1
|
Coquart P, El Haddad A, Koutsouras DA, Bolander J. Organic Bioelectronics in Microphysiological Systems: Bridging the Gap Between Biological Systems and Electronic Technologies. BIOSENSORS 2025; 15:253. [PMID: 40277566 DOI: 10.3390/bios15040253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/28/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025]
Abstract
The growing burden of degenerative, cardiovascular, neurodegenerative, and cancerous diseases necessitates innovative approaches to improve our pathophysiological understanding and ability to modulate biological processes. Organic bioelectronics has emerged as a powerful tool in this pursuit, offering a unique ability to interact with biology due to the mixed ionic-electronic conduction and tissue-mimetic mechanical properties of conducting polymers (CPs). These materials enable seamless integration with biological systems across different levels of complexity, from monolayers to complex 3D models, microfluidic chips, and even clinical applications. CPs can be processed into diverse formats, including thin films, hydrogels, 3D scaffolds, and electrospun fibers, allowing the fabrication of advanced bioelectronic devices such as multi-electrode arrays, transistors (EGOFETs, OECTs), ion pumps, and photoactuators. This review examines the integration of CP-based bioelectronics in vivo and in in vitro microphysiological systems, focusing on their ability to monitor key biological events, including electrical activity, metabolic changes, and biomarker concentrations, as well as their potential for electrical, mechanical, and chemical stimulation. We highlight the versatility and biocompatibility of CPs and their role in advancing personalized medicine and regenerative therapies and discuss future directions for organic bioelectronics to bridge the gap between biological systems and electronic technologies.
Collapse
Affiliation(s)
- Pauline Coquart
- Research Unit 'Soft Matter and Biophysics', Department 'Physics and Astronomy', KU Leuven, B-3000 Leuven, Belgium
- IMEC, Kapeldreef 75, B-3001 Leuven, Belgium
| | - Andrea El Haddad
- IMEC, Kapeldreef 75, B-3001 Leuven, Belgium
- Research Unit 'Assiocated Division ESAT-INSYS (INSYS), Integrated Systems', Department 'Electrical Engineering (ESAT)', KU Leuven, B-3000 Leuven, Belgium
| | - Dimitrios A Koutsouras
- IMEC NL, 5656 AE Eindhoven, The Netherlands
- Department of Electronic & Electrical Engineering, University of Bath, Claverton Down, Bath BA2 7AY, UK
- Centre for Bioengineering & Biomedical Technologies (CBio), University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Johanna Bolander
- IMEC, Kapeldreef 75, B-3001 Leuven, Belgium
- Berlin Institute of Health Center for Regenerative Therapied (BCRT), Berlin Institute of Health at Charité-Universitätmedizin Berlin, 13353 Berlin, Germany
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité-Universitätmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
2
|
Roslan A, Paulus K, Yang J, Matt L, Bischof H, Längst N, Schanz S, Luczak A, Cruz Santos M, Burgstaller S, Skrabak D, Bork NI, Malli R, Schmidtko A, Gawaz M, Nikolaev VO, Ruth P, Ehinger R, Lukowski R. Slack K+ channels confer protection against myocardial ischaemia/reperfusion injury. Cardiovasc Res 2025; 121:174-189. [PMID: 39102831 DOI: 10.1093/cvr/cvae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/26/2024] [Accepted: 06/01/2024] [Indexed: 08/07/2024] Open
Abstract
AIMS Na+-activated Slack potassium (K+) channels are increasingly recognized as regulators of neuronal activity, yet little is known about their role in the cardiovascular system. Slack activity increases when intracellular Na+ concentration ([Na+]i) reaches pathophysiological levels. Elevated [Na+]i is a major determinant of the ischaemia and reperfusion (I/R)-induced myocardial injury; thus, we hypothesized that Slack plays a role under these conditions. METHODS AND RESULTS K+ currents in cardiomyocytes (CMs) obtained from wildtype but not from global Slack knockout mice were sensitive to electrical inactivation of voltage-sensitive Na+ channels. Live-cell imaging demonstrated that K+ fluxes across the sarcolemma rely on Slack, while the depolarized resting membrane potential in Slack-deficient CMs led to excessive cytosolic Ca2+ accumulation and finally to hypoxia/reoxygenation-induced cell death. Cardiac damage in an in vivo model of I/R was exacerbated in global and CM-specific conditional Slack mutants and largely insensitive to mechanical conditioning manoeuvres. Finally, the protection conferred by mitochondrial ATP-sensitive K+ (mitoKATP) channels required functional Slack in CMs. CONCLUSION Collectively, our study provides evidence for Slack's crucial involvement in the ion homeostasis of no or low O2-stressed CMs. Thereby, Slack activity opposes the I/R-induced fatal Ca2+-uptake to CMs supporting the cardioprotective signaling attributed to mitoKATP function.
Collapse
Affiliation(s)
- Anna Roslan
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Katharina Paulus
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Jiaqi Yang
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Lucas Matt
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Helmut Bischof
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Natalie Längst
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Sophia Schanz
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Annika Luczak
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Sandra Burgstaller
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - David Skrabak
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Nadja I Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, and Center for Medical Research, CF Bioimaging, Medical University of Graz, Graz, Austria
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical Pharmacy, Goethe-Universität Frankfurt a.M., Frankfurt a.M., Germany
| | - Meinrad Gawaz
- Department of Cardiology & Cardiovascular Diseases, University Hospital Tuebingen, Tuebingen, Germany
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Rebekka Ehinger
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, Auf der Morgenstelle 8, University of Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
3
|
Chen L, Yao F, Xue S, Yao K, Huang Y, Zhao H, Shao Q. Dynamic membrane changes and osmotic effects by sugar alcohols. NPJ Sci Food 2025; 9:46. [PMID: 40159509 PMCID: PMC11955526 DOI: 10.1038/s41538-025-00410-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025] Open
Abstract
Sugar alcohols are natural sweeteners with various physiological functions, commonly used in low-calorie foods and pharmaceuticals. Current research primarily focuses on their sweetening properties and metabolic effects, often overlooking their interactions with cell membranes. This study built a giant phospholipid vesicle model to examine vesicle deformation in erythritol (Ery) and xylitol (Xyl) environments. The permeation of these sugar alcohols through real cell membranes was also investigated. Fluorescence microscopy and zeta potential measurements showed that osmotic stress from concentration gradients disrupted vesicle membrane structure. Ery and Xyl reduced ROS levels in HEK-293 cells and influenced membrane permeability. Notably, Xyl increased vesicle adsorption on the cell membrane at the same concentration. The findings indicate that sugar alcohols interact with membrane lipids through hydrogen bonds or other non-covalent interactions, modifying cell membrane structure and properties, thus providing a theoretical foundation for understanding their role in physiological environments.
Collapse
Affiliation(s)
- Lichun Chen
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
- Physical Chemistry and Soft Matter, Wageningen University and Research, Wageningen, WE, Netherlands
| | - Feng Yao
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Songwen Xue
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Kuang Yao
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yun Huang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Huimin Zhao
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Qiong Shao
- China National Bamboo Research Center, Key Laboratory of State Forestry and Grassland Administration on Bamboo Forest Ecology and Resource Utilization, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
4
|
Li J, Shao N, Zhang Y, Liu X, Zhang H, Tian L, Piatkevich KD, Zhang D, Lee HJ. Screening of Vibrational Spectroscopic Voltage Indicator by Stimulated Raman Scattering Microscopy. SMALL METHODS 2025:e2402124. [PMID: 40095346 DOI: 10.1002/smtd.202402124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Genetically encoded voltage indicators (GEVIs) have significantly advanced voltage imaging, offering spatial details at cellular and subcellular levels not easily accessible with electrophysiology. In addition to fluorescence imaging, certain chemical bond vibrations are sensitive to membrane potential changes, presenting an alternative imaging strategy; however, challenges in signal sensitivity and membrane specificity highlight the need to develop vibrational spectroscopic GEVIs (vGEVIs) in mammalian cells. To address this need, a vGEVI screening approach is developed that employs hyperspectral stimulated Raman scattering (hSRS) imaging synchronized with an induced transmembrane voltage (ITV) stimulation, revealing unique spectroscopic signatures of sensors expressed on membranes. Specifically, by screening various rhodopsin-based voltage sensors in live mammalian cells, a characteristic peak associated with retinal bound to the sensor is identified in one of the GEVIs, Archon, which exhibited a 70 cm-1 red shift relative to the membrane-bound retinal. Notably, this peak is responsive to changes in membrane potential. Overall, hSRS-ITV presents a promising platform for screening vGEVIs, paving the way for advancements in vibrational spectroscopic voltage imaging.
Collapse
Affiliation(s)
- Jingyuan Li
- College of Biomedical Engineering & Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310058, China
| | - Ninghui Shao
- College of Biomedical Engineering & Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310058, China
| | - Yongqing Zhang
- Zhejiang Key Laboratory of Micro-nano Quantum Chips and Quantum Control, School of Physics, Zhejiang University, Hangzhou, 310027, China
| | - Xingxin Liu
- Zhejiang Key Laboratory of Micro-nano Quantum Chips and Quantum Control, School of Physics, Zhejiang University, Hangzhou, 310027, China
| | - Hanbin Zhang
- School of Life Sciences, Westlake Laboratory of Life Sciences and Biomedicine, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Westlake University, Hangzhou, 310024, China
| | - Liangfei Tian
- College of Biomedical Engineering & Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310058, China
| | - Kiryl D Piatkevich
- School of Life Sciences, Westlake Laboratory of Life Sciences and Biomedicine, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Westlake University, Hangzhou, 310024, China
| | - Delong Zhang
- Zhejiang Key Laboratory of Micro-nano Quantum Chips and Quantum Control, School of Physics, Zhejiang University, Hangzhou, 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration of Zhejiang University, Hangzhou, 310027, China
| | - Hyeon Jeong Lee
- College of Biomedical Engineering & Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310058, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration of Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
5
|
Roy D, Michalet X, Miller EW, Bharadwaj K, Weiss S. Toward measurements of absolute membrane potential in Bacillus subtilis using fluorescence lifetime. BIOPHYSICAL REPORTS 2025; 5:100196. [PMID: 39798601 PMCID: PMC11835658 DOI: 10.1016/j.bpr.2025.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Membrane potential (MP) changes can provide a simple readout of bacterial functional and metabolic state or stress levels. While several optical methods exist for measuring fast changes in MP in excitable cells, there is a dearth of such methods for absolute and precise measurements of steady-state MPs in bacterial cells. Conventional electrode-based methods for the measurement of MP are not suitable for calibrating optical methods in small bacterial cells. While optical measurement based on Nernstian indicators have been successfully used, they do not provide absolute or precise quantification of MP or its changes. We present a novel, calibrated MP recording approach to address this gap. In this study, we used a fluorescence lifetime-based approach to obtain a single-cell-resolved distribution of the membrane potential and its changes upon extracellular chemical perturbation in a population of bacterial cells for the first time. Our method is based on 1) a unique VoltageFluor (VF) optical transducer, whose fluorescence lifetime varies as a function of MP via photoinduced electron transfer and 2) a quantitative phasor-FLIM analysis for high-throughput readout. This method allows MP changes to be easily visualized, recorded and quantified. By artificially modulating potassium concentration gradients across the membrane using an ionophore, we have obtained a Bacillus subtilis-specific MP versus VF lifetime calibration and estimated the MP for unperturbed B. subtilis cells to be -65 mV (in minimal salts glycerol glutamate [MSgg]), -127 mV (in M9), and that for chemically depolarized cells as -14 mV (in MSgg). We observed a population-level MP heterogeneity of ∼6-10 mV indicating a considerable degree of diversity of physiological and metabolic states among individual cells. Our work paves the way for deeper insights into bacterial electrophysiology and bioelectricity research.
