: 05/05/2023]
Abstract
Systemic acquired acclimation and wound signaling require the transmission of electrical, calcium, and reactive oxygen species (ROS) signals between local and systemic tissues of the same plant. However, whether such signals can be transmitted between two different plants is largely unknown. Here, we reveal a new type of plant-to-plant aboveground direct communication involving electrical signaling detected at the surface of leaves, ROS, and photosystem networks. A foliar electrical signal induced by wounding or high light stress applied to a single dandelion leaf can be transmitted to a neighboring plant that is in direct contact with the stimulated plant, resulting in systemic photosynthetic, oxidative, molecular, and physiological changes in both plants. Furthermore, similar aboveground changes can be induced in a network of plants serially connected via touch. Such signals can also induce responses even if the neighboring plant is from a different plant species. Our study demonstrates that electrical signals can function as a communication link between transmitter and receiver plants that are organized as a network (community) of plants. This process can be described as network-acquired acclimation.
Collapse
Affiliation(s)
| | | | - Damian Witoń
- Department of Plant Genetics, Breeding and Biotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Yosef Fichman
- The Division of Plant Sciences and Technology and Interdisciplinary Plant Group, College of Agriculture, Food and Natural Resources, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65201, USA
| | - Ron Mittler
- The Division of Plant Sciences and Technology and Interdisciplinary Plant Group, College of Agriculture, Food and Natural Resources, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65201, USA
| | | |
Collapse
|
19
|
Carvalho J. A computational model of organism development and carcinogenesis resulting from cells' bioelectric properties and communication. Sci Rep 2022; 12:9206. [PMID: 35654933 PMCID: PMC9163332 DOI: 10.1038/s41598-022-13281-3] [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: 11/15/2021] [Accepted: 05/23/2022] [Indexed: 11/15/2022] Open
Abstract
A sound theory of biological organization is clearly missing for a better interpretation of observational results and faster progress in understanding life complexity. The availability of such a theory represents a fundamental progress in explaining both normal and pathological organism development. The present work introduces a computational implementation of some principles of a theory of organism development, namely that the default state of cells is proliferation and motility, and includes the principle of variation and organization by closure of constraints. In the present model, the bioelectric context of cells and tissue is the field responsible for organization, as it regulates cell proliferation and the level of communication driving the system's evolution. Starting from a depolarized (proliferative) cell, the organism grows to a certain size, limited by the increasingly polarized state after successive proliferation events. The system reaches homeostasis, with a depolarized core (proliferative cells) surrounded by a rim of polarized cells (non-proliferative in this condition). This state is resilient to cell death (random or due to injure) and to limited depolarization (potentially carcinogenic) events. Carcinogenesis is introduced through a localized event (a spot of depolarized cells) or by random depolarization of cells in the tissue, which returns cells to their initial proliferative state. The normalization of the bioelectric condition can reverse this out-of-equilibrium state to a new homeostatic one. This simplified model of embryogenesis, tissue organization and carcinogenesis, based on non-excitable cells' bioelectric properties, can be made more realistic with the introduction of other components, like biochemical fields and mechanical interactions, which are fundamental for a more faithful representation of reality. However, even a simple model can give insight for new approaches in complex systems and suggest new experimental tests, focused in its predictions and interpreted under a new paradigm.
Collapse
Affiliation(s)
- Joao Carvalho
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
20
|
Chaudhari SD, Sharma KK, Marchetto JJ, Hydren JR, Burton BM, Moreno AP. Modulating OPG and TGF-β1 mRNA expression via bioelectrical stimulation. Bone Rep 2021; 15:101141. [PMID: 34692946 PMCID: PMC8517839 DOI: 10.1016/j.bonr.2021.101141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Bone remodeling is a lifelong process that ranges from orthodontic tooth movement/alignment to bone damage/healing, to overall bone health. Osteoprotegerin (OPG) and transforming growth factor β1 (TGF-β1) are secreted by osteoblasts and participate in bone remodeling. OPG promotes bone remineralization and stabilization prominent in post-mechanical repositioning of the teeth in the dental alveolus. TGF-β1 participates in regulatory processes to promote osteoblast and osteoclast equilibrium. In the context of orthodontic tooth movement, post-treatment fixation requires additional, exogenous, stabilization support. Recent research showcases supplementary solutions, in conjunction to standard tooth fixation techniques, such as OPG injections into gum and periodontal tissues to accelerate tooth anchorage; however, injections are prone to post-procedure complications and discomfort. This study utilizes noninvasive bioelectric stimulation (BES) to modulate OPG and TGF-β1 as a novel solution to regulate bone remineralization specifically in the context of post-orthodontic tooth movement. PURPOSE The aim of this study was to investigate a spectrum of BES parameters that would modulate OPG and TGF-β1 expression in osteoblasts. METHODS Osteoblasts were cultured and stimulated using frequencies from 25 Hz to 3 MHz. RT-qPCR was used to quantify changes in OPG and TGFb-1 mRNA expression. RESULTS OPG mRNA expression was significantly increased at frequencies above 10,000 Hz with a maximum expression increase of 332 ± 8% at 100 kHz. Conversely, OPG mRNA expression was downregulated at frequencies lower than 1000 Hz. TGF-β1 mRNA expression increased throughout all stimulation frequencies with a peak of 332 ± 72% at 250 kHz. Alizarin Red tests for calcium, indicated that mineralization of stimulated osteoblasts in vitro increased 28% after 6 weeks in culture. DISCUSSION Results support the working hypothesis that OPG and TGF-β1 mRNA expression can be modulated through BES. Noninvasive BES approaches have the potential to accelerate bone remineralization by providing a novel tool to supplement the anchorage process, reduce complications, and promote patient compliance and reduce post-treatment relapse. Noninvasive BES may be applicable to other clinical applications as a novel therapeutic tool to modulate bone remodeling.
