1
|
Sözer EB, Semenov I, Vernier PT. Dihydroethidium-derived fluorescence in electrically stressed cells indicates intracellular microenvironment modifications independent of ROS. Bioelectrochemistry 2024; 160:108751. [PMID: 38851174 DOI: 10.1016/j.bioelechem.2024.108751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Intracellular reactive oxygen species (ROS) generation is widely suggested as a trigger for biological consequences of electric field exposures, such as those in electroporation applications. ROS are linked with membrane barrier function degradation, genetic damage, and complex events like immunological cell death. Dihydroethidium (DHE) is commonly used to monitor ROS in cells. DHE is linked to intracellular ROS by a primary oxidation product, Ethidium (Eth+), that shows increased fluorescence upon binding to polynucleotides. We observed changes in DHE-derived fluorescence in Chinese hamster ovary (CHO) cells post 300-ns electric pulse exposures, comparing them to tert-butyl-hydroperoxide (t-BHP) induced oxidative stress. Immediate intracellular fluorescence changes were noted in both cases, but with distinct localization patterns. After electrical stress, cytosolic DHE-derived fluorescence intensity decreases, and nucleolar intensity increases. Conversely, t-BHP exposure increases DHE-derived fluorescence uniformly across the cell. Surprisingly, fluorescence patterns after electrical stress in Eth+-loaded cells is identical to those in DHE-loaded cells, in kinetics and localization patterns. These findings indicate that DHE-derived fluorescence changes after pulsed electric field stress are not due to intracellular ROS generation leading to DHE oxidation, but rather indicate stress-induced intracellular microenvironment alterations affecting Eth+ fluorescence.
Collapse
Affiliation(s)
- Esin B Sözer
- Old Dominion University, Frank Reidy Research Center for Bioelectrics, Norfolk, VA, USA.
| | - Iurii Semenov
- Old Dominion University, Frank Reidy Research Center for Bioelectrics, Norfolk, VA, USA
| | - P Thomas Vernier
- Old Dominion University, Frank Reidy Research Center for Bioelectrics, Norfolk, VA, USA.
| |
Collapse
|
2
|
Baker C, Willis A, Milestone W, Baker M, Garner AL, Joshi RP. Numerical assessments of geometry, proximity and multi-electrode effects on electroporation in mitochondria and the endoplasmic reticulum to nanosecond electric pulses. Sci Rep 2024; 14:23854. [PMID: 39394381 PMCID: PMC11470013 DOI: 10.1038/s41598-024-74659-z] [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: 10/28/2023] [Accepted: 09/27/2024] [Indexed: 10/13/2024] Open
Abstract
Most simulations of electric field driven bioeffects have considered spherical cellular geometries or probed symmetrical structures for simplicity. This work assesses cellular transmembrane potential build-up and electroporation in a Jurkat cell that includes the endoplasmic reticulum (ER) and mitochondria, both of which have complex shapes, in response to external nanosecond electric pulses. The simulations are based on a time-domain nodal analysis that incorporates membrane poration utilizing the Smoluchowski model with angular-dependent changes in membrane conductivity. Consistent with prior experimental reports, the simulations show that the ER requires the largest electric field for electroporation, while the inner mitochondrial membrane (IMM) is the easiest membrane to porate. Our results suggest that the experimentally observed increase in intracellular calcium could be due to a calcium induced calcium release (CICR) process that is initiated by outer cell membrane breakdown. Repeated pulsing and/or using multiple electrodes are shown to create a stronger poration. The role of mutual coupling, screening, and proximity effects in bringing about electric field modifications is also probed. Finally, while including greater geometric details might refine predictions, the qualitative trends are expected to remain.
Collapse
Affiliation(s)
- C Baker
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, 79409, USA
| | - A Willis
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, 79409, USA
- Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - W Milestone
- Nanohmics, Inc, 6201 E Oltorf St, Austin, TX, 78717, USA
| | - M Baker
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, 79409, USA
| | - A L Garner
- School of Nuclear Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Elmore Family School of Electrical and Computer Engineering, West Lafayette, IN, 47907, USA
| | - R P Joshi
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, 79409, USA.
| |
Collapse
|
3
|
Yun SH, Mansurov V, Yang L, Yoon J, Leblanc N, Craviso GL, Zaklit J. Modulating Ca 2+ influx into adrenal chromaffin cells with short-duration nanosecond electric pulses. Biophys J 2024; 123:2537-2556. [PMID: 38909279 PMCID: PMC11365113 DOI: 10.1016/j.bpj.2024.06.021] [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: 11/14/2023] [Revised: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 06/24/2024] Open
Abstract
Isolated bovine adrenal chromaffin cells exposed to single 2-, 4-, or 5-ns pulses undergo a rapid, transient rise in intracellular Ca2+ mediated by Ca2+ entry via voltage-gated Ca2+ channels (VGCCs), mimicking the activation of these cells in vivo by acetylcholine. However, pulse durations 150 ns or longer elicit larger amplitude and longer-lived Ca2+ responses due to Ca2+ influx via both VGCCs and a yet to be identified plasma membrane pathway(s). To further our understanding of the differential effects of ultrashort versus longer pulse durations on Ca2+ influx, chromaffin cells were loaded with calcium green-1 and exposed to single 3-, 5-, 11-, 25-, or 50-ns pulses applied at their respective Ca2+ activation threshold electric fields. Increasing pulse duration from 3 or 5 ns to only 11 ns was sufficient to elicit increased amplitude and longer-lived Ca2+ responses in the majority of cells, a trend that continued as pulse duration increased to 50 ns. The amplification of Ca2+ responses was not the result of Ca2+ release from intracellular stores and was accompanied by a decreased effectiveness of VGCC inhibitors to block the responses and a reduced reliance on extracellular Na+ and membrane depolarization to evoke the responses. Inhibitors of pannexin channels, P2X receptors, or non-selective cation channels failed to attenuate 50-ns-elicited Ca2+ responses, ruling out these Ca2+-permeable channels as secondary Ca2+ entry pathways. Analytical calculations and numerical modeling suggest that the parameter that best determines the response of chromaffin cells to increasing pulse durations is the time the membrane charges to its peak voltage. These results highlight the pronounced sensitivity of a neuroendocrine cell to pulse durations differing by only tens of nanoseconds, which has important implications for the future development of nanosecond pulse technologies enabling electrostimulation applications for spatially focused and graded in vivo neuromodulation.
Collapse
Affiliation(s)
- Sung Hae Yun
- Department of Electrical and Biomedical Engineering, College of Engineering, University of Nevada, Reno, Nevada
| | - Vasilii Mansurov
- Department of Electrical and Biomedical Engineering, College of Engineering, University of Nevada, Reno, Nevada
| | - Lisha Yang
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada
| | - Jihwan Yoon
- Department of Electrical and Biomedical Engineering, College of Engineering, University of Nevada, Reno, Nevada
| | - Normand Leblanc
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada
| | - Gale L Craviso
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada
| | - Josette Zaklit
- Department of Electrical and Biomedical Engineering, College of Engineering, University of Nevada, Reno, Nevada.
| |
Collapse
|
4
|
Rems L, Rainot A, Wiczew D, Szulc N, Tarek M. Cellular excitability and ns-pulsed electric fields: Potential involvement of lipid oxidation in the action potential activation. Bioelectrochemistry 2024; 155:108588. [PMID: 37879163 DOI: 10.1016/j.bioelechem.2023.108588] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
Recent studies showed that nanosecond pulsed electric fields (nsPEFs) can activate voltage-gated ion channels (VGICs) and trigger action potentials (APs) in excitable cells. Under physiological conditions, VGICs' activation takes place on time scales of the order 10-100 µs. These time scales are considerably longer than the applied pulse duration, thus activation of VGICs by nsPEFs remains puzzling and there is no clear consensus on the mechanisms involved. Here we propose that changes in local electrical properties of the cell membrane due to lipid oxidation might be implicated in AP activation. We first use MD simulations of model lipid bilayers with increasing concentration of primary and secondary lipid oxidation products and demonstrate that oxidation not only increases the bilayer conductance, but also the bilayer capacitance. Equipped with MD-based characterization of electrical properties of oxidized bilayers, we then resort to AP modelling at the cell level with Hodgkin-Huxley-type models. We confirm that a local change in membrane properties, particularly the increase in membrane conductance, due to formation of oxidized membrane lesions can be high enough to trigger an AP, even when no external stimulus is applied. However, excessive accumulation of oxidized lesions (or other conductive defects) can lead to altered cell excitability.
Collapse
Affiliation(s)
- Lea Rems
- University of Ljubljana, Faculty of Electrical Engineering, SI-1000 Ljubljana, Slovenia.
| | | | - Daniel Wiczew
- Université de Lorraine, CNRS, LPCT, F-54000 Nancy, France
| | - Natalia Szulc
- Université de Lorraine, CNRS, LPCT, F-54000 Nancy, France
| | - Mounir Tarek
- Université de Lorraine, CNRS, LPCT, F-54000 Nancy, France.
| |
Collapse
|
5
|
Ruiz-Fernández AR, Campos L, Villanelo F, Garate JA, Perez-Acle T. Protein-Mediated Electroporation in a Cardiac Voltage-Sensing Domain Due to an nsPEF Stimulus. Int J Mol Sci 2023; 24:11397. [PMID: 37511161 PMCID: PMC10379607 DOI: 10.3390/ijms241411397] [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: 04/29/2023] [Revised: 06/15/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
This study takes a step in understanding the physiological implications of the nanosecond pulsed electric field (nsPEF) by integrating molecular dynamics simulations and machine learning techniques. nsPEF, a state-of-the-art technology, uses high-voltage electric field pulses with a nanosecond duration to modulate cellular activity. This investigation reveals a relatively new and underexplored phenomenon: protein-mediated electroporation. Our research focused on the voltage-sensing domain (VSD) of the NaV1.5 sodium cardiac channel in response to nsPEF stimulation. We scrutinized the VSD structures that form pores and thereby contribute to the physical chemistry that governs the defibrillation effect of nsPEF. To do so, we conducted a comprehensive analysis involving the clustering of 142 replicas simulated for 50 ns under nsPEF stimuli. We subsequently pinpointed the representative structures of each cluster and computed the free energy between them. We find that the selected VSD of NaV1.5 forms pores under nsPEF stimulation, but in a way that significant differs from the traditional VSD opening. This study not only extends our understanding of nsPEF and its interaction with protein channels but also adds a new effect to further study.