Collapse
Affiliation(s)
- Debjit Roy
- UCLA-DOE Institute for Genomics and Proteomics, Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, California; Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, California
| | - Xavier Michalet
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, California; California Nano Systems Institute, University of California at Los Angeles, Los Angeles, California.
| | - Evan W Miller
- Departments of Chemistry, Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, California
| | - Kiran Bharadwaj
- UCLA-DOE Institute for Genomics and Proteomics, Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, California; Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, California
| | - Shimon Weiss
- UCLA-DOE Institute for Genomics and Proteomics, Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, California; Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, California; Department of Physiology, University of California at Los Angeles, Los Angeles, California; California Nano Systems Institute, University of California at Los Angeles, Los Angeles, California; Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
6
|
Wang T, Kim DH, Ding C, Wang D, Zhang W, Silic M, Cheng X, Shao K, Ku T, Zheng C, Xie J, Yuan C, Chubykin A, Staiger CJ, Zhang G, Deng Q. Inwardly rectifying potassium channels regulate membrane potential polarization and direction sensing during neutrophil chemotaxis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641746. [PMID: 40093039 PMCID: PMC11908270 DOI: 10.1101/2025.03.06.641746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Potassium channels regulate membrane potential and diverse physiological processes, including cell migration. However, the specific function of the inwardly rectifying potassium channels in immune cell chemotaxis is unknown. Here, we identified that the inwardly rectifying potassium channel Kir7.1 (KCNJ13) maintains the resting membrane potential and is required for directional sensing during neutrophil chemotaxis. Pharmacological or genetic inhibition of Kir7.1 in neutrophils impaired direction sensing toward various chemoattractants without affecting cell polarization in multiple neutrophil models. Using genetically encoded voltage indicators, we observed oscillating depolarization of the membrane potential in protrusions in zebrafish neutrophils, and Kir7.1 is required for polarized depolarization towards the chemokine source. Focal depolarization with optogenetic tools biases pseudopod selection and induces de novo protrusions. Global hyperpolarizing neutrophils stalled cell migration. Furthermore, Kir7.1 regulates GPCR signaling activation. This work adds membrane potential to the intricate feedforward mechanism, coupling the adaptive and excitable network required to steer immune cells in complex tissue environments.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Daniel H Kim
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Chang Ding
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Dingxun Wang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Weiwei Zhang
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN47907, USA
| | - Martin Silic
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Xi Cheng
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Kunming Shao
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - TingHsuan Ku
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Conwy Zheng
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN47907, USA
| | - Alexander Chubykin
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, West Lafayette, IN, 47907
| | - Christopher J Staiger
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN47907, USA
| | - Guangjun Zhang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Qing Deng
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
7
|
Mahapatra C, Kishore A, Gawad J, Al-Emam A, Kouzeiha RA, Rusho MA. Review of electrophysiological models to study membrane potential changes in breast cancer cell transformation and tumor progression. Front Physiol 2025; 16:1536165. [PMID: 40110186 PMCID: PMC11920174 DOI: 10.3389/fphys.2025.1536165] [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: 12/02/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The transformation of normal breast cells into cancerous cells is a complex process influenced by both genetic and microenvironmental factors. Recent studies highlight the significant role of membrane potential (Vm) alterations in this transformation. Cancer cells typically exhibit a depolarized resting membrane potential (RMP) compared to normal cells, which correlates with increased cellular activity and more aggressive cancer behavior. These RMP and Vm changes are associated with altered ion channel activity, altered calcium dynamics, mitochondrial dysfunction, modified gap junction communication, and disrupted signaling pathways. Such fluctuations in RMP and Vm influence key processes in cancer progression, including cell proliferation, migration, and invasion. Notably, more aggressive subtypes of breast cancer cells display more frequent and pronounced Vm fluctuations. Understanding the electrical properties of cancer cells provides new insights into their behavior and offers potential therapeutic targets, such as ion channels and Vm regulation. This review synthesizes current research on how various factors modulate membrane potential and proposes an electrophysiological model of breast cancer cells based on experimental and clinical data from the literature. These findings may pave the way for novel pharmacological targets for clinicians, researchers, and pharmacologists in treating breast cancer.
Collapse
Affiliation(s)
| | - Arnaw Kishore
- Microbiology and Immunology, Xavier University School of Medicine, Aruba, Netherlands
| | - Jineetkumar Gawad
- Department of Pharmaceutical Chemistry, VIVA Institute of Pharmacy, Virar, India
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Riad Azzam Kouzeiha
- Faculty of Medical Sciences, Lebanese University, Hadath Campus, Beirut, Lebanon
| | - Maher Ali Rusho
- Department of Biomedical Engineering, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
8
|
Cibrão JR, Armada M, Lima MF, Vidinha-Mira A, Campos J, Pinho TS, Salgado AJ, Ainla A, Silva NA. Development and application of a novel multi-channel in vitro electrical stimulator for cellular research. BMC Biomed Eng 2025; 7:3. [PMID: 40025549 PMCID: PMC11874659 DOI: 10.1186/s42490-025-00090-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Exposure to electric fields affects cell membranes impacting their potential and altering cellular excitability, nerve transmission, or muscle contraction. Furthermore, electric stimulation influences cell communication, migration, proliferation, and differentiation, with potential therapeutic applications. In vitro platforms for electrical stimulation are valuable tools for studying these effects and advancing medical research. In this study, we developed and tested a novel multi-channel in vitro electrical stimulator designed for cellular applications. The device aims to facilitate research on the effects of electrical stimulation (ES) on cellular processes, providing a versatile platform that is easy to reproduce and implement in various laboratory settings. METHODS The stimulator was designed to be simple, cost-effective, and versatile, fitting on standard 12-well plates for parallel experimentation. Extensive testing was conducted to evaluate the performance of the stimulator, including 3D finite element modelling to analyse electric field distribution. Moreover, the stimulator was evaluated in vitro using neuronal and stem cell cultures. RESULTS Finite element modelling confirmed that the electric field was sufficiently homogeneous within the stimulation zone, though liquid volume affected field strength. A custom controller was developed to program stimulation protocols, ensuring precise and adjustable current delivery up to 160 V/m. ES promoted neurite outgrowth when applied to SH-SY5Y neural cells or to primary spinal cord-derived cells. In human neuronal progenitor cells (hNPCs), ES enhanced neurite growth as well as differentiation into neurons. In adipose stem cells (ASCs), ES altered the secretome, enriching it in molecules that promoted hNPC differentiation into neurons without enhancing neurite growth. CONCLUSIONS Our results highlight the potential of this multi-channel electrical stimulator as a valuable tool for advancing the understanding of ES mechanisms and its therapeutic applications. The simplicity and adaptability of this novel platform make it a promising addition to the toolkit of researchers studying electrical stimulation in cellular models.
Collapse
Affiliation(s)
- Jorge R Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal
| | - Miguel Armada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal
| | - Marta F Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal
| | - André Vidinha-Mira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal
| | - Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal
| | - Alar Ainla
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal.
| | - Nuna A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal.
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, 4806-909, Portugal.
| |
Collapse
|
9
|
Yang L, Tang C, Cui Y, Zhang J. A High-Throughput Screening Strategy for Bacillus subtilis Producing Menaquinone-7 Based on Fluorescence-Activated Cell Sorting. Microorganisms 2025; 13:536. [PMID: 40142429 PMCID: PMC11946230 DOI: 10.3390/microorganisms13030536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Menaquinone-7 (MK-7) is recognized for its important biological activity, and Bacillus subtilis is the preferred strain for its fermentative production. However, the limited phenotypic diversity among high-yielding strains complicates the development of rapid screening methods. To address this, we utilized the effect of MK-7 on transmembrane potential to develop a high-throughput screening (HTS) strategy for efficiently identifying strains with improved MK-7 production. Among various membrane potential fluorescent dyes tested, Rhodamine 123 was selected for quantifying intracellular MK-7 levels due to its effective staining and minimal impact on cell growth. By optimizing pretreatment protocols and staining conditions, we established an HTS protocol that combines fluorescence-activated cell sorting with HPLC to identify strains with increased MK-7 production. A linear correlation was observed between mean MK-7 content and average fluorescence intensity (R2 = 0.9646). This approach was applied to mutant libraries generated through atmospheric room temperature plasma mutagenesis. After three cycles of mutagenesis and screening, the mutant AR03-27 was identified, showing an 85.65% increase in MK-7 yield compared to the original SJTU2 strain. Resequencing analysis revealed that the top three mutants contained mutations in genes related to membrane transport, suggesting their potential role in enhancing MK-7 yield.
Collapse
Affiliation(s)
- Lina Yang
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
| | - Can Tang
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
| | - Yan Cui
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
| | - Jianhua Zhang
- School of Agriculture and Biology, Bor S. Luh Food Safety Research Center, Shanghai Jiao Tong University, Shanghai 200240, China; (L.Y.); (C.T.); (Y.C.)
- NMPA Key Laboratory for Testing Technology of Pharmaceutical, Shanghai Institute for Food and Drug Control, Shanghai 201203, China
| |
Collapse
|
10
|
Shcheslavskiy VI, Shirmanova MV, Yashin KS, Rück AC, Skala MC, Becker W. Fluorescence Lifetime Imaging Techniques-A Review on Principles, Applications and Clinical Relevance. JOURNAL OF BIOPHOTONICS 2025:e202400450. [PMID: 39973086 DOI: 10.1002/jbio.202400450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 02/21/2025]
Abstract
This article gives an overview of the most frequently used fluorescence-lifetime imaging (FLIM) techniques, their capabilities, and typical applications. Starting from a general introduction to fluorescence and phosphorescence lifetime, we will show that the fluorescence lifetime or, more accurately, the fluorescence decay function of a fluorophore is a direct indicator of the interaction with its molecular environment. FLIM is therefore more than a simple contrast technique in microscopy-it is a technique of molecular imaging. FLIM techniques can be classified into time-domain and frequency-domain techniques, analogue and photon counting techniques, and scanning and wide-field techniques. Starting from an overview of these general technical principles we will describe the features and peculiarities of the different FLIM techniques in use. An extended section is dedicated to TCSPC FLIM, addressing unique capabilities that make the technique especially interesting to FLIM of biological systems.
Collapse
Affiliation(s)
- V I Shcheslavskiy
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
- Becker&Hickl GmbH, Berlin, Germany
| | - M V Shirmanova
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - K S Yashin
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - A C Rück
- Centre for Biomedical Research, Microscopy/Neurology Group, University Ulm, Ulm, Germany
| | - M C Skala
- Morgridge Institute for Research, Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| | - W Becker
- Becker&Hickl GmbH, Berlin, Germany
| |
Collapse
|
11
|
Alamsyah F, Firdausi N, Nugraheni SED, Fadhlurrahman AG, Nurhidayat L, Pratiwi R, Taruno WP. Effects of non-contact electric fields on the kidneys and livers of tumour-bearing rats. F1000Res 2025; 12:117. [PMID: 39931163 PMCID: PMC11809679 DOI: 10.12688/f1000research.110080.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Background A novel modality of cancer treatment based on exposure to non-contact electric fields called Electro-Capacitive Cancer Therapy (ECCT) has been developed. However, the effects of this modality on vital organs during cancer treatment have not been fully investigated. Therefore, this study aimed to investigate the effects of non-contact electric field exposure on kidney and liver structures. Methods Female rats were randomly divided into one control group and three treatment groups with six replications each. Animals were treated with 7,12-dimethylbenz[a]anthracene at a dose of 20 mg/kg body weight for mammary tumour induction. Animals were then exposed to electric fields (100 kHz, 50-60 V/m) for 10 hours a day for three weeks. Two kidney samples and two liver samples from different animals in each group were collected for observation of structural damage to the organs. Histopathological cross-sections of the kidneys and livers were made using the paraffin method and Hematoxylin-Eosin staining. Histological scoring used the post-examination masking method with 100 visual fields per group. Results There was no significant damages to the tubules, glomeruli, and interstitial of the kidneys, including congestion, after exposure to non-contact electric fields. In addition, healthy rats exposed to this electric field showed significantly lower renal interstitial damage. There was no significant cellular damage, congestion, and haemorrhage in the livers of all groups, except in the healthy rat group that showed significantly higher haemorrhage. Conclusions Exposure to non-contact electric fields may cause haemorrhage in the livers of healthy rats. However, in kidney tissue, exposure to this electric field was tolerable, and can even decrease the number of inflammations and haemorrhages in healthy rats.