Collapse
Affiliation(s)
- Sejal D. Chaudhari
- OrthodontiCell Inc., Leonhardt Launchpads Utah, Inc., Salt Lake City, UT 84115, United States of America
| | - Kapil K. Sharma
- OrthodontiCell Inc., Leonhardt Launchpads Utah, Inc., Salt Lake City, UT 84115, United States of America
| | - John J. Marchetto
- OrthodontiCell Inc., Leonhardt Launchpads Utah, Inc., Salt Lake City, UT 84115, United States of America
- John J. Marchetto DMD, Weston, FL 33326, United States of America
| | - Jay R. Hydren
- OrthodontiCell Inc., Leonhardt Launchpads Utah, Inc., Salt Lake City, UT 84115, United States of America
| | - Brett M. Burton
- OrthodontiCell Inc., Leonhardt Launchpads Utah, Inc., Salt Lake City, UT 84115, United States of America
| | - Alonso P. Moreno
- OrthodontiCell Inc., Leonhardt Launchpads Utah, Inc., Salt Lake City, UT 84115, United States of America
| |
Collapse
|
21
|
Cui S, Rouabhia M, Semlali A, Zhang Z. Effects of electrical stimulation on human skin keratinocyte growth and the secretion of cytokines and growth factors. Biomed Mater 2021; 16. [PMID: 34592730 DOI: 10.1088/1748-605x/ac2bba] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022]
Abstract
Electrical stimulation (ES) has been widely explored and found effective in promoting wound healing. However, the role of ES on keratinocytes, a major player in wound healing, has not been well established. The present work investigated the cellular and molecular behaviors of human skin keratinocytes being exposed to ES. HaCaT keratinocytes were seeded on a novel electrically conductive and soft PPy-PU/PLLA membrane and cultured under electrical intensities of 100 or 200 mV mm-1for 6 and 24 h. The factors assessed after ES include cell proliferation, colony formation, cytokines, keratins, as well as phosphorylated ERK1/2 (pERK1/2) kinases. The results showed that the electrically stimulated cells exhibited a higher proliferative ability and secreted more IL-6, IL-1α, IL-8, GROα, FGF2, and VEGF-A. Interestingly, the 24 h ES induced a 'stimulus memory' by showing a significant rise in colony-forming efficiency in post-ES cells that were sub-cultured. Additionally, after stopping the 24 h ES, the productions of keratin 5 and keratin 14 were continuously increased for 3 d. The productions of keratin 10 and keratin 13 were significantly increased post the 6 h ES. Finally, the ES increased pERK1/2 kinases. The overall results demonstrated that the proliferation of keratinocytes and their secretion of cytokines and growth factors can be activated through appropriate ES to benefit skin wound healing.