Collapse
Affiliation(s)
| | - Leonardo Campos
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
| | - Felipe Villanelo
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
| | - Jose Antonio Garate
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
- Millennium Nucleus im NanoBioPhysics, Universidad de Valparaiso, Valparaiso 2351319, Chile
| | - Tomas Perez-Acle
- Computational Biology Lab, Fundación Ciencia & Vida, Santiago 7780272, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago 8420524, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaiso 2360102, Chile
| |
Collapse
|
6
|
Łapińska Z, Novickij V, Rembiałkowska N, Szewczyk A, Dubińska-Magiera M, Kulbacka J, Saczko J. The influence of asymmetrical bipolar pulses and interphase intervals on the bipolar cancellation phenomenon in the ovarian cancer cell line. Bioelectrochemistry 2023; 153:108483. [PMID: 37301162 DOI: 10.1016/j.bioelechem.2023.108483] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023]
Abstract
The application of negative polarity electrical pulse (↓) following positive polarity pulses (↑) may induce bipolar cancellation (BPC), a unique physiological response believed to be specific to nanosecond electroporation (nsEP). The literature lacks analysis of bipolar electroporation (BP EP) involving asymmetrical sequences composed of nanosecond and microsecond pulses. Moreover, the impact of interphase interval on BPC caused by such asymmetrical pulse needs consideration. In this study, the authors utilized the ovarian clear carcinoma cell line (OvBH-1) model to investigate the BPC with asymmetrical sequences. Cells were exposed to pulses delivered in 10-pulse bursts but as uni- or bipolar, symmetrical, or asymmetrical sequences with a duration of 600 ns or 10 µs and electric field strength equal to 7.0 or 1.8 kV/cm, respectively. It was shown that the asymmetry of pulses influences BPC. The obtained results have also been investigated in the context of calcium electrochemotherapy. The reduction of cell membrane poration, and cell survival have been observed following Ca2+ electrochemotherapy. The effects of interphase delays (1 and 10 µs) on the BPC phenomenon were reported. Our findings show that the BPC phenomenon can be controlled using pulse asymmetry or delay between the positive and negative polarity of the pulse.
Collapse
Affiliation(s)
- Zofia Łapińska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland.
| | - 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, 08410 Vilnius, Lithuania
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Magdalena Dubińska-Magiera
- Department of Animal Developmental Biology, Faculty of Biological Science, University of Wroclaw, Sienkiewicza 21, 50-335 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410 Vilnius, Lithuania.
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
7
|
Ibrahimi N, Vallet L, Andre FM, Rivaletto M, Novac BM, Mir LM, Pécastaing L. An Overview of Subnanosecond Pulsed Electric Field Biological Effects: Toward Contactless Technologies for Cancer Treatment. Bioelectricity 2023. [DOI: 10.1089/bioe.2022.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Affiliation(s)
- Njomza Ibrahimi
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
| | - Leslie Vallet
- Université Paris-Saclay, CNRS, Gustave Roussy, UMR 9018, Metabolic and Systemic Aspects of Oncogenesis (METSY), Villejuif, France
| | - Franck M. Andre
- Université Paris-Saclay, CNRS, Gustave Roussy, UMR 9018, Metabolic and Systemic Aspects of Oncogenesis (METSY), Villejuif, France
| | - Marc Rivaletto
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
| | - Bucur M. Novac
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, United Kingdom
| | - Lluis M. Mir
- Université Paris-Saclay, CNRS, Gustave Roussy, UMR 9018, Metabolic and Systemic Aspects of Oncogenesis (METSY), Villejuif, France
| | - Laurent Pécastaing
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
| |
Collapse
|
8
|
Abed T, Ganser K, Eckert F, Stransky N, Huber SM. Ion channels as molecular targets of glioblastoma electrotherapy. Front Cell Neurosci 2023; 17:1133984. [PMID: 37006466 PMCID: PMC10064067 DOI: 10.3389/fncel.2023.1133984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/10/2023] [Indexed: 03/19/2023] Open
Abstract
Therapies with weak, non-ionizing electromagnetic fields comprise FDA-approved treatments such as Tumor Treating Fields (TTFields) that are used for adjuvant therapy of glioblastoma. In vitro data and animal models suggest a variety of biological TTFields effects. In particular, effects ranging from direct tumoricidal, radio- or chemotherapy-sensitizing, metastatic spread-inhibiting, up to immunostimulation have been described. Diverse underlying molecular mechanisms, such as dielectrophoresis of cellular compounds during cytokinesis, disturbing the formation of the spindle apparatus during mitosis, and perforating the plasma membrane have been proposed. Little attention, however, has been paid to molecular structures that are predestinated to percept electromagnetic fields-the voltage sensors of voltage-gated ion channels. The present review article briefly summarizes the mode of action of voltage sensing by ion channels. Moreover, it introduces into the perception of ultra-weak electric fields by specific organs of fishes with voltage-gated ion channels as key functional units therein. Finally, this article provides an overview of the published data on modulation of ion channel function by diverse external electromagnetic field protocols. Combined, these data strongly point to a function of voltage-gated ion channels as transducers between electricity and biology and, hence, to voltage-gated ion channels as primary targets of electrotherapy.
Collapse
Affiliation(s)
- Tayeb Abed
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Radiation Oncology, Medical University Vienna, Vienna, Austria
| | - Nicolai Stransky
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Kaynak A, N’Guessan KF, Patel PH, Lee JH, Kogan AB, Narmoneva DA, Qi X. Electric Fields Regulate In Vitro Surface Phosphatidylserine Exposure of Cancer Cells via a Calcium-Dependent Pathway. Biomedicines 2023; 11:biomedicines11020466. [PMID: 36831002 PMCID: PMC9953458 DOI: 10.3390/biomedicines11020466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Cancer is the second leading cause of death worldwide after heart disease. The current treatment options to fight cancer are limited, and there is a critical need for better treatment strategies. During the last several decades, several electric field (EF)-based approaches for anti-cancer therapies have been introduced, such as electroporation and tumor-treating fields; still, they are far from optimal due to their invasive nature, limited efficacy and significant side effects. In this study, we developed a non-contact EF stimulation system to investigate the in vitro effects of a novel EF modality on cancer biomarkers in normal (human astrocytes, human pancreatic ductal epithelial -HDPE-cells) and cancer cell lines (glioblastoma U87-GBM, human pancreatic cancer cfPac-1, and MiaPaCa-2). Our results demonstrate that this EF modality can successfully modulate an important cancer cell biomarker-cell surface phosphatidylserine (PS). Our results further suggest that moderate, but not low, amplitude EF induces p38 mitogen-activated protein kinase (MAPK), actin polymerization, and cell cycle arrest in cancer cell lines. Based on our results, we propose a mechanism for EF-mediated PS exposure in cancer cells, where the magnitude of induced EF on the cell surface can differentially regulate intracellular calcium (Ca2+) levels, thereby modulating surface PS exposure.
Collapse
Affiliation(s)
- Ahmet Kaynak
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kombo F. N’Guessan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Priyankaben H. Patel
- Department of Biomedical Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jing-Huei Lee
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Andrei B. Kogan
- Department of Physics, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Daria A. Narmoneva
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Xiaoyang Qi
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-513-558-4025
| |
Collapse
|
10
|
Ruiz-Fernández AR, Rosemblatt M, Perez-Acle T. Nanosecond pulsed electric field (nsPEF) and vaccines: a novel technique for the inactivation of SARS-CoV-2 and other viruses? Ann Med 2022; 54:1749-1756. [PMID: 35786157 PMCID: PMC9258060 DOI: 10.1080/07853890.2022.2087898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Since the beginning of 2020, worldwide attention has been being focussed on SARS-CoV-2, the second strain of the severe acute respiratory syndrome virus. Although advances in vaccine technology have been made, particularly considering the advent of mRNA vaccines, up to date, no single antigen design can ensure optimal immune response. Therefore, new technologies must be tested as to their ability to further improve vaccines. Nanosecond Pulsed Electric Field (nsPEF) is one such method showing great promise in different biomedical and industrial fields, including the fight against COVID-19. Of note, available research shows that nsPEF directly damages the cell's DNA, so it is critical to determine if this technology could be able to fragment either viral DNA or RNA so as to be used as a novel technology to produce inactivated pathogenic agents that may, in turn, be used for the production of vaccines. Considering the available evidence, we propose that nsPEF may be used to produce inactivated SARS-CoV-2 viruses that may in turn be used to produce novel vaccines, as another tool to address 20 the current COVID-19 pandemic.Key MessagesViral inactivation by using pulsed electric fields in the nanosecond frequency.DNA fragmentation by a Nanosecond Pulsed Electric Field (nsPEF).Opportunity to apply new technologies in vaccine development.
Collapse
Affiliation(s)
- A R Ruiz-Fernández
- Computational Biology Lab, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile.,Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago, Chile
| | - M Rosemblatt
- Computational Biology Lab, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - T Perez-Acle
- Computational Biology Lab, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile.,Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
11
|
Xu M, Xu D, Dong G, Ren Z, Zhang W, Aji T, Zhao Q, Chen X, Jiang T. The Safety and Efficacy of Nanosecond Pulsed Electric Field in Patients With Hepatocellular Carcinoma: A Prospective Phase 1 Clinical Study Protocol. Front Oncol 2022; 12:869316. [PMID: 35912221 PMCID: PMC9328750 DOI: 10.3389/fonc.2022.869316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly aggressive malignancy. Irreversible electroporation (IRE) is an ablative modality that uses high-voltage electrical pulses to permeabilize the cell membrane leading to cell necrosis. Unlike traditional thermal ablation, IRE is hardly affected by the "heat-sink" effect and can prevent damage of the adjacent vital structures. Nanosecond pulsed electric field (nsPEF) is a new IRE technique using ultra-short pulses (nanosecond duration), can not only penetrate the cell membranes, but also act on the organelles. Sufficient preclinical researches have shown that nsPEF can eliminate HCC without damaging vital organs, and elicit potent anti-tumor immune response. OBJECTIVE This is the first clinical study to evaluate feasibility, efficacy, and safety of nsPEF for the treatment of HCC, where thermal ablation is unsuitable due to proximity to critical structures. METHODS AND ANALYSIS We will conduct an open-labeled, single-arm, prospective, multicenter, and objective performance criteria trial. One hundred and ninety-two patients with HCC, in which the tumor is located immediately (<0.5 cm) adjacent to the portal vein, hepatic veins, bile duct, gastrointestinal tract, or diaphragm, will be enrolled among 4 academic medical centers. The primary outcomes are the rate of complete ablation at 1 month and adverse events. Secondary outcomes include technical success, technique efficacy, nsPEF procedural characteristics, local tumor progression, and local progression-free survival. ETHICS AND DISSEMINATION The trial will be conducted according to the ethical principles of the Declaration of Helsinki and has been approved by the ethics committee of all participating centers. The results of this study will be published in peer-reviewed scientific journals and presented at relevant academic conferences. CONCLUSIONS This study is the Phase 1 clinical trial to evaluate the efficacy and safety of nsPEF in patients with HCC at high-risk locations where thermal ablation is contra-indicated. The results may expand the options and offer an alternative therapy for HCC. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, identifier NCT04309747.