Collapse
Affiliation(s)
- Firman Alamsyah
- Faculty of Science and Technology, Universitas Al-Azhar Indonesia, Jl. Sisingamangaraja, Jakarta, 12110, Indonesia
- Center for Medical Physics and Cancer Research, Ctech Labs Edwar Technology, Tangerang, Banten, 15143, Indonesia
| | - Nisrina Firdausi
- Research Center for Pharmaceutical Ingridients and Traditional Medicine, National Research and Innovation Agency Republic of Indonesia, Jl. Raya Jakarta-Bogor Km. 46, Cibinong West Java, 16911, Indonesia
| | - Subekti Evi Dwi Nugraheni
- Medical Laboratory, Universitas Pendidikan Indonesia, Jl. Dr. Setiabudhi No. 229, Bandung West Java, 40154, Indonesia
| | | | - Luthfi Nurhidayat
- Faculty of Biology, Universitas Gadjah Mada, Sleman, DI Yogyakarta, 55281, Indonesia
| | - Rarastoeti Pratiwi
- Faculty of Biology, Universitas Gadjah Mada, Sleman, DI Yogyakarta, 55281, Indonesia
| | - Warsito Purwo Taruno
- Center for Medical Physics and Cancer Research, Ctech Labs Edwar Technology, Tangerang, Banten, 15143, Indonesia
| |
Collapse
|
12
|
Ota A, Shiozaki A, Shimizu H, Kosuga T, Kudou M, Nishibeppu K, Ohashi T, Arita T, Konishi H, Komatsu S, Kubota T, Fujiwara H, Morinaga Y, Konishi E, Otsuji E. Functions and clinical significance of KCNB1 in esophageal squamous cell carcinoma. J Gastroenterol 2025:10.1007/s00535-025-02219-x. [PMID: 39893596 DOI: 10.1007/s00535-025-02219-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Voltage-gated potassium channel subfamily B member 1 (KCNB1) encodes the α-subunit of the Kv2.1 channel and mediates transmembrane potassium transport. The functions and mechanisms underlying KCNB1 activation have been examined in various cancer types; however, its role in esophageal squamous cell carcinoma (ESCC) remains unclear. Therefore, the present study investigated the involvement of KCNB1 in tumor progression and the clinicopathological significance of its expression in ESCC. METHODS Knockdown experiments using KCNB1 small interfering RNA were performed on the human ESCC cell lines, KYSE70 and TE5, and changes in cell proliferation, the cell cycle, apoptosis, migration, and invasion were assessed. Gene expression profiles were examined using a microarray analysis. An immunohistochemical (IHC) analysis was performed on 129 primary tumor samples from ESCC patients who underwent curative esophagectomy. RESULTS Cell proliferation, G2-M phase progression, migration, and invasion were inhibited, and apoptosis was induced in KCNB1-depleted cells. Microarray results showed that KCNB1 gene expression affected Ephrin receptor signaling by suppressing EPHB1, EPHB2, and ERK1/2 gene expression. IHC results revealed a relationship between high KCNB1 expression and a poor prognosis. High KCNB1 expression was extracted as an independent prognostic factor in a multivariate analysis of 5-year relapse-free survival in ESCC patients (p = 0.0197). CONCLUSIONS Cell proliferation is controlled by KCNB1 through its regulation of ERK1/2 gene expression via ephrin receptor signaling. A relationship was observed between KCNB1 and the prognosis of ESCC patients, indicating its potential as a biomarker for cancer progression and in targeted therapy for ESCC.
Collapse
Affiliation(s)
- Atsuki Ota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Michihiro Kudou
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yukiko Morinaga
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
13
|
Zhang G, Levin M. Bioelectricity is a universal multifaced signaling cue in living organisms. Mol Biol Cell 2025; 36:pe2. [PMID: 39873662 PMCID: PMC11809311 DOI: 10.1091/mbc.e23-08-0312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
The cellular electrical signals of living organisms were discovered more than a century ago and have been extensively investigated in the neuromuscular system. Neuronal depolarization and hyperpolarization are essential for our neuromuscular physiological and pathological functions. Bioelectricity is being recognized as an ancient, intrinsic, fundamental property of all living cells, and it is not limited to the neuromuscular system. Instead, emerging evidence supports a view of bioelectricity as an instructional signaling cue for fundamental cellular physiology, embryonic development, regeneration, and human diseases, including cancers. Here, we highlight the current understanding of bioelectricity and share our views on the challenges and perspectives.
Collapse
Affiliation(s)
- GuangJun Zhang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47906
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155
| |
Collapse
|
14
|
Borah R, Diez Clarke D, Upadhyay J, Monaghan MG. From innovation to clinic: Emerging strategies harnessing electrically conductive polymers to enhance electrically stimulated peripheral nerve repair. Mater Today Bio 2025; 30:101415. [PMID: 39816667 PMCID: PMC11733191 DOI: 10.1016/j.mtbio.2024.101415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
Peripheral nerve repair (PNR) is a major healthcare challenge due to the limited regenerative capacity of the nervous system, often leading to severe functional impairments. While nerve autografts are the gold standard, their implications are constrained by issues such as donor site morbidity and limited availability, necessitating innovative alternatives like nerve guidance conduits (NGCs). However, the inherently slow nerve growth rate (∼1 mm/day) and prolonged neuroinflammation, delay recovery even with the use of passive (no-conductive) NGCs, resulting in muscle atrophy and loss of locomotor function. Electrical stimulation (ES) has the ability to enhance nerve regeneration rate by modulating the innate bioelectrical microenvironment of nerve tissue while simultaneously fostering a reparative environment through immunoregulation. In this context, electrically conductive polymer (ECP)-based biomaterials offer unique advantages for nerve repair combining their flexibility, akin to traditional plastics, and mixed ionic-electronic conductivity, similar to ionically conductive nerve tissue, as well as their biocompatibility and ease of fabrication. This review focuses on the progress, challenges, and emerging techniques for integrating ECP based NGCs with ES for functional nerve regeneration. It critically evaluates the various approaches using ECP based scaffolds, identifying gaps that have hindered clinical translation. Key challenges discussed include designing effective 3D NGCs with high electroactivity, optimizing ES modules, and better understanding of immunoregulation during nerve repair. The review also explores innovative strategies in material development and wireless, self-powered ES methods. Furthermore, it emphasizes the need for non-invasive ES delivery methods combined with hybrid ECP based neural scaffolds, highlighting future directions for advancing preclinical and clinical translation. Together, ECP based NGCs combined with ES represent a promising avenue for advancing PNR and improving patient outcomes.
Collapse
Affiliation(s)
- Rajiv Borah
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Daniel Diez Clarke
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Jnanendra Upadhyay
- Department of Physics, Dakshin Kamrup College, Kamrup, Assam, 781125, India
| | - Michael G. Monaghan
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Research in Medical Devices, University of Galway, H91 W2TY Galway, Ireland
| |
Collapse
|
15
|
Yang H, Wang S, Zhao P, Yong Q, Huang Q, Al-Asmari F, Sameeh MY, Yang B, Zhang C, Wang X, Shi C. Thymoquinone as a potent antimicrobial agent against Yersinia enterocolitica: Mechanisms of action and potential food safety applications. Int J Food Microbiol 2025; 431:111071. [PMID: 39854959 DOI: 10.1016/j.ijfoodmicro.2025.111071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/26/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Yersinia enterocolitica (Y. enterocolitica) is a Gram-negative foodborne pathogen associated with potentially fatal diseases. Herein, the antibacterial activity and possible mechanism of thymoquinone (TQ) against Y. enterocolitica were explored. The minimum inhibitory concentration and the minimum bactericidal concentration of TQ against Y. enterocolitica were determined to be 0.10 and 0.20 mg/mL, respectively. Treatment with TQ increased the lag phase period and reduced the growth rate of Y. enterocolitica. TQ was also effective in preventing Y. enterocolitica contamination of pork. Treatment of Y. enterocolitica with TQ caused a decrease in intracellular ATP, membrane depolarization and an increase in the level of intracellular reactive oxygen species. In addition, TQ caused damage to DNA, a reduction in protein content, loss of membrane integrity and abnormal cell morphology. These findings suggest that TQ exhibits antimicrobial activity against Y. enterocolitica and may be a suitable compound to reduce Y. enterocolitica growth in food.
Collapse
Affiliation(s)
- Hui Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China; Northwest A&F University ShenZhen Research Institute, Shenzhen, Guangdong 518057, China
| | - Shuqi Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Pengyu Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qiyao Yong
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qianning Huang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fahad Al-Asmari
- Department of Food and Nutrition Sciences, College of Agricultural and Food Sciences, King Faisal University, Al Ahsa 31982, Saudi Arabia
| | - Manal Y Sameeh
- Department of chemistry, Al-Leith University College, Umm Al Qura University, Makkah 24831, Saudi Arabia
| | - Baowei Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chunling Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chao Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China; Northwest A&F University ShenZhen Research Institute, Shenzhen, Guangdong 518057, China.
| |
Collapse
|
16
|
Zamay TN, Zamay SS, Zamay GS, Kolovskaya OS, Kichkailo AS, Berezovski MV. Systemic Mechanisms of Ionic Regulation in Carcinogenesis. Cancers (Basel) 2025; 17:286. [PMID: 39858068 PMCID: PMC11764231 DOI: 10.3390/cancers17020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer is a complex disease characterized by uncontrolled cell proliferation at various levels, leading to tumor growth and spread. This review focuses on the role of ion homeostasis in cancer progression. It describes a model of ion-mediated regulation in both normal and cancerous cell proliferation. The main function of this system is to maintain the optimal number of cells in the body by regulating intra- and extracellular ion content. The review discusses the key points of ion regulation and their impact on tumor growth and spread during cancer development. It explains that normal levels of sodium, potassium, calcium, chloride, and hydrogen ions are regulated at different levels. Damage to ion transport mechanisms during carcinogenesis can lead to an increase in sodium cations and water content in cells, disrupting the balance of calcium and hydrogen ions. This, in turn, can lead to chromatin compaction reduction, gene overexpression, and instability at the epigenetic and genomic levels, resulting in increased cell proliferation and mutagenesis. Restoring normal ion balance can reduce the proliferative potential of both normal and tumor cell populations. The proposed model of systemic ionic regulation of proliferation aims to reconcile diverse data related to cell mitotic activity in various physiological conditions and explain tumor growth. Understanding the mechanisms behind pathological cell proliferation is important for developing new approaches to control ion homeostasis in the body, potentially leading to more effective cancer treatment and prevention.
Collapse
Affiliation(s)
- Tatiana N. Zamay
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Laboratory for Digital Controlled Drugs and Theranostics, Molecular Electronics Department, 660036 Krasnoyarsk, Russia; (S.S.Z.); (G.S.Z.); (O.S.K.); (A.S.K.)
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University Laboratory for Biomolecular and Medical Technologies, 660022 Krasnoyarsk, Russia
| | - Sergey S. Zamay
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Laboratory for Digital Controlled Drugs and Theranostics, Molecular Electronics Department, 660036 Krasnoyarsk, Russia; (S.S.Z.); (G.S.Z.); (O.S.K.); (A.S.K.)
| | - Galina S. Zamay
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Laboratory for Digital Controlled Drugs and Theranostics, Molecular Electronics Department, 660036 Krasnoyarsk, Russia; (S.S.Z.); (G.S.Z.); (O.S.K.); (A.S.K.)
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University Laboratory for Biomolecular and Medical Technologies, 660022 Krasnoyarsk, Russia
| | - Olga S. Kolovskaya
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Laboratory for Digital Controlled Drugs and Theranostics, Molecular Electronics Department, 660036 Krasnoyarsk, Russia; (S.S.Z.); (G.S.Z.); (O.S.K.); (A.S.K.)
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University Laboratory for Biomolecular and Medical Technologies, 660022 Krasnoyarsk, Russia
| | - Anna S. Kichkailo
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Laboratory for Digital Controlled Drugs and Theranostics, Molecular Electronics Department, 660036 Krasnoyarsk, Russia; (S.S.Z.); (G.S.Z.); (O.S.K.); (A.S.K.)