Collapse
Affiliation(s)
- Shujun Cui
- Research Group on Oral Ecology, Faculty of Dentistry, Université Laval, Québec (QC), Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Québec (QC), Canada.,Division of Regenerative Medicine, Research Center of CHU-Université Laval, Québec (QC), Canada
| | - Mahmoud Rouabhia
- Research Group on Oral Ecology, Faculty of Dentistry, Université Laval, Québec (QC), Canada
| | - Abdelhabib Semlali
- Research Group on Oral Ecology, Faculty of Dentistry, Université Laval, Québec (QC), Canada
| | - Ze Zhang
- Department of Surgery, Faculty of Medicine, Université Laval, Québec (QC), Canada.,Division of Regenerative Medicine, Research Center of CHU-Université Laval, Québec (QC), Canada
| |
Collapse
|
22
|
PEDOT:PSS-Coated Polybenzimidazole Electroconductive Nanofibers for Biomedical Applications. Polymers (Basel) 2021; 13:polym13162786. [PMID: 34451324 PMCID: PMC8401200 DOI: 10.3390/polym13162786] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 11/18/2022] Open
Abstract
Bioelectricity drives several processes in the human body. The development of new materials that can deliver electrical stimuli is gaining increasing attention in the field of tissue engineering. In this work, novel, highly electrically conductive nanofibers made of poly [2,2′-m-(phenylene)-5,5′-bibenzimidazole] (PBI) have been manufactured by electrospinning and then coated with cross-linked poly (3,4-ethylenedioxythiophene) doped with poly (styrene sulfonic acid) (PEDOT:PSS) by spin coating or dip coating. These scaffolds have been characterized by scanning electron microscopy (SEM) imaging and attenuated total reflectance Fourier-transform infrared (ATR-FTIR) spectroscopy. The electrical conductivity was measured by the four-probe method at values of 28.3 S·m−1 for spin coated fibers and 147 S·m−1 for dip coated samples, which correspond, respectively, to an increase of about 105 and 106 times in relation to the electrical conductivity of PBI fibers. Human bone marrow-derived mesenchymal stromal cells (hBM-MSCs) cultured on the produced scaffolds for one week showed high viability, typical morphology and proliferative capacity, as demonstrated by calcein fluorescence staining, 4′,6-diamidino-2-phenylindole (DAPI)/Phalloidin staining and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide] assay. Therefore, all fiber samples demonstrated biocompatibility. Overall, our findings highlight the great potential of PEDOT:PSS-coated PBI electrospun scaffolds for a wide variety of biomedical applications, including their use as reliable in vitro models to study pathologies and the development of strategies for the regeneration of electroactive tissues or in the design of new electrodes for in vivo electrical stimulation protocols.
Collapse
|
23
|
Abstract
It is well known that electrical signals are deeply associated with living entities. Much of our understanding of excitable tissues is derived from studies of specialized cells of neurons or myocytes. However, electric potential is present in all cell types and results from the differential partitioning of ions across membranes. This electrical potential correlates with cell behavior and tissue organization. In recent years, there has been exciting, and broadly unexpected, evidence linking the regulation of development to bioelectric signals. However, experimental modulation of electrical potential can have multifaceted and pleiotropic effects, which makes dissecting the role of electrical signals in development difficult. Here, I review evidence that bioelectric cues play defined instructional roles in orchestrating development and regeneration, and further outline key areas in which to refine our understanding of this signaling mechanism.
Collapse
Affiliation(s)
- Matthew P. Harris
- Department of Genetics, Harvard Medical School, Department of Orthopaedics, Boston Children's Hospital, 300 Longwood Avenue Enders 260, Boston MA 02115, USA
| |
Collapse
|
24
|
James EC, Tomaskovic-Crook E, Crook JM. Bioengineering Clinically Relevant Cardiomyocytes and Cardiac Tissues from Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22063005. [PMID: 33809429 PMCID: PMC8001925 DOI: 10.3390/ijms22063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The regenerative capacity of cardiomyocytes is insufficient to functionally recover damaged tissue, and as such, ischaemic heart disease forms the largest proportion of cardiovascular associated deaths. Human-induced pluripotent stem cells (hiPSCs) have enormous potential for developing patient specific cardiomyocytes for modelling heart disease, patient-based cardiac toxicity testing and potentially replacement therapy. However, traditional protocols for hiPSC-derived cardiomyocytes yield mixed populations of atrial, ventricular and nodal-like cells with immature cardiac properties. New insights gleaned from embryonic heart development have progressed the precise production of subtype-specific hiPSC-derived cardiomyocytes; however, their physiological immaturity severely limits their utility as model systems and their use for drug screening and cell therapy. The long-entrenched challenges in this field are being addressed by innovative bioengingeering technologies that incorporate biophysical, biochemical and more recently biomimetic electrical cues, with the latter having the potential to be used to both direct hiPSC differentiation and augment maturation and the function of derived cardiomyocytes and cardiac tissues by mimicking endogenous electric fields.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy 3065, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| |
Collapse
|
25
|
Analysis of the Differential Gene and Protein Expression Profiles of Corneal Epithelial Cells Stimulated with Alternating Current Electric Fields. Genes (Basel) 2021; 12:genes12020299. [PMID: 33672614 PMCID: PMC7924190 DOI: 10.3390/genes12020299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/17/2022] Open
Abstract
In cells, intrinsic endogenous direct current (DC) electric fields (EFs) serve as morphogenetic cues and are necessary for several important cellular responses including activation of multiple signaling pathways, cell migration, tissue regeneration and wound healing. Endogenous DC EFs, generated spontaneously following injury in physiological conditions, directly correlate with wound healing rate, and different cell types respond to these EFs via directional orientation and migration. Application of external DC EFs results in electrode polarity and is known to activate intracellular signaling events in specific direction. In contrast, alternating current (AC) EFs are known to induce continuous bidirectional flow of charged particles without electrode polarity and also minimize electrode corrosion. In this context, the present study is designed to study effects of AC EFs on corneal epithelial cell gene and protein expression profiles in vitro. We performed gene and antibody arrays, analyzed the data to study specific influence of AC EFs, and report that AC EFs has no deleterious effect on epithelial cell function. Gene Ontology results, following gene and protein array data analysis, showed that AC EFs influence similar biological processes that are predominantly responsive to organic substance, chemical, or external stimuli. Both arrays activate cytokine–cytokine receptor interaction, MAPK and IL-17 signaling pathways. Further, in comparison to the gene array data, the protein array data show enrichment of diverse activated signaling pathways through several interconnecting networks.