Collapse
Affiliation(s)
- Min Xu
- Department of Ultrasound Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| | - Danxia Xu
- Department of Ultrasound Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| | - Gang Dong
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Ren
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wu Zhang
- Shulan International Medical College, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Hangzhou, China
| | - Tuerganaili Aji
- Department of Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qiyu Zhao
- Department of Ultrasound Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| | - Xinhua Chen
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tian’an Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| |
Collapse
|
12
|
Ruiz-Fernández AR, Campos L, Gutierrez-Maldonado SE, Núñez G, Villanelo F, Perez-Acle T. Nanosecond Pulsed Electric Field (nsPEF): Opening the Biotechnological Pandora’s Box. Int J Mol Sci 2022; 23:ijms23116158. [PMID: 35682837 PMCID: PMC9181413 DOI: 10.3390/ijms23116158] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Nanosecond Pulsed Electric Field (nsPEF) is an electrostimulation technique first developed in 1995; nsPEF requires the delivery of a series of pulses of high electric fields in the order of nanoseconds into biological tissues or cells. They primary effects in cells is the formation of membrane nanopores and the activation of ionic channels, leading to an incremental increase in cytoplasmic Ca2+ concentration, which triggers a signaling cascade producing a variety of effects: from apoptosis up to cell differentiation and proliferation. Further, nsPEF may affect organelles, making nsPEF a unique tool to manipulate and study cells. This technique is exploited in a broad spectrum of applications, such as: sterilization in the food industry, seed germination, anti-parasitic effects, wound healing, increased immune response, activation of neurons and myocites, cell proliferation, cellular phenotype manipulation, modulation of gene expression, and as a novel cancer treatment. This review thoroughly explores both nsPEF’s history and applications, with emphasis on the cellular effects from a biophysics perspective, highlighting the role of ionic channels as a mechanistic driver of the increase in cytoplasmic Ca2+ concentration.
Collapse
Affiliation(s)
- Alvaro R. Ruiz-Fernández
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
- Correspondence: (A.R.R.-F.); (T.P.-A.)
| | - Leonardo Campos
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
| | - Sebastian E. Gutierrez-Maldonado
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
| | - Gonzalo Núñez
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
| | - Felipe Villanelo
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
| | - Tomas Perez-Acle
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
- Correspondence: (A.R.R.-F.); (T.P.-A.)
| |
Collapse
|
13
|
Yang L, Pierce S, Gould TW, Craviso GL, Leblanc N. Ultrashort nanosecond electric pulses activate a conductance in bovine adrenal chromaffin cells that involves cation entry through TRPC and NALCN channels. Arch Biochem Biophys 2022; 723:109252. [DOI: 10.1016/j.abb.2022.109252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/25/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022]
|
14
|
L Pall M. Low Intensity Electromagnetic Fields Act via Voltage-Gated Calcium Channel (VGCC) Activation to Cause Very Early Onset Alzheimer's Disease: 18 Distinct Types of Evidence. Curr Alzheimer Res 2022; 19:119-132. [PMID: 35114921 PMCID: PMC9189734 DOI: 10.2174/1567205019666220202114510] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/22/2021] [Accepted: 12/31/2021] [Indexed: 11/22/2022]
Abstract
Electronically generated electromagnetic fields (EMFs) including those used in wireless communication such as cell phones, Wi-Fi and smart meters, are coherent, producing very high electric and magnetic forces which act on the voltage sensor of voltage-gated calcium channels to produce increases in intracellular calcium [Ca2+]i. The calcium hypothesis of Alzheimer's disease (AD) has shown that each of the important AD-specific and nonspecific causal elements are produced by excessive [Ca2+]i. [Ca2+]i acts in AD via excessive calcium signaling and the peroxynitrite/oxidative stress/inflammation pathway which are each elevated by EMFs. An apparent vicious cycle in AD involves amyloid-beta protein (A) and [Ca2+]i. Three types of epidemiology each suggest EMF causation of AD including early onset AD. Extensive animal model studies show that low intensity EMFs cause neurodegeneration including AD, with AD animals having elevated levels of A, amyloid precursor protein and BACE1. Rats exposed to pulsed EMFs every day are reported to develop universal or near universal very very very early onset neurodegeneration including AD; these findings are superficially similar to humans with digital dementia. EMFs producing modest increases in [Ca2+]i can also produce protective, therapeutic effects. The therapeutic pathway and peroxynitrite pathway inhibit each other. A summary of 18 different findings is provided, which collectively provide powerful evidence for EMF causation of AD. The author is concerned that smarter, more highly pulsed "smart" wireless communication may cause widespread very, very early onset AD in human populations.
Collapse
Affiliation(s)
- Martin L Pall
- Professor Emeritus of Biochemistry & Basic Medical Sciences Washington State University Mailing Address: 638 NE 41stst Ave., Portland OR 97232, USA
| |
Collapse
|
15
|
Pulse Duration Dependent Asymmetry in Molecular Transmembrane Transport Due to Electroporation in H9c2 Rat Cardiac Myoblast Cells In Vitro. Molecules 2021; 26:molecules26216571. [PMID: 34770979 PMCID: PMC8588460 DOI: 10.3390/molecules26216571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/09/2021] [Accepted: 10/22/2021] [Indexed: 11/17/2022] Open
Abstract
Electroporation (EP) is one of the successful physical methods for intracellular drug delivery, which temporarily permeabilizes plasma membrane by exposing cells to electric pulses. Orientation of cells in electric field is important for electroporation and, consequently, for transport of molecules through permeabilized plasma membrane. Uptake of molecules after electroporation are the greatest at poles of cells facing electrodes and is often asymmetrical. However, asymmetry reported was inconsistent and inconclusive-in different reports it was either preferentially anodal or cathodal. We investigated the asymmetry of polar uptake of calcium ions after electroporation with electric pulses of different durations, as the orientation of elongated cells affects electroporation to a different extent when using electric pulses of different durations in the range of 100 ns to 100 µs. The results show that with 1, 10, and 100 µs pulses, the uptake of calcium ions is greater at the pole closer to the cathode than at the pole closer to the anode. With shorter 100 ns pulses, the asymmetry is not observed. A different extent of electroporation at different parts of elongated cells, such as muscle or cardiac cells, may have an impact on electroporation-based treatments such as drug delivery, pulse-field ablation, and gene electrotransfection.
Collapse
|
16
|
Liu H, Zhao Y, Yao C, Schmelz EM, Davalos RV. Differential effects of nanosecond pulsed electric fields on cells representing progressive ovarian cancer. Bioelectrochemistry 2021; 142:107942. [PMID: 34509872 DOI: 10.1016/j.bioelechem.2021.107942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022]
Abstract
Nanosecond pulsed electric fields (nsPEFs) may induce differential effects on tumor cells from different disease stages and could be suitable for treating tumors by preferentially targeting the late-stage/highly aggressive tumor cells. In this study, we investigated the nsPEF responses of mouse ovarian surface epithelial (MOSE) cells representing progressive ovarian cancer from benign to malignant stages and highly aggressive tumor-initiating-like cells. We established the cell-seeded 3D collagen scaffolds cultured with or without Nocodazole (eliminating the influence of cell proliferation on ablation outcome) to observe the ablation effects at 3 h and 24 h after treatment and compared the corresponding thresholds obtained by numerically calculated electric field distribution. The results showed that nsPEFs induced larger ablation areas with lower thresholds as the cell progress from benign, malignant to a highly aggressive phenotype. This differential effect was not affected by the different doubling times of the cells, as apparent by similar ablation induction after a synergistic treatment of nsPEFs and Nocodazole. The result suggests that nsPEFs could induce preferential ablation effects on highly aggressive and malignant ovarian cancer cells than their benign counterparts. This study provides an experimental basis for the research on killing malignant tumor cells via electrical treatments and may have clinical implications for treating tumors and preventing tumor recurrence after treatment.
Collapse
Affiliation(s)
- Hongmei Liu
- School of Electrical Engineering, Chongqing University, Chongqing 400033, China; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| | - Yajun Zhao
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; College of Electrical Engineering and Control Science, Nanjing Tech. University, Nanjing 211816, China
| | - Chenguo Yao
- School of Electrical Engineering, Chongqing University, Chongqing 400033, China.
| | - Eva M Schmelz
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
17
|
Zemlin CW. Safety factor for electrostimulation with nanosecond pulses. Bioelectrochemistry 2021; 141:107882. [PMID: 34274877 DOI: 10.1016/j.bioelechem.2021.107882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 06/08/2021] [Accepted: 06/28/2021] [Indexed: 11/19/2022]
Abstract
While electrical stimulation with pulses of milli- or microsecond duration is possible without electroporation, stimulation with nanosecond pulses typically entails electroporation, and nanosecond pulses can even cause electroporation without stimulation. A recently proposed explanation for this intriguing finding is that stimulation requires not only that a threshold membrane potential is reached, but also that it is sustained for a certain time tmin, while electroporation occurs almost immediately after a higher threshold potential is reached. Here we analytically derive stimulation and electroporation thresholds for membranes that satisfy these assumptions. We analyze the safety factor, i.e. the ratio between electroporation and stimulation threshold and its dependence on pulse duration, membrane charging time constant, and tmin. We find that the safety factor is sharply reduced if both the pulse duration and the membrane charging time constant are below tmin. We discuss different approaches to get models with varying tmin that could be used to experimentally test this theory and cardiac applications.