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University Laboratory for Biomolecular and Medical Technologies, 660022 Krasnoyarsk, Russia
| | - Maxim V. Berezovski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| |
Collapse
|
17
|
Costa FP, Tuszynski J, Iemma AF, Trevizan WA, Wiedenmann B, Schöll E. External low energy electromagnetic fields affect heart dynamics: surrogate for system synchronization, chaos control and cancer patient's health. FRONTIERS IN NETWORK PHYSIOLOGY 2025; 4:1525135. [PMID: 39830523 PMCID: PMC11739291 DOI: 10.3389/fnetp.2024.1525135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025]
Abstract
All cells in the human body, including cancer cells, possess specific electrical properties crucial for their functions. These properties are notably different between normal and cancerous cells. Cancer cells are characterized by autonomous oscillations and damped electromagnetic field (EMF) activation. Cancer reduces physiological variability, implying a systemic disconnection that desynchronizes bodily systems and their inherent random processes. The dynamics of heart rate, in this context, could reflect global physiological network instability in the sense of entrainment. Using a medical device that employs an active closed-loop system, such as administering specifically modulated EMF frequencies at targeted intervals and at low energies, we can evaluate the periodic oscillations of the heart. This procedure serves as a closed-loop control mechanism leading to a temporary alteration in plasma membrane ionic flow and the heart's periodic oscillation dynamics. The understanding of this phenomenon is supported by computer simulations of a mathematical model, which are validated by experimental data. Heart dynamics can be quantified using difference logistic equations, and it correlates with improved overall survival rates in cancer patients.
Collapse
Affiliation(s)
| | - Jack Tuszynski
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Politecnico di Torino, Turin, Italy
| | - Antonio F. Iemma
- Mathematical and Statistics, Autem Therapeutics, Hanover, NH, United States
| | - Willian A. Trevizan
- Physics and Mathematical Modeling, Autem Therapeutics, Hanover, NH, United States
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eckehard Schöll
- Institut für Theoretische Physik, Technische Universität Berlin, Berlin, Germany
| |
Collapse
|
18
|
Hou F, Wang R, Wang X, Dong J, Yu T, Deng Q, Wang Z. A multi-functional platform for stimulating and recording electrical responses of SH-SY5Y cells. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:275-283. [PMID: 39611717 DOI: 10.1039/d4ay01515k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
In recent years, multifunctional cell regulation on a single chip has become an imperative need for cell research. In this study, a novel multi-functional micro-platform integrating wireless electrical stimulation, mechanical stimulation and electrical response recording of cells was proposed. Controlling cell fate by photoexcited radio stimulation of cells on photosensitive films can precisely orchestrate biological activities. This is the first report of combining hydrogenated amorphous silicon (a-Si:H) photosensitive films and a 532 nm green laser for cell stimulation and electrical response recording. Remote wireless electrical stimulation of nerve cells through photoelectric effects based on photosensitive films evoked a change in membrane potential and promoted the neurite growth and neuronal differentiation. These effects were confirmed in a cell model of the human neuroblastoma cell line. The electrical response of cells demonstrated that the photocurrent generated by laser irradiation of the photosensitive film induced a redistribution of ions inside and outside the cell. Furthermore, a mechanical stimulus was applied to cells using a probe placed above them. The chip was used as a signal output to simultaneously obtain the electrical response of cells. The ability of photosensitive films to precisely excite cellular activity offers a novel prospect for wireless electrical stimulation. This work provides a promising strategy for the design of multi-functional biological devices based on a single chip.
Collapse
Affiliation(s)
- Fengyan Hou
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Rui Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Xia Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Jianjun Dong
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Tianzhu Yu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Qiuyang Deng
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
- Institute of Biomedical and Environmental Science & Technology, Institute for Research in Applicable Computing, University of Bedfordshire, Luton LU1 3JU, UK
| |
Collapse
|
19
|
Lissek T. Cancer memory as a mechanism to establish malignancy. Biosystems 2025; 247:105381. [PMID: 39701407 DOI: 10.1016/j.biosystems.2024.105381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/18/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
Cancers during oncogenic progression hold information in epigenetic memory which allows flexible encoding of malignant phenotypes and more rapid reaction to the environment when compared to purely mutation-based clonal evolution mechanisms. Cancer memory describes a proposed mechanism by which complex information such as metastasis phenotypes, therapy resistance and interaction patterns with the tumor environment might be encoded at multiple levels via mechanisms used in memory formation in the brain and immune system (e.g. single-cell epigenetic changes and distributed state modifications in cellular ensembles). Carcinogenesis might hence be the result of physiological multi-level learning mechanisms unleashed by defined heritable oncogenic changes which lead to tumor-specific loss of goal state integration into the whole organism. The formation of cancer memories would create and bind new levels of individuality within the host organism into the entity we call cancer. Translational implications of cancer memory are that cancers could be engaged at higher organizational levels (e.g. be "trained" for memory extinction) and that compounds that are known to interfere with memory processes could be investigated for their potential to block cancer memory formation or recall. It also suggests that diagnostic measures should extend beyond sequencing approaches to functional diagnosis of cancer physiology.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
20
|
Roy D, Michalet X, Miller EW, Bharadwaj K, Weiss S. Towards measurements of absolute membrane potential in Bacillus subtilis using fluorescence lifetime. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598880. [PMID: 38915670 PMCID: PMC11195253 DOI: 10.1101/2024.06.13.598880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Membrane potential (MP) changes can provide a simple readout of bacterial functional and metabolic state or stress levels. While several optical methods exist for measuring fast changes in MP in excitable cells, there is a dearth of such methods for absolute and precise measurements of steady-state membrane potentials (MPs) in bacterial cells. Conventional electrode-based methods for the measurement of MP are not suitable for calibrating optical methods in small bacterial cells. While optical measurement based on Nernstian indicators have been successfully used, they do not provide absolute or precise quantification of MP or its changes. We present a novel, calibrated MP recording approach to address this gap. In this study, we used a fluorescence lifetime-based approach to obtain a single-cell resolved distribution of the membrane potential and its changes upon extracellular chemical perturbation in a population of bacterial cells for the first time. Our method is based on (i) a unique VoltageFluor (VF) optical transducer, whose fluorescence lifetime varies as a function of MP via photoinduced electron transfer (PeT) and (ii) a quantitative phasor-FLIM analysis for high-throughput readout. This method allows MP changes to be easily visualized, recorded and quantified. By artificially modulating potassium concentration gradients across the membrane using an ionophore, we have obtained a Bacillus subtilis-specific MP versus VF lifetime calibration and estimated the MP for unperturbed B. subtilis cells to be -65 mV (in MSgg), 127 mV (in M9) and that for chemically depolarized cells as -14 mV (in MSgg). We observed a population level MP heterogeneity of ~6-10 mV indicating a considerable degree of diversity of physiological and metabolic states among individual cells. Our work paves the way for deeper insights into bacterial electrophysiology and bioelectricity research.
Collapse
Affiliation(s)
- Debjit Roy
- UCLA-DOE Institute for Genomics and Proteomics, Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Xavier Michalet
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, CA 90095, USA
- California Nano Systems Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Evan W. Miller
- Departments of Chemistry, Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California at Berkeley, CA 94720, USA
| | - Kiran Bharadwaj
- UCLA-DOE Institute for Genomics and Proteomics, Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Shimon Weiss
- UCLA-DOE Institute for Genomics and Proteomics, Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Department of Physiology, University of California at Los Angeles, Los Angeles, CA 90095, USA
- California Nano Systems Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 52900, Israel
| |
Collapse
|
21
|
Costa FP, Wiedenmann B, Schöll E, Tuszynski J. Emerging cancer therapies: targeting physiological networks and cellular bioelectrical differences with non-thermal systemic electromagnetic fields in the human body - a comprehensive review. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1483401. [PMID: 39720338 PMCID: PMC11666389 DOI: 10.3389/fnetp.2024.1483401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024]
Abstract
A steadily increasing number of publications support the concept of physiological networks, and how cellular bioelectrical properties drive cell proliferation and cell synchronization. All cells, especially cancer cells, are known to possess characteristic electrical properties critical for physiological behavior, with major differences between normal and cancer cell counterparts. This opportunity can be explored as a novel treatment modality in Oncology. Cancer cells exhibit autonomous oscillations, deviating from normal rhythms. In this context, a shift from a static view of cellular processes is required for a better understanding of the dynamic connections between cellular metabolism, gene expression, cell signaling and membrane polarization as states in constant flux in realistic human models. In oncology, radiofrequency electromagnetic fields have produced sustained responses and improved quality of life in cancer patients with minimal side effects. This review aims to show how non-thermal systemic radiofrequency electromagnetic fields leads to promising therapeutic responses at cellular and tissue levels in humans, supporting this newly emerging cancer treatment modality with early favorable clinical experience specifically in advanced cancer.
Collapse
Affiliation(s)
| | | | - Eckehard Schöll
- Institut für Theoretische Physik, Technische Universität Berlin, Berlin, Germany
| | - Jack Tuszynski
- Department of Physics, University of Alberta, Edmonton, AB, Canada
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Turin, Italy
- Department of Data Science and Engineering, The Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
22
|
Shrivastava A, Kumar A, Aggarwal LM, Pradhan S, Choudhary S, Ashish A, Kashyap K, Mishra S. Evolution of Bioelectric Membrane Potentials: Implications in Cancer Pathogenesis and Therapeutic Strategies. J Membr Biol 2024; 257:281-305. [PMID: 39183198 DOI: 10.1007/s00232-024-00323-2] [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: 05/30/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Electrophysiology typically deals with the electrical properties of excitable cells like neurons and muscles. However, all other cells (non-excitable) also possess bioelectric membrane potentials for intracellular and extracellular communications. These membrane potentials are generated by different ions present in fluids available in and outside the cell, playing a vital role in communication and coordination between the cell and its organelles. Bioelectric membrane potential variations disturb cellular ionic homeostasis and are characteristic of many diseases, including cancers. A rapidly increasing interest has emerged in sorting out the electrophysiology of cancer cells. Compared to healthy cells, the distinct electrical properties exhibited by cancer cells offer a unique way of understanding cancer development, migration, and progression. Decoding the altered bioelectric signals influenced by fluctuating electric fields benefits understanding cancer more closely. While cancer research has predominantly focussed on genetic and molecular traits, the delicate area of electrophysiological characteristics has increasingly gained prominence. This review explores the historical exploration of electrophysiology in the context of cancer cells, shedding light on how alterations in bioelectric membrane potentials, mediated by ion channels and gap junctions, contribute to the pathophysiology of cancer.
Collapse
Affiliation(s)
- Anju Shrivastava
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India.
| | - Amit Kumar
- Department of Anatomy, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Lalit Mohan Aggarwal
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Satyajit Pradhan
- Radiation Oncology, Mahamana Pandit Madhan Mohan Malaviya Cancer Centre, Varanasi, India
| | - Sunil Choudhary
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ashish Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Keshav Kashyap
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Shivani Mishra
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
23
|
Boutonnet M, Bünemann M, Perroy J. The voltage sensitivity of G-protein coupled receptors: Unraveling molecular mechanisms and physiological implications. Pharmacol Ther 2024; 264:108741. [PMID: 39489434 DOI: 10.1016/j.pharmthera.2024.108741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/11/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
In the landscape of proteins controlled by membrane voltage (Vm), like voltage-gated ionotropic channels, the emergence of the voltage sensitivity within the vast family of G-protein coupled receptors (GPCRs) marked a significant milestone at the onset of the 21st century. Since its discovery, extensive research has been devoted to understanding the intricate relationship between Vm and GPCRs. Approximately 30 GPCRs out of a family comprising more than 800 receptors have been implicated in Vm-dependent positive and negative regulation. GPCRs stand out as the quintessential regulators of synaptic transmission in neurons, where they encounter substantial variations in Vm. However, the molecular mechanism underlying the Vm sensor of GPCRs remains enigmatic, hindered by the scarcity of mutant GPCRs insensitive to Vm yet functionally intact, impeding a comprehensive understanding of this unique property in physiology. Nevertheless, two decades of dedicated research have furnished numerous insights into the molecular aspects of GPCR Vm-sensing, accompanied by recently proposed physiological roles as well as pharmacological potential, which we encapsulate in this review. The Vm sensitivity of GPCRs emerges as a pivotal attribute, shedding light on previously unforeseen roles in synaptic transmission and extending beyond, underscoring its significance in cellular signaling and physiological processes.