Collapse
|
26
|
Mohana Sundaram P, Rangharajan KK, Akbari E, Hadick TJ, Song JW, Prakash S. Direct current electric field regulates endothelial permeability under physiologically relevant fluid forces in a microfluidic vessel bifurcation model. LAB ON A CHIP 2021; 21:319-330. [PMID: 33319218 PMCID: PMC7855772 DOI: 10.1039/d0lc00507j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Previous in vitro studies have reported on the use of direct current electric fields (DC-EFs) to regulate vascular endothelial permeability, which is important for tissue regeneration and wound healing. However, these studies have primarily used static 2D culture models that lack the fluid mechanical forces associated with blood flow experienced by endothelial cells (ECs) in vivo. Hence, the effect of DC-EF on ECs under physiologically relevant fluid forces is yet to be systematically evaluated. Using a 3D microfluidic model of a bifurcating vessel, we report the role of DC-EF on regulating endothelial permeability when co-applied with physiologically relevant fluid forces that arise at the vessel bifurcation. The application of a 70 V m-1 DC-EF simultaneously with 1 μL min-1 low perfusion rate (generating 3.8 dyn cm-2 stagnation pressure at the bifurcation point and 0.3 dyn cm-2 laminar shear stress in the branched vessel) increased the endothelial permeability 7-fold compared to the static control condition (i.e., without flow and DC-EF). When the perfusion rate was increased to 10 μL min-1 (generating 38 dyn cm-2 stagnation pressure at the bifurcation point and 3 dyn cm-2 laminar shear stress in the branched vessel) while maintaining the same electrical stimulation, a 4-fold increase in endothelial permeability compared to the static control was observed. The lower increase in endothelial permeability for the higher fluid forces but the same DC-EF suggests a competing role between fluid forces and the applied DC-EF. Moreover, the observed increase in endothelial permeability due to combined DC-EF and flow was transient and dependent on the Akt signalling pathway. Collectively, these findings provide significant new insights into how the endothelium serves as an electro-mechanical interface for regulating vessel permeability.
Collapse
|
27
|
Gibney S, Hicks JM, Robinson A, Jain A, Sanjuan-Alberte P, Rawson FJ. Toward nanobioelectronic medicine: Unlocking new applications using nanotechnology. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1693. [PMID: 33442962 DOI: 10.1002/wnan.1693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/29/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
Abstract
Bioelectronic medicine aims to interface electronic technology with biological components and design more effective therapeutic and diagnostic tools. Advances in nanotechnology have moved the field forward improving the seamless interaction between biological and electronic components. In the lab many of these nanobioelectronic devices have the potential to improve current treatment approaches, including those for cancer, cardiovascular disorders, and disease underpinned by malfunctions in neuronal electrical communication. While promising, many of these devices and technologies require further development before they can be successfully applied in a clinical setting. Here, we highlight recent work which is close to achieving this goal, including discussion of nanoparticles, carbon nanotubes, and nanowires for medical applications. We also look forward toward the next decade to determine how current developments in nanotechnology could shape the growing field of bioelectronic medicine. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Steven Gibney
- Division of Regenerative Medicine and Cellular Therapies, Biodiscovery Institute,School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Jacqueline M Hicks
- Division of Regenerative Medicine and Cellular Therapies, Biodiscovery Institute,School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Andie Robinson
- Division of Regenerative Medicine and Cellular Therapies, Biodiscovery Institute,School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Akhil Jain
- Division of Regenerative Medicine and Cellular Therapies, Biodiscovery Institute,School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Paola Sanjuan-Alberte
- Division of Regenerative Medicine and Cellular Therapies, Biodiscovery Institute,School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK.,Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Frankie J Rawson
- Division of Regenerative Medicine and Cellular Therapies, Biodiscovery Institute,School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| |
Collapse
|
28
|
Calvo P, Baluška F, Trewavas A. Integrated information as a possible basis for plant consciousness. Biochem Biophys Res Commun 2020; 564:158-165. [PMID: 33081970 DOI: 10.1016/j.bbrc.2020.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/23/2020] [Accepted: 10/09/2020] [Indexed: 12/29/2022]
Abstract
It is commonly assumed that plants do not possess consciousness. Since the criterion for this assumption is usually human consciousness this assumption represents a top down attitude. It is obvious that plants are not animals and using animal criteria of consciousness will lead to its rejection in plants. However using a bottom up evolutionary approach and a leading theory of consciousness, Integrated Information Theory, we report that we find evidence that indicates that plant meristems act in a conscious fashion although probably at the level of minimal consciousness. Since many plants contain multiple meristems these observations highlight a very different evolutionary approach to consciousness in biological organisms.