Collapse
Affiliation(s)
- Christian W Zemlin
- Division of Cardiothoracic Surgery, Washington University School of Medicine, Campus Box 8234, 660 S Euclid Ave, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
Exploring the Conformational Changes Induced by Nanosecond Pulsed Electric Fields on the Voltage Sensing Domain of a Ca 2+ Channel. MEMBRANES 2021; 11:membranes11070473. [PMID: 34206827 PMCID: PMC8303878 DOI: 10.3390/membranes11070473] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/21/2022]
Abstract
Nanosecond Pulsed Electric Field (nsPEF or Nano Pulsed Stimulation, NPS) is a technology that delivers a series of pulses of high-voltage electric fields during a short period of time, in the order of nanoseconds. The main consequence of nsPEF upon cells is the formation of nanopores, which is followed by the gating of ionic channels. Literature is conclusive in that the physiological mechanisms governing ion channel gating occur in the order of milliseconds. Hence, understanding how these channels can be activated by a nsPEF would be an important step in order to conciliate fundamental biophysical knowledge with improved nsPEF applications. To get insights on both the kinetics and thermodynamics of ion channel gating induced by nsPEF, in this work, we simulated the Voltage Sensing Domain (VSD) of a voltage-gated Ca2+ channel, inserted in phospholipidic membranes with different concentrations of cholesterol. We studied the conformational changes of the VSD under a nsPEF mimicked by the application of a continuous electric field lasting 50 ns with different intensities as an approach to reveal novel mechanisms leading to ion channel gating in such short timescales. Our results show that using a membrane with high cholesterol content, under an nsPEF of 50 ns and E→ = 0.2 V/nm, the VSD undergoes major conformational changes. As a whole, our work supports the notion that membrane composition may act as an allosteric regulator, specifically cholesterol content, which is fundamental for the response of the VSD to an external electric field. Moreover, changes on the VSD structure suggest that the gating of voltage-gated Ca2+ channels by a nsPEF may be due to major conformational changes elicited in response to the external electric field. Finally, the VSD/cholesterol-bilayer under an nsPEF of 50 ns and E→ = 0.2 V/nm elicits a pore formation across the VSD suggesting a new non-reported effect of nsPEF into cells, which can be called a “protein mediated electroporation”.
Collapse
|
19
|
Interference targeting of bipolar nanosecond electric pulses for spatially focused electroporation, electrostimulation, and tissue ablation. Bioelectrochemistry 2021; 141:107876. [PMID: 34171507 DOI: 10.1016/j.bioelechem.2021.107876] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/17/2022]
Abstract
Stimulation and electroporation by nanosecond electric pulses (nsEP) are distinguished by a phenomenon of bipolar cancellation, which stands for a reduced efficiency of bipolar pulses compared to unipolar ones. When two pairs of stimulating electrodes are arrayed in a quadrupole, bipolar cancellation inhibits nsEP effects near the electrodes, where the electric field is the strongest. Two properly shaped and synchronized bipolar nsEP overlay into a unipolar pulse towards the center of the electrode array, thus canceling the bipolar cancellation (a "CANCAN effect"). High efficiency of the re-created unipolar nsEP outweighs the weakening of the electric field with distance and focuses nsEP effects to the center. In monolayers of CHO, BPAE, and HEK cells, CANCAN effect achieved by the interference of two bipolar nsEP enhanced electroporation up to tenfold, with a peak at the quadrupole center. Introducing a time interval between bipolar nsEP prevented the formation of a unipolar pulse and eliminated the CANCAN effect. Strong electroporation by CANCAN stimuli killed cells over the entire area encompassed by the electrodes, whereas the time-separated pulses caused ablation only in the strongest electric field near the electrodes. The CANCAN approach is promising for uniform tumor ablation and stimulation targeting away from electrodes.
Collapse
|
20
|
Aguilar AA, Ho MC, Chang E, Carlson KW, Natarajan A, Marciano T, Bomzon Z, Patel CB. Permeabilizing Cell Membranes with Electric Fields. Cancers (Basel) 2021; 13:2283. [PMID: 34068775 PMCID: PMC8126200 DOI: 10.3390/cancers13092283] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/29/2022] Open
Abstract
The biological impact of exogenous, alternating electric fields (AEFs) and direct-current electric fields has a long history of study, ranging from effects on embryonic development to influences on wound healing. In this article, we focus on the application of electric fields for the treatment of cancers. In particular, we outline the clinical impact of tumor treating fields (TTFields), a form of AEFs, on the treatment of cancers such as glioblastoma and mesothelioma. We provide an overview of the standard mechanism of action of TTFields, namely, the capability for AEFs (e.g., TTFields) to disrupt the formation and segregation of the mitotic spindle in actively dividing cells. Though this standard mechanism explains a large part of TTFields' action, it is by no means complete. The standard theory does not account for exogenously applied AEFs' influence directly upon DNA nor upon their capacity to alter the functionality and permeability of cancer cell membranes. This review summarizes the current literature to provide a more comprehensive understanding of AEFs' actions on cell membranes. It gives an overview of three mechanistic models that may explain the more recent observations into AEFs' effects: the voltage-gated ion channel, bioelectrorheological, and electroporation models. Inconsistencies were noted in both effective frequency range and field strength between TTFields versus all three proposed models. We addressed these discrepancies through theoretical investigations into the inhomogeneities of electric fields on cellular membranes as a function of disease state, external microenvironment, and tissue or cellular organization. Lastly, future experimental strategies to validate these findings are outlined. Clinical benefits are inevitably forthcoming.
Collapse
Affiliation(s)
- Alondra A. Aguilar
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Michelle C. Ho
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Edwin Chang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Kristen W. Carlson
- Beth Israel Deaconess Medical Center, Department of Neurosurgery, Harvard Medical School, Boston, MA 02215, USA;
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Tal Marciano
- Novocure, Ltd., 31905 Haifa, Israel; (T.M.); (Z.B.)
| | - Ze’ev Bomzon
- Novocure, Ltd., 31905 Haifa, Israel; (T.M.); (Z.B.)
| | - Chirag B. Patel
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
- Department of Neurology & Neurological Sciences, Division of Neuro-Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
21
|
5 ns electric pulses induce Ca 2+-dependent exocytotic release of catecholamine from adrenal chromaffin cells. Bioelectrochemistry 2021; 140:107830. [PMID: 33965669 DOI: 10.1016/j.bioelechem.2021.107830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
Previously we reported that adrenal chromaffin cells exposed to a 5 ns, 5 MV/m pulse release the catecholamines norepinephrine (NE) and epinephrine (EPI) in a Ca2+-dependent manner. Here we determined that NE and EPI release increased with pulse number (one versus five and ten pulses at 1 Hz), established that release occurs by exocytosis, and characterized the exocytotic response in real-time. Evidence of an exocytotic mechanism was the appearance of dopamine-β-hydroxylase on the plasma membrane, and the demonstration by total internal reflection fluorescence microscopy studies that a train of five or ten pulses at 1 Hz triggered the release of the fluorescent dye acridine orange from secretory granules. Release events were Ca2+-dependent, longer-lived relative to those evoked by nicotinic receptor stimulation, and occurred with a delay of several seconds despite an immediate rise in Ca2+. In complementary studies, cells labeled with the plasma membrane fluorescent dye FM 1-43 and exposed to a train of ten pulses at 1 Hz underwent Ca2+-dependent increases in FM 1-43 fluorescence indicative of granule fusion with the plasma membrane due to exocytosis. These results demonstrate the effectiveness of ultrashort electric pulses for stimulating catecholamine release, signifying their promise as a novel electrostimulation modality for neurosecretion.
Collapse
|
22
|
Cantu JC, Tolstykh GP, Tarango M, Beier HT, Ibey BL. Caveolin-1 is Involved in Regulating the Biological Response of Cells to Nanosecond Pulsed Electric Fields. J Membr Biol 2021; 254:141-156. [PMID: 33427940 DOI: 10.1007/s00232-020-00160-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/18/2020] [Indexed: 01/20/2023]
Abstract
Nanosecond pulsed electric fields (nsPEFs) induce changes in the plasma membrane (PM), including PM permeabilization (termed nanoporation), allowing free passage of ions into the cell and, in certain cases, cell death. Recent studies from our laboratory show that the composition of the PM is a critical determinant of PM nanoporation. Thus, we hypothesized that the biological response to nsPEF exposure could be influenced by lipid microdomains, including caveolae, which are specialized invaginations of the PM that are enriched in cholesterol and contain aggregates of important cell signaling proteins, such as caveolin-1 (Cav1). Caveolae play a significant role in cellular signal transduction, including control of calcium influx and cell death by interaction of Cav1 with regulatory signaling proteins. Present results show that depletion of Cav1 increased the influx of calcium, while Cav1 overexpression produced the opposite effect. Additionally, Cav1 is known to bind and sequester important cell signaling proteins within caveolae, rendering the binding partners inactive. Imaging of the PM after nsPEF exposure showed localized depletion of PM Cav1 and results of co-immunoprecipitation studies showed dissociation of two critical Cav1 binding partners (transient receptor potential cation channel subfamily C1 (TRPC1) and inositol trisphosphate receptor (IP3R)) after exposure to nsPEFs. Release of TRPC1 and IP3R from Cav1 would activate downstream signaling cascades, including store-operated calcium entry, which could explain the influx in calcium after nsPEF exposure. Results of the current study establish a significant relationship between Cav1 and the activation of cell signaling pathways in response to nsPEFs.