Collapse
Affiliation(s)
- Marin Boutonnet
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Julie Perroy
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
24
|
Minnaar CA, Szigeti GP, Szasz A. The Synergy of Thermal and Non-Thermal Effects in Hyperthermic Oncology. Cancers (Basel) 2024; 16:3908. [PMID: 39682096 DOI: 10.3390/cancers16233908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Modulated electro-hyperthermia (mEHT) is unique due to its combination of thermal and non-thermal effects. METHOD This report summarizes the literature on the effects of mEHT observed in vitro and in vivo. RESULTS The thermal and electrical heterogeneity of tissues allows the radiofrequency signal to selectively target malignant tissue. The applied modulation appears to activate various apoptotic pathways, predominantly leading to immunogenic cell death (ICD). ICD promotes the release of damage-associated molecular patterns, potentially producing tumour-specific antigen-presenting cells. This abscopal-type effect may target distant metastases while treating the primary tumour locally. This immune memory effect is like vaccination mechanisms. CONCLUSIONS The application of mEHT has the potential to expand from local to systemic disease, enabling the simultaneous treatment of micro- and macro-metastases.
Collapse
Affiliation(s)
- Carrie Anne Minnaar
- Department of Radiation Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Gyula Peter Szigeti
- John von Neumann Faculty of Informatics, Óbuda University, 1034 Budapest, Hungary
- MedTech Innovation and Education Center, University Research and Innovation Center, Óbuda University, 1034 Budapest, Hungary
| | - Andras Szasz
- Department of Biotechnics, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| |
Collapse
|
25
|
Yu H, Nishio H, Barbi J, Mitchell-Flack M, Vignali PDA, Zheng Y, Lebid A, Chang KY, Fu J, Higgins M, Huang CT, Zhang X, Li Z, Blosser L, Tam A, Drake C, Pardoll D. Neurotrophic factor Neuritin modulates T cell electrical and metabolic state for the balance of tolerance and immunity. eLife 2024; 13:RP96812. [PMID: 39565188 DOI: 10.7554/elife.96812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Abstract
The adaptive T cell response is accompanied by continuous rewiring of the T cell's electric and metabolic state. Ion channels and nutrient transporters integrate bioelectric and biochemical signals from the environment, setting cellular electric and metabolic states. Divergent electric and metabolic states contribute to T cell immunity or tolerance. Here, we report in mice that neuritin (Nrn1) contributes to tolerance development by modulating regulatory and effector T cell function. Nrn1 expression in regulatory T cells promotes its expansion and suppression function, while expression in the T effector cell dampens its inflammatory response. Nrn1 deficiency in mice causes dysregulation of ion channel and nutrient transporter expression in Treg and effector T cells, resulting in divergent metabolic outcomes and impacting autoimmune disease progression and recovery. These findings identify a novel immune function of the neurotrophic factor Nrn1 in regulating the T cell metabolic state in a cell context-dependent manner and modulating the outcome of an immune response.
Collapse
Affiliation(s)
- Hong Yu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Hiroshi Nishio
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Joseph Barbi
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Marisa Mitchell-Flack
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Paolo D A Vignali
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ying Zheng
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Andriana Lebid
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Kwang-Yu Chang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Juan Fu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Makenzie Higgins
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ching-Tai Huang
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Xuehong Zhang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Zhiguang Li
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Lee Blosser
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ada Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Charles Drake
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Drew Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
26
|
Zimmermann J, Farooqi AR, van Rienen U. Electrical stimulation for cartilage tissue engineering - A critical review from an engineer's perspective. Heliyon 2024; 10:e38112. [PMID: 39416819 PMCID: PMC11481755 DOI: 10.1016/j.heliyon.2024.e38112] [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: 04/05/2024] [Revised: 08/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Cartilage has a limited intrinsic healing capacity. Hence, cartilage degradation and lesions pose a huge clinical challenge, particularly in an ageing society. Osteoarthritis impacts a significant number of the population and requires the development of repair and tissue engineering methods for hyaline articular cartilage. In this context, electrical stimulation has been investigated for more than 50 years already. Yet, no well-established clinical therapy to treat osteoarthritis by means of electrical stimulation exists. We argue that one reason is the lack of replicability of electrical stimulation devices from a technical perspective together with lacking hypotheses of the biophysical mechanism. Hence, first, the electrical stimulation studies reported in the context of cartilage tissue engineering with a special focus on technical details are summarized. Then, an experimental and numerical approach is discussed to make the electrical stimulation experiments replicable. Finally, biophysical hypotheses have been reviewed on the interaction of electric fields and cells that are relevant for cartilage tissue engineering. With that, the aim is to inspire future research to enable clinical electrical stimulation therapies to fight osteoarthritis.
Collapse
Affiliation(s)
- Julius Zimmermann
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
| | - Abdul Razzaq Farooqi
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Electronic Engineering, Faculty of Engineering, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Ursula van Rienen
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Ageing of Individuals and Society, Interdisciplinary Faculty, University of Rostock, 18051 Rostock, Germany
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany
| |
Collapse
|
27
|
Maqoud F, Simone L, Tricarico D, Camerino GM, Antonacci M, Nicchia GP. The Functional Interaction of KATP and BK Channels with Aquaporin-4 in the U87 Glioblastoma Cell. Biomedicines 2024; 12:1891. [PMID: 39200356 PMCID: PMC11351575 DOI: 10.3390/biomedicines12081891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
K+ channels do play a role in cell shape changes observed during cell proliferation and apoptosis. Research suggested that the dynamics of the aggregation of Aquaporin-4 (AQP4) into AQP4-OAP isoforms can trigger cell shape changes in malignant glioma cells. Here, we investigated the relationship between AQP4 and some K+ channels in the malignant glioma U87 line. The U87 cells transfected with the human M1-AQP4 and M23-AQP4 isoforms were investigated for morphology, the gene expression of KCNJ8, KCNJ11, ABCC8, ABCC9, KCNMA1, and Cyclin genes by RT-PCR, recording the whole-cell K+ ion currents by patch-clamp experiments. AQP4 aggregation into OAPs increases the plasma membrane functional expression of the Kir6.2 and SUR2 subunits of the KATP channels and of the KCNMA1 of the BK channels in U87 cells leading to a large increase in inward and outward K+ ion currents. These changes were associated with changes in morphology, with a decrease in cell volume in the U87 cells and an increase in the ER density. These U87 cells accumulate in the mitotic and G2 cell cycle. The KATP channel blocker zoledronic acid reduced cell proliferation in both M23 AQP4-OAP and M1 AQP4-tetramer-transfected cells, leading to early and late apoptosis, respectively. The BK channel sustains the efflux of K+ ions associated with the M23 AQP4-OAP expression in the U87 cells, but it is downregulated in the M1 AQP4-tetramer cells. The KATP channels are effective in the M1 AQP4-tetramer and M23 AQP4-OAP cells. Zoledronic acid can be effective in targeting pathogenic M1 AQP4-tetramer cell phenotypes inhibiting KATP channels and inducing early apoptosis.
Collapse
Affiliation(s)
- Fatima Maqoud
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.M.); (G.M.C.); (M.A.)
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology Saverio de Bellis, I.R.C.C.S. Research Hospital, 70013 Castellana Grotte, Italy
| | - Laura Simone
- Cancer Stem Cells Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, snc, 71013 San Giovanni Rotondo, Italy;
| | - Domenico Tricarico
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.M.); (G.M.C.); (M.A.)
| | - Giulia Maria Camerino
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.M.); (G.M.C.); (M.A.)
| | - Marina Antonacci
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.M.); (G.M.C.); (M.A.)
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
| |
Collapse
|
28
|
Tota M, Jonderko L, Witek J, Novickij V, Kulbacka J. Cellular and Molecular Effects of Magnetic Fields. Int J Mol Sci 2024; 25:8973. [PMID: 39201657 PMCID: PMC11354277 DOI: 10.3390/ijms25168973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Recently, magnetic fields (MFs) have received major attention due to their potential therapeutic applications and biological effects. This review provides a comprehensive analysis of the cellular and molecular impacts of MFs, with a focus on both in vitro and in vivo studies. We investigate the mechanisms by which MFs influence cell behavior, including modifications in gene expression, protein synthesis, and cellular signaling pathways. The interaction of MFs with cellular components such as ion channels, membranes, and the cytoskeleton is analyzed, along with their effects on cellular processes like proliferation, differentiation, and apoptosis. Molecular insights are offered into how MFs modulate oxidative stress and inflammatory responses, which are pivotal in various pathological conditions. Furthermore, we explore the therapeutic potential of MFs in regenerative medicine, cancer treatment, and neurodegenerative diseases. By synthesizing current findings, this article aims to elucidate the complex bioeffects of MFs, thereby facilitating their optimized application in medical and biotechnological fields.
Collapse
Affiliation(s)
- Maciej Tota
- Student Research Group № K148, Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Laura Jonderko
- Student Research Group № K148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland; (L.J.); (J.W.)
| | - Julia Witek
- Student Research Group № K148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland; (L.J.); (J.W.)
| | - Vitalij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, LT-03227 Vilnius, Lithuania;
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08410 Vilnius, Lithuania
| | - Julita Kulbacka
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08410 Vilnius, Lithuania
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wrocław, Poland
| |
Collapse
|
29
|
Karal MAS, Billah MM, Nasrin T, Moniruzzaman M. Interaction of anionic Fe 3O 4 nanoparticles with lipid vesicles: a review on deformation and poration under various conditions. RSC Adv 2024; 14:25986-26001. [PMID: 39161454 PMCID: PMC11331399 DOI: 10.1039/d4ra05686h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024] Open
Abstract
This review focuses on the deformation and poration of lipid vesicles caused by the interaction of anionic magnetite nanoparticles (MNPs). Effects of various factors, such as surface charge density, salt and sugar concentrations in buffer, membrane cholesterol content, polymer-grafted phospholipid, and membrane potential have been discussed for the interaction of MNPs with lipid vesicles. To quantify these effects on the vesicles, compactness, fraction of deformation and poration, dynamics of membrane permeation, and kinetics of membrane permeation have been critically evaluated. The review explores the potential advancements as well as future directions of the research field in the biomedical application of MNPs.
Collapse
Affiliation(s)
- Mohammad Abu Sayem Karal
- Department of Physics, Bangladesh University of Engineering and Technology Dhaka 1000 Bangladesh +880-2-58613046 +880-2-9665613
| | - Md Masum Billah
- Department of Physics, Jashore University of Science and Technology Jashore 7408 Bangladesh +880-2-42142012 +880-242142046
| | - Tawfika Nasrin
- Department of Physics, Bangladesh University of Engineering and Technology Dhaka 1000 Bangladesh +880-2-58613046 +880-2-9665613
| | - Md Moniruzzaman
- Department of Physics, Bangladesh University of Engineering and Technology Dhaka 1000 Bangladesh +880-2-58613046 +880-2-9665613
| |
Collapse
|
30
|
Hughes MP, Clarke KSP, Hoque R, Griffiths OV, Kruchek EJ, Johnson MP, Tariq MH, Kohli N, Lewis R, Labeed FH. Label-free, non-contact determination of resting membrane potential using dielectrophoresis. Sci Rep 2024; 14:18477. [PMID: 39122771 PMCID: PMC11316104 DOI: 10.1038/s41598-024-69000-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Measurement of cellular resting membrane potential (RMP) is important in understanding ion channels and their role in regulation of cell function across a wide range of cell types. However, methods available for the measurement of RMP (including patch clamp, microelectrodes, and potential-sensitive fluorophores) are expensive, slow, open to operator bias, and often result in cell destruction. We present non-contact, label-free membrane potential estimation which uses dielectrophoresis to determine the cytoplasm conductivity slope as a function of medium conductivity. By comparing this to patch clamp data available in the literature, we have demonstratet the accuracy of this approach using seven different cell types, including primary suspension cells (red blood cells, platelets), cultured suspension cells (THP-1), primary adherent cells (chondrocytes, human umbilical mesenchymal stem cells), and adherent (HeLa) and suspension (Jurkat) cancer cell lines. Analysis of the effect of ion channel inhibitors suggests the effects of pharmaceutical agents (TEA on HeLa; DMSO and neuraminidase on red blood cells) can also be measured. Comparison with published values of membrane potential suggest that the differences between our estimates and values recorded by patch clamp are accurate to within published margins of error. The method is low-cost, non-destructive, operator-independent and label-free, and has previously been shown to allow cells to be recovered after measurement.