Collapse
Affiliation(s)
- Paco Calvo
- Minimal Intelligence Laboratory, Universidad de Murcia, Murcia, Spain.
| | - František Baluška
- Institute of Cellular and Molecular Botany, University of Bonn, Bonn, Germany
| | - Anthony Trewavas
- Institute of Molecular Plant Science, Kings Buildings, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
29
|
Sen CK, Mathew-Steiner SS, Das A, Sundaresan VB, Roy S. Electroceutical Management of Bacterial Biofilms and Surgical Infection. Antioxid Redox Signal 2020; 33:713-724. [PMID: 32466673 PMCID: PMC7475090 DOI: 10.1089/ars.2020.8086] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 01/04/2023]
Abstract
Significance: In the host-microbe microenvironment, bioelectrical factors influence microbes and hosts as well as host-microbe interactions. This article discusses relevant mechanistic underpinnings of this novel paradigm. It also addresses how such knowledge may be leveraged to develop novel electroceutical solutions to manage biofilm infection. Recent Advances: Systematic review and meta-analysis of several hundred wound studies reported a 78.2% prevalence of biofilms in chronic wounds. Biofilm infection is a major cause of delayed wound healing. In the host-microbe microenvironment, bioelectrical factors influence interactions between microbes and hosts. Critical Issues: Rapid biological responses are driven by electrical signals generated by ion currents moving across cell membranes. Bacterial life, growth, and function rely on a bioelectrical milieu, which when perturbed impairs their ability to form a biofilm, a major threat to health care. Electrokinetic stability of several viral particles depend on electrostatic forces. Weak electrical field strength, otherwise safe for humans, can be anti-microbial in this context. In the host, the electric field enhanced keratinocyte migration, bolstered immune defenses, improved mitochondrial function, and demonstrated multiple other effects consistent with supporting wound healing. A deeper mechanistic understanding of bioelectrical principles will inform the design of next-generation electroceuticals. Future Directions: This is an opportune moment in time as there is a surge of interest in electroceuticals in medicine. Projected to reach $35.5 billion by 2025, electroceuticals are becoming a cynosure in the global market. The World Health Organization reports that more than 50% of surgical site infections can be antibiotic resistant. Electroceuticals offer a serious alternative.
Collapse
Affiliation(s)
- Chandan K. Sen
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shomita S. Mathew-Steiner
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Amitava Das
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Vishnu Baba Sundaresan
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Li Z, Liu X, Zhang W, Zhuang X. Electrostatic reaction for the detection of circulating tumor cells as a potential diagnostic biomarker for metastasis in solid tumor. Nanotheranostics 2020; 4:233-241. [PMID: 32923313 PMCID: PMC7484632 DOI: 10.7150/ntno.46928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/28/2020] [Indexed: 12/24/2022] Open
Abstract
The detection of circulating tumor cells (CTCs) from blood samples is important to predict metastatic spread of cancer cells. However, effective quantification and identification of CTCs in solid tumors remain a challenge. Aerobic glycolysis is a hallmark of cancer cells, which makes cancer cells have more negative membrane potentials than that of normal cells. Herein, we reported a CTC isolation method with 80.7% capture efficiency based on electrostatic reaction, which was accomplished within 30 min in mimic clinical samples. Following in vitro verification using Lewis lung carcinoma (LLC1) (EpCAM-positive) and B16F10 (EpCAM-negative) melanoma cells, syngeneic tumor models were used to evaluate specificity and sensitivity of the surface charged nanoparticles. After subcutaneous implantation, blood was drawn from mice every three days, and CTCs were successfully detected in all implanted subjects. From 100 µl blood samples, the minimum amount found in blood was 9-34 CTCs on 3 day and the maximum was 94-107 CTCs on 15 day. Besides, the isolated CTCs cells remained viable and verified by re-implantation. This study confirms that our multifunctional nanoparticles are highly efficient in detecting CTCs in tumor metastasis and has huge potential in translational medicine.