Collapse
Affiliation(s)
- Jody C Cantu
- General Dynamics Information Technology, JBSA Fort Sam Houston, 4141 Petroleum Road, Bldg. 3260, San Antonio, TX, 78234-2644, USA.
| | - Gleb P Tolstykh
- General Dynamics Information Technology, JBSA Fort Sam Houston, 4141 Petroleum Road, Bldg. 3260, San Antonio, TX, 78234-2644, USA
| | - Melissa Tarango
- General Dynamics Information Technology, JBSA Fort Sam Houston, 4141 Petroleum Road, Bldg. 3260, San Antonio, TX, 78234-2644, USA
| | - Hope T Beier
- Air Force Research Laboratory, 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, Optical Radiation Bioeffects Branch, JBSA Fort Sam Houston, San Antonio, TX, 78234, USA
| | - Bennett L Ibey
- Air Force Research Laboratory, 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, Radio Frequency Bioeffects Branch, JBSA Fort Sam Houston, San Antonio, TX, 78234, USA
| |
Collapse
|
23
|
2-ns Electrostimulation of Ca 2+ Influx into Chromaffin Cells: Rapid Modulation by Field Reversal. Biophys J 2020; 120:556-567. [PMID: 33359835 PMCID: PMC7895993 DOI: 10.1016/j.bpj.2020.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/01/2020] [Accepted: 12/16/2020] [Indexed: 12/15/2022] Open
Abstract
Cellular effects of nanosecond-pulsed electric field exposures can be attenuated by an electric field reversal, a phenomenon called bipolar pulse cancellation. Our investigations of this phenomenon in neuroendocrine adrenal chromaffin cells show that a single 2-ns, 16 MV/m unipolar pulse elicited a rapid, transient rise in intracellular Ca2+ levels due to Ca2+ influx through voltage-gated calcium channels. The response was eliminated by a 2-ns bipolar pulse with positive and negative phases of equal duration and amplitude and fully restored (unipolar-equivalent response) when the delay between each phase of the bipolar pulse was 30 ns. Longer interphase intervals evoked Ca2+ responses that were greater in magnitude than those evoked by a unipolar pulse (stimulation). Cancellation was also observed when the amplitude of the second (negative) phase of the bipolar pulse was half that of the first (positive) phase but progressively lost as the amplitude of the second phase was incrementally increased above that of the first phase. When the amplitude of the second phase was twice that of the first phase, there was stimulation. By comparing the experimental results for each manipulation of the bipolar pulse waveform with analytical calculations of capacitive membrane charging/discharging, also known as accelerated membrane discharge mechanism, we show that the transition from cancellation to unipolar-equivalent stimulation broadly agrees with this model. Taken as a whole, our results demonstrate that electrostimulation of adrenal chromaffin cells with ultrashort pulses can be modulated with interphase intervals of tens of nanoseconds, a prediction of the accelerated membrane discharge mechanism not previously observed in other bipolar pulse cancellation studies. Such modulation of Ca2+ responses in a neural-type cell is promising for the potential use of nanosecond bipolar pulse technologies for remote electrostimulation applications for neuromodulation.
Collapse
|
24
|
Pakhomov AG, Pakhomova ON. The interplay of excitation and electroporation in nanosecond pulse stimulation. Bioelectrochemistry 2020; 136:107598. [PMID: 32711366 DOI: 10.1016/j.bioelechem.2020.107598] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Conventional electric stimuli of micro- and millisecond duration excite or activate cells at voltages 10-100 times below the electroporation threshold. This ratio is remarkably different for nanosecond electric pulses (nsEP), which caused excitation and activation only at or above the electroporation threshold in diverse cell lines, primary cardiomyocytes, neurons, and chromaffin cells. Depolarization to the excitation threshold often results from (or is assisted by) the loss of the resting membrane potential due to ion leaks across the membrane permeabilized by nsEP. Slow membrane resealing and the build-up of electroporation damages prevent repetitive excitation by nsEP. However, peripheral nerves and muscles are exempt from this rule and withstand multiple cycles of excitation by nsEP without the loss of function or signs of electroporation. We show that the damage-free excitation by nsEP may be enabled by the membrane charging time constant sufficiently large to (1) cap the peak transmembrane voltage during nsEP below the electroporation threshold, and (2) extend the post-nsEP depolarization long enough to activate voltage-gated ion channels. The low excitatory efficacy of nsEP compared to longer pulses makes them advantageous for medical applications where the neuromuscular excitation is an unwanted side effect, such as electroporation-based cancer and tissue ablation.
Collapse
Affiliation(s)
- Andrei G Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA.
| | - Olga N Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| |
Collapse
|
25
|
Yang L, Pierce S, Chatterjee I, Craviso GL, Leblanc N. Paradoxical effects on voltage-gated Na+ conductance in adrenal chromaffin cells by twin vs single high intensity nanosecond electric pulses. PLoS One 2020; 15:e0234114. [PMID: 32516325 PMCID: PMC7282663 DOI: 10.1371/journal.pone.0234114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/19/2020] [Indexed: 01/17/2023] Open
Abstract
We previously reported that a single 5 ns high intensity electric pulse (NEP) caused an E-field-dependent decrease in peak inward voltage-gated Na+ current (INa) in isolated bovine adrenal chromaffin cells. This study explored the effects of a pair of 5 ns pulses on INa recorded in the same cell type, and how varying the E-field amplitude and interval between the pulses altered its response. Regardless of the E-field strength (5 to 10 MV/m), twin NEPs having interpulse intervals ≥ than 5 s caused the inhibition of TTX-sensitive INa to approximately double relative to that produced by a single pulse. However, reducing the interval from 1 s to 10 ms between twin NEPs at E-fields of 5 and 8 MV/m but not 10 MV/m decreased the magnitude of the additive inhibitory effect by the second pulse in a pair on INa. The enhanced inhibitory effects of twin vs single NEPs on INa were not due to a shift in the voltage-dependence of steady-state activation and inactivation but were associated with a reduction in maximal Na+ conductance. Paradoxically, reducing the interval between twin NEPs at 5 or 8 MV/m but not 10 MV/m led to a progressive interval-dependent recovery of INa, which after 9 min exceeded the level of INa reached following the application of a single NEP. Disrupting lipid rafts by depleting membrane cholesterol with methyl-β-cyclodextrin enhanced the inhibitory effects of twin NEPs on INa and ablated the progressive recovery of this current at short twin pulse intervals, suggesting a complete dissociation of the inhibitory effects of twin NEPs on this current from their ability to stimulate its recovery. Our results suggest that in contrast to a single NEP, twin NEPs may influence membrane lipid rafts in a manner that enhances the trafficking of newly synthesized and/or recycling of endocytosed voltage-gated Na+ channels, thereby pointing to novel means to regulate ion channels in excitable cells.
Collapse
Affiliation(s)
- Lisha Yang
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, United States of America
| | - Sophia Pierce
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, United States of America
| | - Indira Chatterjee
- Department of Electrical and Biomedical Engineering, College of Engineering, University of Nevada, Reno, NV, United States of America
| | - Gale L. Craviso
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, United States of America
| | - Normand Leblanc
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, United States of America
| |
Collapse
|
26
|
Rems L, Kasimova MA, Testa I, Delemotte L. Pulsed Electric Fields Can Create Pores in the Voltage Sensors of Voltage-Gated Ion Channels. Biophys J 2020; 119:190-205. [PMID: 32559411 PMCID: PMC7335976 DOI: 10.1016/j.bpj.2020.05.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/31/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
Pulsed electric fields are increasingly used in medicine to transiently increase the cell membrane permeability via electroporation to deliver therapeutic molecules into the cell. One type of event that contributes to this increase in membrane permeability is the formation of pores in the membrane lipid bilayer. However, electrophysiological measurements suggest that membrane proteins are affected as well, particularly voltage-gated ion channels (VGICs). The molecular mechanisms by which the electric field could affects these molecules remain unidentified. In this study, we used molecular dynamics simulations to unravel the molecular events that take place in different VGICs when exposing them to electric fields mimicking electroporation conditions. We show that electric fields can induce pores in the voltage-sensor domains (VSDs) of different VGICs and that these pores form more easily in some channels than in others. We demonstrate that poration is more likely in VSDs that are more hydrated and are electrostatically more favorable for the entry of ions. We further show that pores in VSDs can expand into so-called complex pores, which become stabilized by lipid headgroups. Our results suggest that such complex pores are considerably more stable than conventional lipid pores, and their formation can lead to severe unfolding of VSDs from the channel. We anticipate that such VSDs become dysfunctional and unable to respond to changes in transmembrane voltage, which is in agreement with previous electrophysiological measurements showing a decrease in the voltage-dependent transmembrane ionic currents after pulse treatment. Finally, we discuss the possibility of activation of VGICs by submicrosecond-duration pulses. Overall, our study reveals a new, to our knowledge, mechanism of electroporation through membranes containing VGICs.
Collapse
Affiliation(s)
- Lea Rems
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Marina A Kasimova
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Ilaria Testa
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Solna, Sweden.
| |
Collapse
|
27
|
Probing Nanoelectroporation and Resealing of the Cell Membrane by the Entry of Ca 2+ and Ba 2+ Ions. Int J Mol Sci 2020; 21:ijms21093386. [PMID: 32403282 PMCID: PMC7247012 DOI: 10.3390/ijms21093386] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022] Open
Abstract
The principal bioeffect of the nanosecond pulsed electric field (nsPEF) is a lasting cell membrane permeabilization, which is often attributed to the formation of nanometer-sized pores. Such pores may be too small for detection by the uptake of fluorescent dyes. We tested if Ca2+, Cd2+, Zn2+, and Ba2+ ions can be used as nanoporation markers. Time-lapse imaging was performed in CHO, BPAE, and HEK cells loaded with Fluo-4, Calbryte, or Fluo-8 dyes. Ca2+ and Ba2+ did not change fluorescence in intact cells, whereas their entry after nsPEF increased fluorescence within <1 ms. The threshold for one 300-ns pulse was at 1.5–2 kV/cm, much lower than >7 kV/cm for the formation of larger pores that admitted YO-PRO-1, TO-PRO-3, or propidium dye into the cells. Ba2+ entry caused a gradual emission rise, which reached a stable level in 2 min or, with more intense nsPEF, kept rising steadily for at least 30 min. Ca2+ entry could elicit calcium-induced calcium release (CICR) followed by Ca2+ removal from the cytosol, which markedly affected the time course, polarity, amplitude, and the dose-dependence of fluorescence change. Both Ca2+ and Ba2+ proved as sensitive nanoporation markers, with Ba2+ being more reliable for monitoring membrane damage and resealing.