Collapse
Affiliation(s)
- Michael Pycraft Hughes
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE.
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, UAE.
| | - Krista S P Clarke
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Rashedul Hoque
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Oreoluwa V Griffiths
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Emily J Kruchek
- Centre for Biomedical Engineering, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Matthew P Johnson
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Muhammad Hamza Tariq
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Nupur Kohli
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Rebecca Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Fatima H Labeed
- Department of Biology, United Arab Emirates University, Al Ain, UAE.
| |
Collapse
|
31
|
Yu H, Nishio H, Barbi J, Mitchell-Flack M, Vignali PDA, Zheng Y, Lebid A, Chang KY, Fu J, Higgins M, Huang CT, Zhang X, Li Z, Blosser L, Tam A, Drake CG, Pardoll DM. Neurotrophic factor Neuritin modulates T cell electrical and metabolic state for the balance of tolerance and immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578284. [PMID: 38352414 PMCID: PMC10862906 DOI: 10.1101/2024.01.31.578284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The adaptive T cell response is accompanied by continuous rewiring of the T cell's electric and metabolic state. Ion channels and nutrient transporters integrate bioelectric and biochemical signals from the environment, setting cellular electric and metabolic states. Divergent electric and metabolic states contribute to T cell immunity or tolerance. Here, we report that neuritin (Nrn1) contributes to tolerance development by modulating regulatory and effector T cell function. Nrn1 expression in regulatory T cells promotes its expansion and suppression function, while expression in the T effector cell dampens its inflammatory response. Nrn1 deficiency causes dysregulation of ion channel and nutrient transporter expression in Treg and effector T cells, resulting in divergent metabolic outcomes and impacting autoimmune disease progression and recovery. These findings identify a novel immune function of the neurotrophic factor Nrn1 in regulating the T cell metabolic state in a cell context-dependent manner and modulating the outcome of an immune response.
Collapse
Affiliation(s)
- Hong Yu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hiroshi Nishio
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Joseph Barbi
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY14263, USA
| | - Marisa Mitchell-Flack
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Paolo D A Vignali
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: University of Pittsburgh, Carnegie Mellon
| | - Ying Zheng
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andriana Lebid
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kwang-Yu Chang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Juan Fu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Makenzie Higgins
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ching-Tai Huang
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Infectious Diseases, Department of Medicine, Chang Gung Memorial Hospital, Taiwan
| | - Xuehong Zhang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian 116044, China
| | - Zhiguang Li
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian 116044, China
| | - Lee Blosser
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ada Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles G Drake
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Current address: Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| | - Drew M Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Immunology and Hematopoiesis Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Nakajima T. Unification of Mind and Matter through Hierarchical Extension of Cognition: A New Framework for Adaptation of Living Systems. ENTROPY (BASEL, SWITZERLAND) 2024; 26:660. [PMID: 39202130 PMCID: PMC11354174 DOI: 10.3390/e26080660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024]
Abstract
Living systems (LSs) must solve the problem of adapting to their environment by identifying external states and acting appropriately to maintain external relationships and internal order for survival and reproduction. This challenge is akin to the philosophical enigma of how the self can escape solipsism. In this study, a comprehensive model is developed to address the adaptation problem. LSs are composed of material entities capable of detecting their external states. This detection is conceptualized as "cognition", a state change in relation to its external states. This study extends the concept of cognition to include three hierarchical levels of the world: physical, chemical, and semiotic cognitions, with semiotic cognition being closest to the conventional meaning of cognition. This radical extension of the cognition concept to all levels of the world provides a monistic model named the cognizers system model, in which mind and matter are unified as a single entity, the "cognizer". During evolution, LSs invented semiotic cognition based on physical and chemical cognitions to manage the probability distribution of events that occur to them. This study proposes a theoretical model in which semiotic cognition is an adaptive process wherein the inverse causality operation produces particular internal states as symbols that signify hidden external states. This operation makes LSs aware of the external world.
Collapse
|
33
|
Jiang T, Yu F, Zhou Y, Li R, Zheng M, Jiang Y, Li Z, Pan J, Ouyang N. Synergistic effect of ultrasound and reinforced electrical environment by bioinspired periosteum for enhanced osteogenesis via immunomodulation of macrophage polarization through Piezo1. Mater Today Bio 2024; 27:101147. [PMID: 39045313 PMCID: PMC11263955 DOI: 10.1016/j.mtbio.2024.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/18/2024] [Accepted: 07/03/2024] [Indexed: 07/25/2024] Open
Abstract
The periosteum plays a vital role in repairing bone defects. Researchers have demonstrated the existence of electrical potential in the periosteum and native bone, indicating that electrical signals are essential for functional bone regeneration. However, the clinical use of external electrical treatments has been limited due to their inconvenience and inefficacy. As an alternative, low-intensity pulsed ultrasound (LIPUS) is a noninvasive form of physical therapy that enhances bone regeneration. Furthermore, the wireless activation of piezoelectric biomaterials through ultrasound stimulation would generate electric charges precisely at the defect area, compensating for the insufficiency of external electrical stimulation and potentially promoting bone regeneration through the synergistic effect of mechanical and electrical stimulation. However, the optimal integration of LIPUS with an appropriate piezoelectric periosteum is yet to be explored. Herein, the BaTiO3/multiwalled-carbon nanotubes/collagen (BMC) membranes have been fabricated, possessing physicochemical properties including improved surface hydrophilicity, enhanced mechanical performance, ideal piezoelectricity, and outstanding biocompatibility, all of which are conducive to bone regeneration. When combined with LIPUS, the endogenous electrical microenvironment of native bone was recreated. After that, the wireless-generated electrical signals, along with the mechanical signals induced by LIPUS, were transferred to macrophages and activated Ca2+ influx through Piezo1. Ultimately, the regenerative effect of the BMC membrane with LIPUS stimulation (BMC + L) was confirmed in a mouse cranial defect model. Together, this research presents a co-engineering strategy that involves fabricating a novel biomimetic periosteum and utilizing the synergistic effect of ultrasound to enhance bone regeneration, which is achieved through the reinforcement of the electrical environment and the immunomodulation of macrophage polarization.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
- Oral Bioengineering Lab, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Fei Yu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yuqi Zhou
- Department of Stomatology, Weifang People's Hospital Stomatological Hospital, Weifang, 261041, China
| | - Ruomei Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
- Oral Bioengineering Lab, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Mengting Zheng
- Oral Bioengineering Lab, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Yangyang Jiang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Zhenxia Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Jun Pan
- School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| | - Ningjuan Ouyang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| |
Collapse
|
34
|
Liu H, Weng J, Huang CLH, Jackson AP. Voltage-gated sodium channels in cancers. Biomark Res 2024; 12:70. [PMID: 39060933 PMCID: PMC11282680 DOI: 10.1186/s40364-024-00620-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Voltage-gated sodium channels (VGSCs) initiate action potentials in electrically excitable cells and tissues. Surprisingly, some VGSC genes are aberrantly expressed in a variety of cancers, derived from "non-excitable" tissues that do not generate classic action potentials, showing potential as a promising pharmacological target for cancer. Most of the previous review articles on this topic are limited in scope, and largely unable to provide researchers with a comprehensive understanding of the role of VGSC in cancers. Here, we review the expression patterns of all nine VGSC α-subunit genes (SCN1A-11A) and their four regulatory β-subunit genes (SCN1B-4B). We reviewed data from the Cancer Genome Atlas (TCGA) database, complemented by an extensive search of the published papers. We summarized and reviewed previous independent studies and analyzed the VGSC genes in the TCGA database regarding the potential impact of VGSC on cancers. A comparison between evidence gathered from independent studies and data review was performed to scrutinize potential biases in prior research and provide insights into future research directions. The review supports the view that VGSCs play an important role in diagnostics as well as therapeutics of some cancer types, such as breast, colon, prostate, and lung cancer. This paper provides an overview of the current knowledge on voltage-gated sodium channels in cancer, as well as potential avenues for further research. While further research is required to fully understand the role of VGSCs in cancer, the potential of VGSCs for clinical diagnosis and treatment is promising.
Collapse
Affiliation(s)
- Hengrui Liu
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK.
| | - Jieling Weng
- Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Christopher L-H Huang
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Antony P Jackson
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK.
| |
Collapse
|
35
|
Pukkanasut P, Jaskula-Sztul R, Gomora JC, Velu SE. Therapeutic targeting of voltage-gated sodium channel Na V1.7 for cancer metastasis. Front Pharmacol 2024; 15:1416705. [PMID: 39045054 PMCID: PMC11263763 DOI: 10.3389/fphar.2024.1416705] [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: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/25/2024] Open
Abstract
This review focuses on the expression and function of voltage-gated sodium channel subtype NaV1.7 in various cancers and explores its impact on the metastasis driving cell functions such as proliferation, migration, and invasiveness. An overview of its structural characteristics, drug binding sites, inhibitors and their likely mechanisms of action are presented. Despite the lack of clarity on the precise mechanism by which NaV1.7 contributes to cancer progression and metastasis; many studies have suggested a connection between NaV1.7 and proteins involved in multiple signaling pathways such as PKA and EGF/EGFR-ERK1/2. Moreover, the functional activity of NaV1.7 appears to elevate the expression levels of MACC1 and NHE-1, which are controlled by p38 MAPK activity, HGF/c-MET signaling and c-Jun activity. This cascade potentially enhances the secretion of extracellular matrix proteases, such as MMPs which play critical roles in cell migration and invasion activities. Furthermore, the NaV1.7 activity may indirectly upregulate Rho GTPases Rac activity, which is critical for cytoskeleton reorganization, cell adhesion, and actin polymerization. The relationship between NaV1.7 and cancer progression has prompted researchers to investigate the therapeutic potential of targeting NaV1.7 using inhibitors. The positive outcome of such studies resulted in the discovery of several inhibitors with the ability to reduce cancer cell migration, invasion, and tumor growth underscoring the significance of NaV1.7 as a promising pharmacological target for attenuating cancer cell proliferation and metastasis. The research findings summarized in this review suggest that the regulation of NaV1.7 expression and function by small molecules and/or by genetic engineering is a viable approach to discover novel therapeutics for the prevention and treatment of metastasis of cancers with elevated NaV1.7 expression.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
36
|
Tauber M, Ben-Chaim Y. Voltage Sensors Embedded in G Protein-Coupled Receptors. Int J Mol Sci 2024; 25:5295. [PMID: 38791333 PMCID: PMC11120775 DOI: 10.3390/ijms25105295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Some signaling processes mediated by G protein-coupled receptors (GPCRs) are modulated by membrane potential. In recent years, increasing evidence that GPCRs are intrinsically voltage-dependent has accumulated. A recent publication challenged the view that voltage sensors are embedded in muscarinic receptors. Herein, we briefly discuss the evidence that supports the notion that GPCRs themselves are voltage-sensitive proteins and an alternative mechanism that suggests that voltage-gated sodium channels are the voltage-sensing molecules involved in such processes.