Collapse
Affiliation(s)
- Zhiming Li
- Institue of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Department of Urology, the First Affiliated Hospital of Xiamen University, Xiamen 361003, Fujian, China
| | - Xingping Liu
- Institue of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Weidong Zhang
- Clinical Trial Management Platform, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang, China
| | - Xuan Zhuang
- Department of Urology, the First Affiliated Hospital of Xiamen University, Xiamen 361003, Fujian, China.,Department of Clinical Medicine, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| |
Collapse
|
31
|
Bhavsar MB, Leppik L, Costa Oliveira KM, Barker JH. Role of Bioelectricity During Cell Proliferation in Different Cell Types. Front Bioeng Biotechnol 2020; 8:603. [PMID: 32714900 PMCID: PMC7343900 DOI: 10.3389/fbioe.2020.00603] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/18/2020] [Indexed: 11/26/2022] Open
Abstract
Most living organisms possess varying degrees of regenerative capabilities but how these regenerative processes are controlled is still poorly understood. Naturally occurring bioelectric voltages (like Vmem) are thought to be playing instructive role in tissue regeneration, as well as embryonic development. The different distribution of ions on the either side of the cell membrane results in intra- and extra-cellular voltage differences, known as membrane potential or Vmem. The relationship between Vmem and cell physiology is conserved in a wide range of cell types and suggests that Vmem regulation is a fundamental control mechanism for regeneration related processes e.g., proliferation and differentiation. In the present study we measured Vmem in three different cell types (human osteogenic sarcoma cell line (OSC), rat bone marrow derived mesenchymal stem cells (BM-MSC), and rat dermal fibroblasts) and characterized the relationship between their Vmem and proliferation. In order to find out if Vmem controls proliferation, or visa-versa, we blocked and then unblocked Na+/K+-exchanging ATPase using ouabain and measured the proliferation. Our results demonstrate that Vmem can be pharmacologically manipulated to control proliferation in certain cell types like BM-MSC. Taken together, it is clear that control of bioelectrical properties in non-excitable cells could prove to be potentially a useful tool in regenerative medicine efforts.
Collapse
Affiliation(s)
- Mit Balvantray Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt, Germany
| | - Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt, Germany
| | - Karla Mychellyne Costa Oliveira
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt, Germany
| | - John H Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt, Germany
| |
Collapse
|
32
|
Das B, Shrirao A, Golberg A, Berthiaume F, Schloss R, Yarmush ML. Differential Cell Death and Regrowth of Dermal Fibroblasts and Keratinocytes After Application of Pulsed Electric Fields. Bioelectricity 2020; 2:175-185. [PMID: 34471845 PMCID: PMC8370327 DOI: 10.1089/bioe.2020.0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: High-powered pulsed electric fields (PEF) may be used for tissue debridement and disinfection, while lower PEF intensities may stimulate beneficial cellular responses for wound healing. We investigated the dual effects of nonuniform PEF on cellular death and stimulation. Methods: Dermal fibroblast or keratinocyte monolayers were exposed to PEF induced by two needle electrodes (2 mm apart). Voltages (100-600 V; 1 Hz; 70 micros pulse width; 90 pulses/cycle) were applied between the two electrodes. Controls consisted of similar monolayers subjected to a scratch mechanical injury. Results: Cell growth and closure of the cell-free gap was faster in PEF-treated cell monolayers versus scratched ones. Media conditioned from cells pre-exposed to PEF, when applied to responder cells, stimulated greater proliferation than media from scratched monolayers. Conclusions: PEF treatment causes the release of soluble factors that promote cell growth, and thus may play a role in the accelerated healing of wounds post PEF.
Collapse
Affiliation(s)
- Bodhisatwa Das
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Anil Shrirao
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Alexander Golberg
- Department of Environmental Studies, Tel Aviv University, Tel Aviv, Israel
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Rene Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Martin L. Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
- Center for Engineering in Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Shriners Hospitals for Children, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Shin J, Bae S, Seo PJ. De novo shoot organogenesis during plant regeneration. JOURNAL OF EXPERIMENTAL BOTANY 2020; 71:63-72. [PMID: 31504722 DOI: 10.1093/jxb/erz395] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/22/2019] [Indexed: 05/08/2023]
Abstract
Plants exhibit remarkable regeneration capacity, ensuring developmental plasticity. In vitro tissue culture techniques are based on plant regeneration ability and facilitate production of new organs and even the whole plant from explants. Plant somatic cells can be reprogrammed to form a pluripotent cell mass called the callus. A portion of pluripotent callus cells gives rise to a fertile shoot via de novo shoot organogenesis (DNSO). Here, we reconstitute the shoot regeneration process with four phases, namely pluripotency acquisition, shoot promeristem formation, establishment of the confined shoot progenitor, and shoot outgrowth. Additionally, other biological processes, including cell cycle progression and reactive oxygen species metabolism, which further contribute to successful completion of DNSO, are also summarized. Overall, this study highlights recent advances in the molecular and cellular events involved in DNSO, as well as the regulatory mechanisms behind key steps of DNSO.