Collapse
|
28
|
Nanosecond Pulsed Electric Fields Induce Endoplasmic Reticulum Stress Accompanied by Immunogenic Cell Death in Murine Models of Lymphoma and Colorectal Cancer. Cancers (Basel) 2019; 11:cancers11122034. [PMID: 31861079 PMCID: PMC6966635 DOI: 10.3390/cancers11122034] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Depending on the initiating stimulus, cancer cell death can be immunogenic or non-immunogenic. Inducers of immunogenic cell death (ICD) rely on endoplasmic reticulum (ER) stress for the trafficking of danger signals such as calreticulin (CRT) and ATP. We found that nanosecond pulsed electric fields (nsPEF), an emerging new modality for tumor ablation, cause the activation of the ER-resident stress sensor PERK in both CT-26 colon carcinoma and EL-4 lymphoma cells. PERK activation correlates with sustained CRT exposure on the cell plasma membrane and apoptosis induction in both nsPEF-treated cell lines. Our results show that, in CT-26 cells, the activity of caspase-3/7 was increased fourteen-fold as compared with four-fold in EL-4 cells. Moreover, while nsPEF treatments induced the release of the ICD hallmark HMGB1 in both cell lines, extracellular ATP was detected only in CT-26. Finally, in vaccination assays, CT-26 cells treated with nsPEF or doxorubicin equally impaired the growth of tumors at challenge sites eliciting a protective anticancer immune response in 78% and 80% of the animals, respectively. As compared to CT-26, both nsPEF- and mitoxantrone-treated EL-4 cells had a less pronounced effect and protected 50% and 20% of the animals, respectively. These results support our conclusion that nsPEF induce ER stress, accompanied by bona fide ICD.
Collapse
|
29
|
Bagalkot TR, Leblanc N, Craviso GL. Stimulation or Cancellation of Ca 2+ Influx by Bipolar Nanosecond Pulsed Electric Fields in Adrenal Chromaffin Cells Can Be Achieved by Tuning Pulse Waveform. Sci Rep 2019; 9:11545. [PMID: 31395918 PMCID: PMC6687888 DOI: 10.1038/s41598-019-47929-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 07/26/2019] [Indexed: 12/30/2022] Open
Abstract
Exposing adrenal chromaffin cells to single 150 to 400 ns electric pulses triggers a rise in intracellular Ca2+ ([Ca2+]i) that is due to Ca2+ influx through voltage-gated Ca2+ channels (VGCC) and plasma membrane electropores. Immediate delivery of a second pulse of the opposite polarity in which the duration and amplitude were the same as the first pulse (a symmetrical bipolar pulse) or greater than the first pulse (an asymmetrical bipolar pulse) had a stimulatory effect, evoking larger Ca2+ responses than the corresponding unipolar pulse. Progressively decreasing the amplitude of the opposite polarity pulse while also increasing its duration converted stimulation to attenuation, which reached a maximum of 43% when the positive phase was 150 ns at 3.1 kV/cm, and the negative phase was 800 ns at 0.2 kV/cm. When VGCCs were blocked, Ca2+ responses evoked by asymmetrical and even symmetrical bipolar pulses were significantly reduced relative to those evoked by the corresponding unipolar pulse under the same conditions, indicating that attenuation involved mainly the portion of Ca2+ influx attributable to membrane electropermeabilization. Thus, by tuning the shape of the bipolar pulse, Ca2+ entry into chromaffin cells through electropores could be attenuated while preserving Ca2+ influx through VGCCs.
Collapse
Affiliation(s)
- Tarique R Bagalkot
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA.
| | - Normand Leblanc
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Gale L Craviso
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
30
|
Ultrashort nanosecond electric pulses evoke heterogeneous patterns of Ca 2+ release from the endoplasmic reticulum of adrenal chromaffin cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1180-1188. [PMID: 30986385 DOI: 10.1016/j.bbamem.2019.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 01/19/2023]
|
31
|
Lamberti P, Compitiello M, Romeo S, Lamberti P, Compitiello M, Romeo S, Lamberti P, Romeo S, Compitiello M. ns Pulsed Electric Field-Induced Action Potentials in the Circuital Model of an Axon. IEEE Trans Nanobioscience 2019; 17:110-116. [PMID: 29870334 DOI: 10.1109/tnb.2018.2822840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pulsed electric fields with duration in the sub- and ns time scale (nsPEFs) increase the permeability of cell membranes, enabling the transport of normally impermeant molecules into or out of the cell (electroporation). Such effect is associated to intracellular alterations and indicates nsPEFs as a new stimulus to modulate cell functions. In particular, studies dealing with the application of nsPEFs to excitable cells suggest their use for the stimulation/inhibition of cell excitation. In this paper, the circuital model per surface unit of the plasma membrane of an axon was developed to implement the Hodgkin and Huxley equations, describing the action potential activation process. For the first time, a power electronics circuital simulator was adopted. The model was first validated with conventional microsecond stimuli, and then it was employed to identify the conditions for cell excitation by nsPEFs. The results demonstrated the possibility of electrostimulation by nsPEFs at depolarization levels far below those required for inducing electroporation, and with ionic current dynamics similar to that induced by conventional stimuli, confirming recent experimental findings. Moreover, by using a power electronics tool, easier integration of the cell modeling with the design and optimization of pulse generation systems can be gained.
Collapse
|
32
|
Kotnik T, Rems L, Tarek M, Miklavčič D. Membrane Electroporation and Electropermeabilization: Mechanisms and Models. Annu Rev Biophys 2019; 48:63-91. [PMID: 30786231 DOI: 10.1146/annurev-biophys-052118-115451] [Citation(s) in RCA: 399] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Exposure of biological cells to high-voltage, short-duration electric pulses causes a transient increase in their plasma membrane permeability, allowing transmembrane transport of otherwise impermeant molecules. In recent years, large steps were made in the understanding of underlying events. Formation of aqueous pores in the lipid bilayer is now a widely recognized mechanism, but evidence is growing that changes to individual membrane lipids and proteins also contribute, substantiating the need for terminological distinction between electroporation and electropermeabilization. We first revisit experimental evidence for electrically induced membrane permeability, its correlation with transmembrane voltage, and continuum models of electropermeabilization that disregard the molecular-level structure and events. We then present insights from molecular-level modeling, particularly atomistic simulations that enhance understanding of pore formation, and evidence of chemical modifications of membrane lipids and functional modulation of membrane proteins affecting membrane permeability. Finally, we discuss the remaining challenges to our full understanding of electroporation and electropermeabilization.
Collapse
Affiliation(s)
- Tadej Kotnik
- Faculty of Electrical Engineering, University of Ljubljana, SI-1000 Ljubljana, Slovenia; ,
| | - Lea Rems
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, 17165 Solna, Sweden;
| | - Mounir Tarek
- Université de Lorraine, CNRS, LPCT, F-54000 Nancy, France;
| | - Damijan Miklavčič
- Faculty of Electrical Engineering, University of Ljubljana, SI-1000 Ljubljana, Slovenia; ,
| |
Collapse
|
33
|
Mechanisms and immunogenicity of nsPEF-induced cell death in B16F10 melanoma tumors. Sci Rep 2019; 9:431. [PMID: 30674926 PMCID: PMC6344591 DOI: 10.1038/s41598-018-36527-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
Accumulating data indicates that some cancer treatments can restore anticancer immunosurveillance through the induction of tumor immunogenic cell death (ICD). Nanosecond pulsed electric fields (nsPEF) have been shown to efficiently ablate melanoma tumors. In this study we investigated the mechanisms and immunogenicity of nsPEF-induced cell death in B16F10 melanoma tumors. Our data show that in vitro nsPEF (20–200, 200-ns pulses, 7 kV/cm, 2 Hz) caused a rapid dose-dependent cell death which was not accompanied by caspase activation or PARP cleavage. The lack of nsPEF-induced apoptosis was confirmed in vivo in B16F10 tumors. NsPEF also failed to trigger ICD-linked responses such as necroptosis and autophagy. Our results point at necrosis as the primary mechanism of cell death induced by nsPEF in B16F10 cells. We finally compared the antitumor immunity in animals treated with nsPEF (750, 200-ns, 25 kV/cm, 2 Hz) with animals were tumors were surgically removed. Compared to the naïve group where all animals developed tumors, nsPEF and surgery protected 33% (6/18) and 28.6% (4/14) of the animals, respectively. Our data suggest that, under our experimental conditions, the local ablation by nsPEF restored but did not boost the natural antitumor immunity which stays dormant in the tumor-bearing host.
Collapse
|
34
|
Azarov JE, Semenov I, Casciola M, Pakhomov AG. Excitation of murine cardiac myocytes by nanosecond pulsed electric field. J Cardiovasc Electrophysiol 2019; 30:392-401. [PMID: 30582656 DOI: 10.1111/jce.13834] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Opening of voltage-gated sodium channels takes tens to hundreds of microseconds, and mechanisms of their opening by nanosecond pulsed electric field (nsPEF) stimuli remain elusive. This study was aimed at uncovering the mechanisms of how nsPEF elicits action potentials (APs) in cardiomyocytes. METHODS AND RESULTS Fluorescent imaging of optical APs (FluoVolt) and Ca2+ -transients (Fluo-4) was performed in enzymatically isolated murine ventricular cardiomyocytes stimulated by 200-nanosecond trapezoidal pulses. nsPEF stimulation evoked tetrodotoxin-sensitive APs accompanied or preceded by slow sustained depolarization (SSD) and, in most cells, by transient afterdepolarization waves. SSD threshold was lower than the AP threshold (1.26 ± 0.03 vs 1.34 ± 0.03 kV/cm, respectively, P < 0.001). Inhibition of l-type calcium and sodium-calcium exchanger currents reduced the SSD amplitude and increased the AP threshold ( P < 0.05). The threshold for Ca 2+ -transients (1.40 ± 0.04 kV/cm) was not significantly affected by a tetrodotoxin-verapamil cocktail, suggesting the activation of a Ca 2+ entry pathway independent from the opening of Na + or Ca 2+ voltage-gated channels. Removal of external Ca 2+ decreased the SSD amplitude ( P = 0.004) and blocked Ca 2+ -transients but not APs. The incidence of transient afterdepolarization waves was decreased by verapamil and by removal of external Ca 2+ ( P = 0.002). CONCLUSIONS The study established that nsPEF stimulation caused calcium entry into cardiac myocytes (including routes other than voltage-gated calcium channels) and SSD. Tetrodotoxin-sensitive APs were mediated by SSD, whose amplitude depended on the calcium entry. Plasma membrane electroporation was the most likely primary mechanism of SSD with additional contribution from l-type calcium and sodium-calcium exchanger currents.