Collapse
Affiliation(s)
| | - Yair Ben-Chaim
- Department of Natural Sciences, The Open University of Israel, Ra’anana 4353701, Israel
| |
Collapse
|
37
|
Rühl P, Nair AG, Gawande N, Dehiwalage SNCW, Münster L, Schönherr R, Heinemann SH. An Ultrasensitive Genetically Encoded Voltage Indicator Uncovers the Electrical Activity of Non-Excitable Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307938. [PMID: 38526185 PMCID: PMC11132041 DOI: 10.1002/advs.202307938] [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: 10/20/2023] [Revised: 02/10/2024] [Indexed: 03/26/2024]
Abstract
Most animal cell types are classified as non-excitable because they do not generate action potentials observed in excitable cells, such as neurons and muscle cells. Thus, resolving voltage signals in non-excitable cells demands sensors with exceptionally high voltage sensitivity. In this study, the ultrabright, ultrasensitive, and calibratable genetically encoded voltage sensor rEstus is developed using structure-guided engineering. rEstus is most sensitive in the resting voltage range of non-excitable cells and offers a 3.6-fold improvement in brightness change for fast voltage spikes over its precursor ASAP3. Using rEstus, it is uncovered that the membrane voltage in several non-excitable cell lines (A375, HEK293T, MCF7) undergoes spontaneous endogenous alterations on a second to millisecond timescale. Correlation analysis of these optically recorded voltage alterations provides a direct, real-time readout of electrical cell-cell coupling, showing that visually connected A375 and HEK293T cells are also largely electrically connected, while MCF7 cells are only weakly coupled. The presented work provides enhanced tools and methods for non-invasive voltage imaging in living cells and demonstrates that spontaneous endogenous membrane voltage alterations are not limited to excitable cells but also occur in a variety of non-excitable cell types.
Collapse
Affiliation(s)
- Philipp Rühl
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Anagha G. Nair
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Namrata Gawande
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Sassrika N. C. W. Dehiwalage
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Lukas Münster
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Roland Schönherr
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| | - Stefan H. Heinemann
- Center for Molecular Biomedicine, Department of BiophysicsFriedrich Schiller University Jena and Jena University HospitalD‐07745JenaGermany
| |
Collapse
|
38
|
Bisht K, Lomholt MA, Khandelia H. Sensing membrane voltage by reorientation of dipolar transmembrane peptides. Biophys J 2024; 123:584-597. [PMID: 38308436 PMCID: PMC10938080 DOI: 10.1016/j.bpj.2024.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/23/2023] [Accepted: 01/29/2024] [Indexed: 02/04/2024] Open
Abstract
Membrane voltage plays a vital role in the behavior and functions of the lipid bilayer membrane. For instance, it regulates the exchange of molecules across the membrane through transmembrane proteins such as ion channels. In this paper, we study the membrane voltage-sensing mechanism, which entails the reorientation of α-helices with a change in the membrane voltage. We consider a helix having a large electrical macrodipole embedded in a lipid bilayer as a model system. We performed extensive molecular dynamics simulations to study the effect of variation of membrane voltage on the tilt angle of peptides and ascertain the optimal parameters for designing such a voltage-sensing peptide. A theoretical model for the system is also developed to investigate the interplay of competing effects of hydrophobic mismatch and dipole-electric field coupling on the tilt of the peptide and further explore the parameter space. This work opens the possibility for the design and fabrication of artificial dipolar membrane voltage-sensing elements for biomedical applications.
Collapse
Affiliation(s)
- Konark Bisht
- PHYLIFE: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark.
| | - Michael A Lomholt
- PHYLIFE: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Himanshu Khandelia
- PHYLIFE: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
39
|
Gest AM, Grenier V, Miller EW. Optical Estimation of Membrane Potential Values Using Fluorescence Lifetime Imaging Microscopy and Hybrid Chemical-Genetic Voltage Indicators. Bioelectricity 2024; 6:34-41. [PMID: 38516638 PMCID: PMC10951690 DOI: 10.1089/bioe.2023.0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
Introduction Membrane potential (Vm), the voltage across a cell membrane, is an important biophysical phenomenon, central to the physiology of cells, tissues, and organisms. Voltage-sensitive fluorescent indicators are a powerful method for interrogating membrane potential in living systems, but most indicators are best suited for detecting changes in membrane potential rather than measuring values of the membrane potential. One promising approach is to use fluorescence lifetime imaging microscopy (FLIM) in combination of chemically synthesized dyes to estimate a value of membrane potential. However, a drawback is that chemically synthesized dyes show poor specificity of staining. Objectives To address this problem, we applied a chemical-genetic voltage imaging approach to FLIM to enable optical estimation of membrane potential values from genetically defined cells. Results In this report, we detail the characterization and evaluation of two of these systems in mammalian cells. We further validate the use of a FLIM-based chemical genetic voltage indicator in mammalian neurons. Conclusions Finally, we discuss opportunities for future improvements to chemical-genetic FLIM-based voltage indicators.
Collapse
Affiliation(s)
- Anneliese M.M. Gest
- Department of Chemistry, University of California, Berkeley, California, USA
| | - Vincent Grenier
- Department of Chemistry, University of California, Berkeley, California, USA
| | - Evan W. Miller
- Department of Chemistry, University of California, Berkeley, California, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| |
Collapse
|
40
|
Rifat A, Ossola B, Bürli RW, Dawson LA, Brice NL, Rowland A, Lizio M, Xu X, Page K, Fidzinski P, Onken J, Holtkamp M, Heppner FL, Geiger JRP, Madry C. Differential contribution of THIK-1 K + channels and P2X7 receptors to ATP-mediated neuroinflammation by human microglia. J Neuroinflammation 2024; 21:58. [PMID: 38409076 PMCID: PMC10895799 DOI: 10.1186/s12974-024-03042-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/12/2024] [Indexed: 02/28/2024] Open
Abstract
Neuroinflammation is highly influenced by microglia, particularly through activation of the NLRP3 inflammasome and subsequent release of IL-1β. Extracellular ATP is a strong activator of NLRP3 by inducing K+ efflux as a key signaling event, suggesting that K+-permeable ion channels could have high therapeutic potential. In microglia, these include ATP-gated THIK-1 K+ channels and P2X7 receptors, but their interactions and potential therapeutic role in the human brain are unknown. Using a novel specific inhibitor of THIK-1 in combination with patch-clamp electrophysiology in slices of human neocortex, we found that THIK-1 generated the main tonic K+ conductance in microglia that sets the resting membrane potential. Extracellular ATP stimulated K+ efflux in a concentration-dependent manner only via P2X7 and metabotropic potentiation of THIK-1. We further demonstrated that activation of P2X7 was mandatory for ATP-evoked IL-1β release, which was strongly suppressed by blocking THIK-1. Surprisingly, THIK-1 contributed only marginally to the total K+ conductance in the presence of ATP, which was dominated by P2X7. This suggests a previously unknown, K+-independent mechanism of THIK-1 for NLRP3 activation. Nuclear sequencing revealed almost selective expression of THIK-1 in human brain microglia, while P2X7 had a much broader expression. Thus, inhibition of THIK-1 could be an effective and, in contrast to P2X7, microglia-specific therapeutic strategy to contain neuroinflammation.
Collapse
Affiliation(s)
- Ali Rifat
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Bernardino Ossola
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Roland W Bürli
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Lee A Dawson
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Nicola L Brice
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Anna Rowland
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Marina Lizio
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Xiao Xu
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Keith Page
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Pawel Fidzinski
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurology, Epilepsy-Center Berlin-Brandenburg, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Neurocure Cluster of Excellence, Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Julia Onken
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Martin Holtkamp
- Department of Neurology, Epilepsy-Center Berlin-Brandenburg, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Frank L Heppner
- Neurocure Cluster of Excellence, Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
| | - Jörg R P Geiger
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christian Madry
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
41
|
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
|
42
|
Monteil A, Guérineau NC, Gil-Nagel A, Parra-Diaz P, Lory P, Senatore A. New insights into the physiology and pathophysiology of the atypical sodium leak channel NALCN. Physiol Rev 2024; 104:399-472. [PMID: 37615954 DOI: 10.1152/physrev.00014.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cell excitability and its modulation by hormones and neurotransmitters involve the concerted action of a large repertoire of membrane proteins, especially ion channels. Unique complements of coexpressed ion channels are exquisitely balanced against each other in different excitable cell types, establishing distinct electrical properties that are tailored for diverse physiological contributions, and dysfunction of any component may induce a disease state. A crucial parameter controlling cell excitability is the resting membrane potential (RMP) set by extra- and intracellular concentrations of ions, mainly Na+, K+, and Cl-, and their passive permeation across the cell membrane through leak ion channels. Indeed, dysregulation of RMP causes significant effects on cellular excitability. This review describes the molecular and physiological properties of the Na+ leak channel NALCN, which associates with its accessory subunits UNC-79, UNC-80, and NLF-1/FAM155 to conduct depolarizing background Na+ currents in various excitable cell types, especially neurons. Studies of animal models clearly demonstrate that NALCN contributes to fundamental physiological processes in the nervous system including the control of respiratory rhythm, circadian rhythm, sleep, and locomotor behavior. Furthermore, dysfunction of NALCN and its subunits is associated with severe pathological states in humans. The critical involvement of NALCN in physiology is now well established, but its study has been hampered by the lack of specific drugs that can block or agonize NALCN currents in vitro and in vivo. Molecular tools and animal models are now available to accelerate our understanding of how NALCN contributes to key physiological functions and the development of novel therapies for NALCN channelopathies.
Collapse
Affiliation(s)
- Arnaud Monteil
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Antonio Gil-Nagel
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Paloma Parra-Diaz
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Adriano Senatore
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
43
|
Liu C, Xue L, Song C. Calcium binding and permeation in TRPV channels: Insights from molecular dynamics simulations. J Gen Physiol 2023; 155:e202213261. [PMID: 37728593 PMCID: PMC10510737 DOI: 10.1085/jgp.202213261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/21/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023] Open
Abstract
Some calcium channels selectively permeate Ca2+, despite the high concentration of monovalent ions in the surrounding environment, which is essential for many physiological processes. Without atomistic and dynamical ion permeation details, the underlying mechanism of Ca2+ selectivity has long been an intensively studied, yet controversial, topic. This study takes advantage of the homologous Ca2+-selective TRPV6 and non-selective TRPV1 and utilizes the recently solved open-state structures and a newly developed multisite calcium model to investigate the ion binding and permeation features in TRPV channels by molecular dynamics simulations. Our results revealed that the open-state TRPV6 and TRPV1 show distinct ion binding patterns in the selectivity filter, which lead to different ion permeation features. Two Ca2+ ions simultaneously bind to the selectivity filter of TRPV6 compared with only one Ca2+ in the case of TRPV1. Multiple Ca2+ binding at the selectivity filter of TRPV6 permeated in a concerted manner, which could efficiently block the permeation of Na+. Cations of various valences differentiate between the binding sites at the entrance of the selectivity filter in TRPV6. Ca2+ preferentially binds to the central site with a higher probability of permeation, repelling Na+ to a peripheral site. Therefore, we believe that ion binding competition at the selectivity filter of calcium channels, including the binding strength and number of binding sites, determines Ca2+ selectivity under physiological conditions.
Collapse
Affiliation(s)
- Chunhong Liu
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Lingfeng Xue
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Chen Song
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
44
|
Taylor KR, Monje M. Neuron-oligodendroglial interactions in health and malignant disease. Nat Rev Neurosci 2023; 24:733-746. [PMID: 37857838 PMCID: PMC10859969 DOI: 10.1038/s41583-023-00744-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2023] [Indexed: 10/21/2023]
Abstract
Experience sculpts brain structure and function. Activity-dependent modulation of the myelinated infrastructure of the nervous system has emerged as a dimension of adaptive change during childhood development and in adulthood. Myelination is a richly dynamic process, with neuronal activity regulating oligodendrocyte precursor cell proliferation, oligodendrogenesis and myelin structural changes in some axonal subtypes and in some regions of the nervous system. This myelin plasticity and consequent changes to conduction velocity and circuit dynamics can powerfully influence neurological functions, including learning and memory. Conversely, disruption of the mechanisms mediating adaptive myelination can contribute to cognitive impairment. The robust effects of neuronal activity on normal oligodendroglial precursor cells, a putative cellular origin for many forms of glioma, indicates that dysregulated or 'hijacked' mechanisms of myelin plasticity could similarly promote growth in this devastating group of brain cancers. Indeed, neuronal activity promotes the pathogenesis of many forms of glioma in preclinical models through activity-regulated paracrine factors and direct neuron-to-glioma synapses. This synaptic integration of glioma into neural circuits is central to tumour growth and invasion. Thus, not only do neuron-oligodendroglial interactions modulate neural circuit structure and function in the healthy brain, but neuron-glioma interactions also have important roles in the pathogenesis of glial malignancies.