Collapse
Affiliation(s)
- Jinwoo Shin
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Soonhyung Bae
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Pil Joon Seo
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
- Plant Genomics and Breeding Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
34
|
Ebrahimdamavandi S, Mobasheri H. Application of a static magnetic field as a complementary aid to healing in an in vitro wound model. J Wound Care 2019; 28:40-52. [PMID: 30625046 DOI: 10.12968/jowc.2019.28.1.40] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Static magnetic field (SMF) has long been used as a therapeutic means, though its effects on the activity of cells and the mechanism(s) involved remain unknown. The purpose of this study is to determine the effect of a moderate-intensity SMF on the activity, growth and migration of mouse embryonic fibroblast (NIH 3T3), aiming to mimic wound healing and to study it in real time. METHOD A cell-free area (a scratch with a 200-500µm width) was formed in NIH 3T3 cultured cells and used as a wound model. The effects of a SMF (10, 50, 80 and 100mT) on the survival rate (MTT assay), integrity of cell membranes (lactate dehydrogenase (LDH) assay), the morphology of the cell (circularity, number and length of filopodia), cell orientation, and migration (speed, direction, rate) were studied as a function of the incubation time in a time-lapse manner. RESULTS The exposure of cells to SMF at all intensities had no cytotoxic effect, as revealed by the MTT assay. The integrity of the membranes of the SMF-treated cells studied by the LDH assay test showed no effects. The structure of the membrane at the leading edge of the cells changed and showed several filopodia extended parallel to the field direction. The exposure to the SMF elongated the cells and decreased their circularity at SMF 10mT. The migration of the cells from one edge of the gap towards the other was affected by the applied SMF. The maximum and minimum effects were monitored at 80mT and 10mT, respectively. Analysis of cell migration revealed an average directness of 0.73, 0.66, 0.78, 0.78 and 0.69 under SMF 10, 50, 80, 100mT and control, respectively. CONCLUSION The morphological and functional changes of the cells in the presence of SMF revealed particular effects on the membrane and cytoskeleton. Cells were affected by physicochemical changes caused by the applied SMF, though the extent of the incurred effects was not a linear function of the field intensity. This low cost, non-invasive approach can be used as a magneto-manipulative means to tailor a practical, independent or complementary means of manipulating the activities of cells and tissues for clinical purposes.
Collapse
Affiliation(s)
- Sajedeh Ebrahimdamavandi
- PhD student, Laboratory of Membrane Biophysics and Macromolecules. Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Hamid Mobasheri
- Professor of Biophysics, Head of Laboratory, Laboratory of Membrane Biophysics and Macromolecules. Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran; Biomaterials Research Center (BRC), University of Tehran and Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Ross CL, Zhou Y, McCall CE, Soker S, Criswell TL. The Use of Pulsed Electromagnetic Field to Modulate Inflammation and Improve Tissue Regeneration: A Review. Bioelectricity 2019; 1:247-259. [PMID: 34471827 PMCID: PMC8370292 DOI: 10.1089/bioe.2019.0026] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pulsed electromagnetic field (PEMF) is emerging as innovative treatment for regulation of inflammation, which could have significant effects on tissue regeneration. PEMF modulates inflammatory processes through the regulation of pro- and anti-inflammatory cytokine secretion during different stages of inflammatory response. Consistent outcomes in studies involving animal and human tissue have shown promise for the use of PEMF as an alternative or complementary treatment to pharmaceutical therapies. Thus, PEMF treatment could provide a novel nonpharmaceutical means of modulating inflammation in injured tissues resulting in enhanced functional recovery. This review examines the effect of PEMF on immunomodulatory cells (e.g., mesenchymal stem/stromal cells [MSCs] and macrophages [MΦ]) to better understand the potential for PEMF therapy to modulate inflammatory signaling pathways and improve tissue regeneration. This review cites published data that support the use of PEMF to improve tissue regeneration. Our studies included herein confirm anti-inflammatory effects of PEMF on MSCs and MΦ.
Collapse
Affiliation(s)
- Christina L. Ross
- Center for Integrative Medicine, Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Yu Zhou
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Charles E. McCall
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Shay Soker
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tracy L. Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
36
|
A journey in the complex interactions between electrochemistry and bacteriology: From electroactivity to electromodulation of bacterial biofilms. Bioelectrochemistry 2019; 131:107401. [PMID: 31707278 DOI: 10.1016/j.bioelechem.2019.107401] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 02/06/2023]
Abstract
Although the term bioelectrochemistry tends to be associated with animal and human tissues, bioelectric currents exist also in plants and bacteria. Especially the latter, when agglomerated in the form of biofilms, can exhibit electroactivity and susceptibility to electrical stimulation. Therefore, electrochemical methods appear to become powerful techniques to expand the conventional strategies of biofilm characterization and modification. In this review, we aim to provide the insight into the electrochemical behaviour of bacteria and present the variety of electrochemical techniques that can be used either for the non-destructive monitoring of bacterial communities or modulation of their growth. The most common applications of electrical stimulation on biofilms are presented, including the prevention of bacterial growth by charging the surface of the materials, changing the direction of bacterial movement under the influence of the electric field and increasing of the potency of antibiotics when bactericides are coupled with the electric field. Also, the industrial applications of microbial electro-technologies are described, such as bioremediation, wastewater treatment, and microbial fuel cells. Consequently, we are showing the complexity of interactions that exist between electrochemistry and bacteriology that can be used for the benefit of these two disciplines.