Collapse
Affiliation(s)
- Jan E Azarov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia.,Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, Syktyvkar, Russia.,Department of Physiology, Medical Institute of Pitirim Sorokin Syktyvkar State University, Syktyvkar, Russia
| | - Iurii Semenov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Maura Casciola
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Andrei G Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
35
|
Tolstykh GP, Cantu JC, Tarango M, Ibey BL. Receptor- and store-operated mechanisms of calcium entry during the nanosecond electric pulse-induced cellular response. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:685-696. [PMID: 30552899 DOI: 10.1016/j.bbamem.2018.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 11/16/2022]
Abstract
Nanosecond electric pulses have been shown to open nanopores in the cell plasma membrane by fluorescent imaging of calcium uptake and fluorescent dyes, including propidium (Pr) iodide and YO-PRO-1 (YP1). Recently, we demonstrated that nsEPs also induce the phosphoinositide intracellular signaling cascade by phosphatidylinositol-4,5-bisphosphate (PIP2) depletion resulting in physiological responses similar to those observed following stimulation of Gq11-coupled receptors. In this paper, we explore the role of receptor- and store-operated calcium entry (ROCE/SOCE) mechanisms in the observed response of cells to nsEP. We show that addition of the ROCE/SOCE and transient receptor potential channel (TRPC) blocker gadolinium (Gd3+, 300 μM) slows PIP2 depletion following 1 and 20 nsEP exposures. Lipid rafts, regions of the plasma membrane rich in PIP2 and TRPC, are also disrupted by nsEP exposure suggesting that ROCE/SOCE mechanisms are likely impacted. Reducing the expression of stromal interaction molecule 1 (STIM1) protein, a key protein in ROCE and SOCE, in cells exposure to nsEP resulted in a reduction in induced intracellular calcium rise. Additionally, after exposure to 1 and 20 nsEPs (16.2 kV/cm, 5 Hz), intracellular calcium rises were significantly reduced by the addition of GD3+ and SKF-96365 (1-[2-(4-methoxyphenyl)-2-[3-(4-methoxyphenyl) propoxy] ethyl-1H-imidazole hydrochloride, 100 μM), a blocker of STIM1 interaction. However, using similar nsEP exposure parameters, SKF-96365 was less effective at reducing YP1 uptake compared to Gd3+. Thus, it is possible that SKF-96365 could block STIM1 interactions within the cell, while Gd3+ could acts on TRPC/nanopores from outside of the cell. Our results present evidence of nsEP induces ROCE and SOCE mechanisms and demonstrate that YP1 and Ca2+ cannot be used solely as markers of nsEP-induced nanoporation.
Collapse
Affiliation(s)
- Gleb P Tolstykh
- General Dynamics Information Technology, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA.
| | - Jody C Cantu
- General Dynamics Information Technology, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| | - Melissa Tarango
- General Dynamics Information Technology, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| | - Bennett L Ibey
- Air Force Research Laboratory, 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, Radio Frequency Bioeffects Branch, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| |
Collapse
|
36
|
Zhou P, He F, Han Y, Liu B, Wei S. Nanosecond pulsed electric field induces calcium mobilization in osteoblasts. Bioelectrochemistry 2018; 124:7-12. [DOI: 10.1016/j.bioelechem.2018.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 01/19/2023]
|
37
|
Marracino P, Bernardi M, Liberti M, Del Signore F, Trapani E, Gárate JA, Burnham CJ, Apollonio F, English NJ. Transprotein-Electropore Characterization: A Molecular Dynamics Investigation on Human AQP4. ACS OMEGA 2018; 3:15361-15369. [PMID: 30556005 PMCID: PMC6288775 DOI: 10.1021/acsomega.8b02230] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/22/2018] [Indexed: 06/09/2023]
Abstract
Electroporation characterization is a topic of intensive interest probed by extensive ongoing research efforts. Usually, these studies are carried out on lipid-bilayer electroporation. Surprisingly, the possibility of water-channel electropore formation across transmembrane proteins themselves, particularly in view of such a promising application, has not yet been elucidated. The present work examines the geometrical and kinetic aspects of electropores and their stability in such a protein milieux (as opposed through the phospholipid membranes) in depth, by means of scrutiny of such a process in human-AQP4 as a well-representative prototype. The residues forming the electropore's walls, organized in loops, reveal the formation mechanism by their dipole alignment and translational response in response to applied axial electric fields in nonequilibrium molecular dynamics simulation. The magnitude of sustaining electric fields (keeping a stable electropore open) were determined. This suggests that transmembrane proteins could play a central role in electroporation applications, e.g., in medicine and biotechnology.
Collapse
Affiliation(s)
- Paolo Marracino
- Department
of Information Engineering, Electronics and Telecommunications, La Sapienza University, 00184 Rome, Italy
| | - Mario Bernardi
- Department
of Information Engineering, Electronics and Telecommunications, La Sapienza University, 00184 Rome, Italy
| | - Micaela Liberti
- Department
of Information Engineering, Electronics and Telecommunications, La Sapienza University, 00184 Rome, Italy
| | - Federico Del Signore
- Department
of Information Engineering, Electronics and Telecommunications, La Sapienza University, 00184 Rome, Italy
| | - Erika Trapani
- Department
of Information Engineering, Electronics and Telecommunications, La Sapienza University, 00184 Rome, Italy
| | - José-Antonio Gárate
- Centro
Interdisciplinario de neurociencia de Valparaiso, Universidad de Valparaiso, 05101 Valparaiso, Chile
| | - Christian J. Burnham
- School
of Chemical and Bioprocess Engineering, University College Dublin, Belfield, D4 Dublin, Ireland
| | - Francesca Apollonio
- Department
of Information Engineering, Electronics and Telecommunications, La Sapienza University, 00184 Rome, Italy
| | - Niall J. English
- School
of Chemical and Bioprocess Engineering, University College Dublin, Belfield, D4 Dublin, Ireland
| |
Collapse
|
38
|
Guo Y, Lv Q, Zou XQ, Yan ZX, Yan YX. Mechanical Strain Regulates Osteoblast Proliferation Through Ca 2+-CaMK-CREB Signal Pathway. ACTA ACUST UNITED AC 2018; 31:100-106. [PMID: 28031098 DOI: 10.1016/s1001-9294(16)30033-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Objective To investigate the effects of mechanical strain on Ca2+-calmodulin dependent kinase (CaMK)-cAMP response element binding protein (CREB) signal pathway and proliferation of osteoblasts.Methods Using a four-point bending device, MC3T3-E1 cells were exposed to mechanical tensile strains of 2500 µs and 5000 µs at 0.5 Hz respectively. The intracellular free Ca2+ ([Ca2+]i) concentration and calmodulin activity were assayed by fluorospectrophotometry, CaMK II β, CREB, and phosphorylated (activated) CREB (p-CREB) were assessed by Western blot, and cells proliferation was assayed with MTT. Pretreatment with verapamil was carried out to block Ca2+ channel, and inhibitor U73122 was used to inhibit phospholipase C (PLC).Results Mechanical strains of 2500 µs and 5000 µs for 1 to 10 minutes both increased [Ca2+]i level of the cells. The 2500 µs strain, a periodicity of 1 h/d for 3 days, activated calmodulin, elevated protein levels of CaMK II β and p-CREB, and promoted cells proliferation, which were attenuated by pretreatment of verapamil or U73122. The effects of 5000 µs strain on calmodulin, CaMK II β, p-CREB and proliferation were contrary to 2500 µs strain.Conclusion The mechanical strain regulates osteoblasts proliferation through Ca2+-CaMK-CREB signal pathway via Ca2+ channel and PLC/IP3 transduction cascades.
Collapse
Affiliation(s)
- Yong Guo
- Depantment of Bioengineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, China; Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, China
| | - Qi Lv
- Experiment Management Center, Logistical College of People Armed Police Forces, Tianjin 300162, China
| | - Xian-Qiong Zou
- Depantment of Bioengineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, China
| | - Zhi-Xiong Yan
- Depantment of Bioengineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, China
| | - Yu-Xian Yan
- Depantment of Bioengineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, China; Experiment Management Center, Logistical College of People Armed Police Forces, Tianjin 300162, China
| |
Collapse
|
39
|
Expression of voltage-gated calcium channels augments cell susceptibility to membrane disruption by nanosecond pulsed electric field. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:2175-2183. [PMID: 30409513 DOI: 10.1016/j.bbamem.2018.08.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022]
Abstract
We compared membrane permeabilization by nanosecond pulsed electric field (nsPEF) in HEK293 cells with and without assembled CaV1.3 L-type voltage-gated calcium channel (VGCC). Individual cells were subjected to one 300-ns pulse at 0 (sham exposure); 1.4; 1.8; or 2.3 kV/cm, and membrane permeabilization was evaluated by measuring whole-cell currents and by optical monitoring of cytosolic Ca2+. nsPEF had either no effect (0 and 1.4 kV/cm), or caused a lasting (>80 s) increase in the membrane conductance in about 50% of cells (1.8 kV/cm), or in all cells (2.3 kV/cm). The conductance pathway opened by nsPEF showed strong inward rectification, with maximum conductance increase for the inward current at the most negative membrane potentials. Although these potentials were below the depolarization threshold for VGCC activation, the increase in conductance in cells which expressed VGCC (VGCC+ cells) was about twofold greater than in cells which did not (VGCC- cells). Among VGCC+ cells, the nsPEF-induced increase in membrane conductance showed a positive correlation with the amplitude of VGCC current measured in the same cells prior to nsPEF exposure. These findings demonstrate that the expression of VGCC makes cells more susceptible to membrane permeabilization by nsPEF. Time-lapse imaging of nsPEF-induced Ca2+ transients confirmed permeabilization by a single 300-ns pulse at 1.8 or 2.3 kV/cm, but not at 1.4 kV/cm, and the transients were expectedly larger in VGCC+ cells. However, it remains to be established whether larger transients reflected additional Ca2+ entry through VGCC, or were a result of more severe electropermeabilization of VGCC+ cells.