Collapse
Affiliation(s)
- Kathryn R Taylor
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
45
|
Kirchhofer SB, Lim VJY, Ernst S, Karsai N, Ruland JG, Canals M, Kolb P, Bünemann M. Differential interaction patterns of opioid analgesics with µ opioid receptors correlate with ligand-specific voltage sensitivity. eLife 2023; 12:e91291. [PMID: 37983079 PMCID: PMC10849675 DOI: 10.7554/elife.91291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/19/2023] [Indexed: 11/21/2023] Open
Abstract
The µ opioid receptor (MOR) is the key target for analgesia, but the application of opioids is accompanied by several issues. There is a wide range of opioid analgesics, differing in their chemical structure and their properties of receptor activation and subsequent effects. A better understanding of ligand-receptor interactions and the resulting effects is important. Here, we calculated the respective binding poses for several opioids and analyzed interaction fingerprints between ligand and receptor. We further corroborated the interactions experimentally by cellular assays. As MOR was observed to display ligand-induced modulation of activity due to changes in membrane potential, we further analyzed the effects of voltage sensitivity on this receptor. Combining in silico and in vitro approaches, we defined discriminating interaction patterns responsible for ligand-specific voltage sensitivity and present new insights into their specific effects on activation of the MOR.
Collapse
Affiliation(s)
- Sina B Kirchhofer
- Department of Pharmacology and Clinical Pharmacy, University of MarburgMarburgGermany
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of NottinghamNottinghamUnited Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and NottinghamMidlandsUnited Kingdom
| | - Victor Jun Yu Lim
- Department of Pharmaceutical Chemistry, University of MarburgMarburgGermany
| | - Sebastian Ernst
- Department of Pharmacology and Clinical Pharmacy, University of MarburgMarburgGermany
| | - Noemi Karsai
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of NottinghamNottinghamUnited Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and NottinghamMidlandsUnited Kingdom
| | - Julia G Ruland
- Department of Pharmacology and Clinical Pharmacy, University of MarburgMarburgGermany
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of NottinghamNottinghamUnited Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and NottinghamMidlandsUnited Kingdom
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of MarburgMarburgGermany
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, University of MarburgMarburgGermany
| |
Collapse
|
46
|
Zuccolini P, Barbieri R, Sbrana F, Picco C, Gavazzo P, Pusch M. IK Channel-Independent Effects of Clotrimazole and Senicapoc on Cancer Cells Viability and Migration. Int J Mol Sci 2023; 24:16285. [PMID: 38003471 PMCID: PMC10671816 DOI: 10.3390/ijms242216285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Many studies highlighted the importance of the IK channel for the proliferation and the migration of different types of cancer cells, showing how IK blockers could slow down cancer growth. Based on these data, we wanted to characterize the effects of IK blockers on melanoma metastatic cells and to understand if such effects were exclusively IK-dependent. For this purpose, we employed two different blockers, namely clotrimazole and senicapoc, and two cell lines: metastatic melanoma WM266-4 and pancreatic cancer Panc-1, which is reported to have little or no IK expression. Clotrimazole and senicapoc induced a decrease in viability and the migration of both WM266-4 and Panc-1 cells irrespective of IK expression levels. Patch-clamp experiments on WM266-4 cells revealed Ca2+-dependent, IK-like, clotrimazole- and senicapoc-sensitive currents, which could not be detected in Panc-1 cells. Neither clotrimazole nor senicapoc altered the intracellular Ca2+ concentration. These results suggest that the effects of IK blockers on cancer cells are not strictly dependent on a robust presence of the channel in the plasma membrane, but they might be due to off-target effects on other cellular targets or to the blockade of IK channels localized in intracellular organelles.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael Pusch
- Biophysics Institute, National Research Council, 16149 Genova, Italy; (P.Z.); (R.B.); (F.S.); (C.P.); (P.G.)
| |
Collapse
|
47
|
Sheroziya M, Khazipov R. Synaptic Origin of Early Sensory-evoked Oscillations in the Immature Thalamus. Neuroscience 2023; 532:50-64. [PMID: 37769898 DOI: 10.1016/j.neuroscience.2023.09.005] [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: 06/10/2023] [Revised: 08/22/2023] [Accepted: 09/08/2023] [Indexed: 10/03/2023]
Abstract
During the critical period of postnatal development, brain maturation is extremely sensitive to external stimuli. Newborn rodents already have functional somatosensory pathways and the thalamus, but the cortex is still forming. Immature thalamic synapses may produce large postsynaptic potentials in immature neurons, while non-synaptic membrane currents remain relatively weak and slow. The thalamocortical system generates spontaneous and evoked early gamma and spindle-burst oscillations in newborn rodents. How relatively strong synapses and weak intrinsic currents interact with each other and how they contribute to early thalamic activities remains largely unknown. Here, we performed local field potential (LFP), juxtacellular, and patch-clamp recordings in the somatosensory thalamus of urethane-anesthetized rat pups at postnatal days 6-7 with one whisker stimulation. We removed the overlying cortex and hippocampus to reach the thalamus with electrodes. Deflection of only one (the principal) whisker induced spikes in a particular thalamic cell. Whisker deflection evoked a group of large-amplitude excitatory events, likely originating from lemniscal synapses and multiple inhibitory postsynaptic events in thalamocortical cells. Large-amplitude excitatory events produced a group of spike bursts and could evoke a depolarization block. Juxtacellular recordings confirmed the partial inactivation of spikes. Inhibitory events prevented inactivation of action potentials and gamma-modulated neuronal firing. We conclude that the interplay of strong excitatory and inhibitory synapses and relatively weak intrinsic currents produces sensory-evoked early gamma oscillations in thalamocortical cells. We also propose that sensory-evoked large-amplitude excitatory events contribute to evoked spindle-bursts.
Collapse
Affiliation(s)
- Maxim Sheroziya
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia.
| | - Roustem Khazipov
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia; Aix Marseille University, INSERM, INMED, Marseille, France
| |
Collapse
|
48
|
Alvarez-Lorenzo C, Zarur M, Seijo-Rabina A, Blanco-Fernandez B, Rodríguez-Moldes I, Concheiro A. Physical stimuli-emitting scaffolds: The role of piezoelectricity in tissue regeneration. Mater Today Bio 2023; 22:100740. [PMID: 37521523 PMCID: PMC10374602 DOI: 10.1016/j.mtbio.2023.100740] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/01/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023] Open
Abstract
The imbalance between life expectancy and quality of life is increasing due to the raising prevalence of chronic diseases. Musculoskeletal disorders and chronic wounds affect a growing percentage of people and demand more efficient tools for regenerative medicine. Scaffolds that can better mimic the natural physical stimuli that tissues receive under healthy conditions and during healing may significantly aid the regeneration process. Shape, mechanical properties, pore size and interconnectivity have already been demonstrated to be relevant scaffold features that can determine cell adhesion and differentiation. Much less attention has been paid to scaffolds that can deliver more dynamic physical stimuli, such as electrical signals. Recent developments in the precise measurement of electrical fields in vivo have revealed their key role in cell movement (galvanotaxis), growth, activation of secondary cascades, and differentiation to different lineages in a variety of tissues, not just neural. Piezoelectric scaffolds can mimic the natural bioelectric potentials and gradients in an autonomous way by generating the electric stimuli themselves when subjected to mechanical loads or, if the patient or the tissue lacks mobility, ultrasound irradiation. This review provides an analysis on endogenous bioelectrical signals, recent developments on piezoelectric scaffolds for bone, cartilage, tendon and nerve regeneration, and their main outcomes in vivo. Wound healing with piezoelectric dressings is addressed in the last section with relevant examples of performance in animal models. Results evidence that a fine adjustment of material composition and processing (electrospinning, corona poling, 3D printing, annealing) provides scaffolds that act as true emitters of electrical stimuli that activate endogenous signaling pathways for more efficient and long-term tissue repair.
Collapse
Affiliation(s)
- Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Mariana Zarur
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Alejandro Seijo-Rabina
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Barbara Blanco-Fernandez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Isabel Rodríguez-Moldes
- Grupo NEURODEVO, Departamento de Bioloxía Funcional, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| |
Collapse
|
49
|
Eskandari N, Gentile S. Potassium channels activity unveils cancer vulnerability. CURRENT TOPICS IN MEMBRANES 2023; 92:1-14. [PMID: 38007264 DOI: 10.1016/bs.ctm.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
"No cell could exist without ion channels" (Clay Armstrong; 1999). Since the discovery in the early 1950s, that ions move across biological membranes, the idea that changes of ionic gradients can generate biological signals has fascinated scientists in any fields. Soon later (1960s) it was found that ionic flows were controlled by a class of specific and selective proteins called ion channels. Thus, it became clear that the concerted activities of these proteins can initiate, arrest, and finely tune a variety of biochemical cascades which offered the opportunity to better understand both biology and pathology. Cancer is a disease that is notoriously difficult to treat due its heterogeneous nature which makes it the deadliest disease in the developed world. Recently, emerging evidence has established that potassium channels are critical modulators of several hallmarks of cancer including tumor growth, metastasis, and metabolism. Nevertheless, the role of potassium ion channels in cancer biology and the therapeutic potential offered by targeting these proteins has not been explored thoroughly. This chapter is addressed to both cancer biologists and ion channels scientists and it aims to shine a light on the established and potential roles of potassium ion channels in cancer biology and on the therapeutic benefit of targeting potassium channels with activator molecules.
Collapse
Affiliation(s)
- Najmeh Eskandari
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Saverio Gentile
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
50
|
Moniruzzaman M, Karal MAS, Wadud MA, Rashid MMO. Increase in anionic Fe 3O 4 nanoparticle-induced membrane poration and vesicle deformation due to membrane potential - an experimental study. Phys Chem Chem Phys 2023; 25:23111-23124. [PMID: 37602684 DOI: 10.1039/d3cp02702c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
The membrane potential plays a significant role in various cellular processes while interacting with membrane active agents. So far, all the investigations of the interaction of nanoparticles (NPs) with lipid vesicles have been performed in the absence of membrane potential. In this study, the anionic magnetite NP-induced poration along with deformation of cell-mimetic giant unilamellar vesicles (GUVs) has been studied in the presence of various membrane potentials. Lipids 1,2-dioleoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DOPG), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), and channel forming protein gramicidin A (GrA) are used to synthesize the DOPG/DOPC/GrA-GUVs. The static and dynamic nature of GUVs is investigated using phase contrast fluorescent microscopy. The presence of GrA in the membrane decreases the leakage constant of the encapsulating fluorescent probe (calcein) in the absence of membrane potential. With the increase of negative membrane potential, the leakage shifts from a single exponential to two exponential functions, obtaining two leakage constants. The leakage became faster at the initial stage, and at the final stage, it became slower with the increase in negative membrane potential. Both the fraction of poration and deformation increase with the increase of negative membrane potential. These results suggested that the membrane potential enhances the NP-induced poration along with the deformation of DOPG/DOPC/GrA-GUVs. The increase of the binding constant in the NPs with membrane potential is one of the important factors for increasing membrane permeation and vesicle deformation.
Collapse
Affiliation(s)
- Md Moniruzzaman
- Department of Physics, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh.
| | - Mohammad Abu Sayem Karal
- Department of Physics, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh.
| | - Md Abdul Wadud
- Department of Physics, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh.
| | - Md Mamun Or Rashid
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| |
Collapse
|