Collapse
|
37
|
Electric-Induced Reversal of Morphogenesis in Hydra. Biophys J 2019; 117:1514-1523. [PMID: 31570230 PMCID: PMC6817546 DOI: 10.1016/j.bpj.2019.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023] Open
Abstract
Morphogenesis involves the dynamic interplay of biochemical, mechanical, and electrical processes. Here, we ask to what extent can the course of morphogenesis be modulated and controlled by an external electric field? We show that at a critical amplitude, an external electric field can halt morphogenesis in Hydra regeneration. Moreover, above this critical amplitude, the electric field can lead to reversal dynamics: a fully developed Hydra folds back into its incipient spheroid morphology. The potential to renew morphogenesis is reexposed when the field is reduced back to amplitudes below criticality. These dynamics are accompanied by modulations of the Wnt3 activity, a central component of the head organizer in Hydra. The controlled backward-forward cycle of morphogenesis can be repeated several times, showing that the reversal trajectory maintains the integrity of the tissue and its regeneration capability. Each cycle of morphogenesis leads to a newly emerged body plan in the redeveloped folded tissue, which is not necessarily similar to the one before the reversal process. Reversal of morphogenesis is shown to be triggered by enhanced electrical excitations in the Hydra tissue, leading to intensified calcium activity. Folding back of the body-plan morphology together with the decay of a central biosignaling system, indicate that electrical processes are tightly integrated with biochemical and mechanical-structural processes in morphogenesis and play an instructive role to a level that can direct developmental trajectories. Reversal of morphogenesis by external fields calls for extending its framework beyond program-like, forward-driven, hierarchical processes based on reaction diffusion and positional information.
Collapse
|
38
|
Mehta AS, Singh A. Insights into regeneration tool box: An animal model approach. Dev Biol 2019; 453:111-129. [PMID: 30986388 PMCID: PMC6684456 DOI: 10.1016/j.ydbio.2019.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/20/2022]
Abstract
For ages, regeneration has intrigued countless biologists, clinicians, and biomedical engineers. In recent years, significant progress made in identification and characterization of a regeneration tool kit has helped the scientific community to understand the mechanism(s) involved in regeneration across animal kingdom. These mechanistic insights revealed that evolutionarily conserved pathways like Wnt, Notch, Hedgehog, BMP, and JAK/STAT are involved in regeneration. Furthermore, advancement in high throughput screening approaches like transcriptomic analysis followed by proteomic validations have discovered many novel genes, and regeneration specific enhancers that are specific to highly regenerative species like Hydra, Planaria, Newts, and Zebrafish. Since genetic machinery is highly conserved across the animal kingdom, it is possible to engineer these genes and regeneration specific enhancers in species with limited regeneration properties like Drosophila, and mammals. Since these models are highly versatile and genetically tractable, cross-species comparative studies can generate mechanistic insights in regeneration for animals with long gestation periods e.g. Newts. In addition, it will allow extrapolation of regenerative capabilities from highly regenerative species to animals with low regeneration potential, e.g. mammals. In future, these studies, along with advancement in tissue engineering applications, can have strong implications in the field of regenerative medicine and stem cell biology.
Collapse
Affiliation(s)
- Abijeet S Mehta
- Department of Biology, University of Dayton, Dayton, OH, 45469, USA
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, 45469, USA; Premedical Program, University of Dayton, Dayton, OH, 45469, USA; Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, 45469, USA; The Integrative Science and Engineering Center, University of Dayton, Dayton, OH, 45469, USA; Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, IN, USA.
| |
Collapse
|
39
|
Miller WB, Torday JS, Baluška F. The N-space Episenome unifies cellular information space-time within cognition-based evolution. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 150:112-139. [PMID: 31415772 DOI: 10.1016/j.pbiomolbio.2019.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/26/2019] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
Self-referential cellular homeostasis is maintained by the measured assessment of both internal status and external conditions based within an integrated cellular information field. This cellular field attachment to biologic information space-time coordinates environmental inputs by connecting the cellular senome, as the sum of the sensory experiences of the cell, with its genome and epigenome. In multicellular organisms, individual cellular information fields aggregate into a collective information architectural matrix, termed a N-space Episenome, that enables mutualized organism-wide information management. It is hypothesized that biological organization represents a dual heritable system constituted by both its biological materiality and a conjoining N-space Episenome. It is further proposed that morphogenesis derives from reciprocations between these inter-related facets to yield coordinated multicellular growth and development. The N-space Episenome is conceived as a whole cell informational projection that is heritable, transferable via cell division and essential for the synchronous integration of the diverse self-referential cells that constitute holobionts.
Collapse
Affiliation(s)
| | - John S Torday
- Department of Pediatrics, Harbor-UCLA Medical Center, USA.
| | | |
Collapse
|
40
|
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: 86] [Impact Index Per Article: 17.2] [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
|
41
|
Srinivasan TM. Resonance Signaling and Yoga. Int J Yoga 2018; 11:89-90. [PMID: 29755215 PMCID: PMC5934955 DOI: 10.4103/ijoy.ijoy_16_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- T M Srinivasan
- Division of Yoga and Physical Sciences, Swami Vivekananda Yoga Anusandhana Samsthana, Bengaluru, Karnataka, India. E-mail:
| |
Collapse
|