Collapse
|
40
|
Semenov I, Grigoryev S, Neuber JU, Zemlin CW, Pakhomova ON, Casciola M, Pakhomov AG. Excitation and injury of adult ventricular cardiomyocytes by nano- to millisecond electric shocks. Sci Rep 2018; 8:8233. [PMID: 29844431 PMCID: PMC5974370 DOI: 10.1038/s41598-018-26521-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/08/2018] [Indexed: 12/13/2022] Open
Abstract
Intense electric shocks of nanosecond (ns) duration can become a new modality for more efficient but safer defibrillation. We extended strength-duration curves for excitation of cardiomyocytes down to 200 ns, and compared electroporative damage by proportionally more intense shocks of different duration. Enzymatically isolated murine, rabbit, and swine adult ventricular cardiomyocytes (VCM) were loaded with a Ca2+ indicator Fluo-4 or Fluo-5N and subjected to shocks of increasing amplitude until a Ca2+ transient was optically detected. Then, the voltage was increased 5-fold, and the electric cell injury was quantified by the uptake of a membrane permeability marker dye, propidium iodide. We established that: (1) Stimuli down to 200-ns duration can elicit Ca2+ transients, although repeated ns shocks often evoke abnormal responses, (2) Stimulation thresholds expectedly increase as the shock duration decreases, similarly for VCMs from different species, (3) Stimulation threshold energy is minimal for the shortest shocks, (4) VCM orientation with respect to the electric field does not affect the threshold for ns shocks, and (5) The shortest shocks cause the least electroporation injury. These findings support further exploration of ns defibrillation, although abnormal response patterns to repetitive ns stimuli are of a concern and require mechanistic analysis.
Collapse
Affiliation(s)
- Iurii Semenov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Sergey Grigoryev
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Johanna U Neuber
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA.,Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, 23508, USA
| | - Christian W Zemlin
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA.,Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, 23508, USA
| | - Olga N Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Maura Casciola
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Andrei G Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, 23508, USA.
| |
Collapse
|
41
|
Batista Napotnik T, Miklavčič D. In vitro electroporation detection methods – An overview. Bioelectrochemistry 2018; 120:166-182. [DOI: 10.1016/j.bioelechem.2017.12.005] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022]
|
42
|
Different Membrane Pathways Mediate Ca 2+ Influx in Adrenal Chromaffin Cells Exposed to 150-400 ns Electric Pulses. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9046891. [PMID: 29789806 PMCID: PMC5896273 DOI: 10.1155/2018/9046891] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/18/2018] [Indexed: 12/21/2022]
Abstract
Exposing adrenal chromaffin cells to 5 ns electric pulses (nsPEF) causes a rapid rise in intracellular Ca2+ ([Ca2+]i) that is solely the result of Ca2+ influx through voltage-gated Ca2+ channels (VGCCs). This study explored the effect of longer duration nsPEF on [Ca2+]i. Single 150, 200, or 400 ns pulses at 3.1 kV/cm evoked rapid increases in [Ca2+]i, the magnitude of which increased linearly with pulse width and electric field amplitude. Recovery of [Ca2+]i to prestimulus levels was faster for 150 ns exposures. Regardless of pulse width, no rise in [Ca2+]i occurred in the absence of extracellular Ca2+, indicating that the source of Ca2+ was from outside the cell. Ca2+ responses evoked by a 150 ns pulse were inhibited to varying degrees by ω-agatoxin IVA, ω-conotoxin GVIA, nitrendipine or nimodipine, antagonists of P/Q-, N-, and L-type VGCCs, respectively, and by 67% when all four types of VGCCs were blocked simultaneously. The remaining Ca2+ influx insensitive to VGCC inhibitors was attributed to plasma membrane nanoporation, which comprised the E-field sensitive component of the response. Both pathways of Ca2+ entry were inhibited by 200 μM Cd2+. These results demonstrate that, in excitable chromaffin cells, single 150-400 ns pulses increased the permeability of the plasma membrane to Ca2+ in addition to causing Ca2+ influx via VGCCs.
Collapse
|
43
|
Dermol-Černe J, Miklavčič D, Reberšek M, Mekuč P, Bardet SM, Burke R, Arnaud-Cormos D, Leveque P, O'Connor R. Plasma membrane depolarization and permeabilization due to electric pulses in cell lines of different excitability. Bioelectrochemistry 2018; 122:103-114. [PMID: 29621662 DOI: 10.1016/j.bioelechem.2018.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/13/2018] [Accepted: 03/19/2018] [Indexed: 12/18/2022]
Abstract
In electroporation-based medical treatments, excitable tissues are treated, either intentionally (irreversible electroporation of brain cancer, gene electrotransfer or ablation of the heart muscle, gene electrotransfer of skeletal muscles), or unintentionally (excitable tissues near the target area). We investigated how excitable and non-excitable cells respond to electric pulses, and if electroporation could be an effective treatment of the tumours of the central nervous system. For three non-excitable and one excitable cell line, we determined a strength-duration curve for a single pulse of 10ns-10ms. The threshold for depolarization decreased with longer pulses and was higher for excitable cells. We modelled the response with the Lapicque curve and the Hodgkin-Huxley model. At 1μs a plateau of excitability was reached which could explain why high-frequency irreversible electroporation (H-FIRE) electroporates but does not excite cells. We exposed cells to standard electrochemotherapy parameters (8×100μs pulses, 1Hz, different voltages). Cells behaved similarly which indicates that electroporation most probably occurs at the level of lipid bilayer, independently of the voltage-gated channels. These results could be used for optimization of electric pulses to achieve maximal permeabilization and minimal excitation/pain sensation. In the future, it should be established whether the in vitro depolarization correlates to nerve/muscle stimulation and pain sensation in vivo.
Collapse
Affiliation(s)
- Janja Dermol-Černe
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, SI-1000 Ljubljana, Slovenia.
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, SI-1000 Ljubljana, Slovenia.
| | - Matej Reberšek
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, SI-1000 Ljubljana, Slovenia.
| | - Primož Mekuč
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, SI-1000 Ljubljana, Slovenia
| | - Sylvia M Bardet
- University of Limoges, CNRS, XLIM, UMR 7252, F-87000 Limoges, France.
| | - Ryan Burke
- University of Limoges, CNRS, XLIM, UMR 7252, F-87000 Limoges, France
| | | | - Philippe Leveque
- University of Limoges, CNRS, XLIM, UMR 7252, F-87000 Limoges, France.
| | - Rodney O'Connor
- École des Mines de Saint-Étienne, Department of Bioelectronics, Georges Charpak Campus, Centre Microélectronique de Provence, 880 Route de Mimet, 13120 Gardanne, France.
| |
Collapse
|
44
|
Muratori C, Pakhomov AG, Gianulis E, Meads J, Casciola M, Mollica PA, Pakhomova ON. Activation of the phospholipid scramblase TMEM16F by nanosecond pulsed electric fields (nsPEF) facilitates its diverse cytophysiological effects. J Biol Chem 2017; 292:19381-19391. [PMID: 28982976 PMCID: PMC5702676 DOI: 10.1074/jbc.m117.803049] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/20/2017] [Indexed: 12/17/2022] Open
Abstract
Nanosecond pulsed electric fields (nsPEF) are emerging as a novel modality for cell stimulation and tissue ablation. However, the downstream protein effectors responsible for nsPEF bioeffects remain to be established. Here we demonstrate that nsPEF activate TMEM16F (or Anoctamin 6), a protein functioning as a Ca2+-dependent phospholipid scramblase and Ca2+-activated chloride channel. Using confocal microscopy and patch clamp recordings, we investigated the relevance of TMEM16F activation for several bioeffects triggered by nsPEF, including phosphatidylserine (PS) externalization, nanopore-conducted currents, membrane blebbing, and cell death. In HEK 293 cells treated with a single 300-ns pulse of 25.5 kV/cm, Tmem16f expression knockdown and TMEM16F-specific inhibition decreased nsPEF-induced PS exposure by 49 and 42%, respectively. Moreover, the Tmem16f silencing significantly decreased Ca2+-dependent chloride channel currents activated in response to the nanoporation. Tmem16f expression also affected nsPEF-induced cell blebbing, with only 20% of the silenced cells developing blebs compared with 53% of the control cells. This inhibition of cellular blebbing correlated with a 25% decrease in cytosolic free Ca2+ transient at 30 s after nanoporation. Finally, in TMEM16F-overexpressing cells, a train of 120 pulses (300 ns, 20 Hz, 6 kV/cm) decreased cell survival to 34% compared with 51% in control cells (*, p < 0.01). Taken together, these results indicate that TMEM16F activation by nanoporation mediates and enhances the diverse cellular effects of nsPEF.
Collapse
Affiliation(s)
| | | | - Elena Gianulis
- From the Frank Reidy Research Center for Bioelectrics, and
| | - Jade Meads
- From the Frank Reidy Research Center for Bioelectrics, and
| | - Maura Casciola
- From the Frank Reidy Research Center for Bioelectrics, and
| | - Peter A Mollica
- the Department of Medical Diagnostics and Translational Sciences, Old Dominion University, Norfolk, Virginia 23508
| | | |
Collapse
|
45
|
Hanna H, Denzi A, Liberti M, André FM, Mir LM. Electropermeabilization of Inner and Outer Cell Membranes with Microsecond Pulsed Electric Fields: Quantitative Study with Calcium Ions. Sci Rep 2017; 7:13079. [PMID: 29026094 PMCID: PMC5638809 DOI: 10.1038/s41598-017-12960-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/15/2017] [Indexed: 12/18/2022] Open
Abstract
Microsecond pulsed electric fields (μsPEF) permeabilize the plasma membrane (PM) and are widely used in research, medicine and biotechnology. For internal membranes permeabilization, nanosecond pulsed electric fields (nsPEF) are applied but this technology is complex to use. Here we report that the endoplasmic reticulum (ER) membrane can also be electropermeabilized by one 100 µs pulse without affecting the cell viability. Indeed, using Ca2+ as a permeabilization marker, we observed cytosolic Ca2+ peaks in two different cell types after one 100 µs pulse in a medium without Ca2+. Thapsigargin abolished these Ca2+ peaks demonstrating that the calcium is released from the ER. Moreover, IP3R and RyR inhibitors did not modify these peaks showing that they are due to the electropermeabilization of the ER membrane and not to ER Ca2+ channels activation. Finally, the comparison of the two cell types suggests that the PM and the ER permeabilization thresholds are affected by the sizes of the cell and the ER. In conclusion, this study demonstrates that µsPEF, which are easier to control than nsPEF, can permeabilize internal membranes. Besides, μsPEF interaction with either the PM or ER, can be an efficient tool to modulate the cytosolic calcium concentration and study Ca2+ roles in cell physiology.
Collapse
Affiliation(s)
- Hanna Hanna
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94 805, Villejuif, France
| | - Agnese Denzi
- Department of Information Engineering, Electronics and Telecommunication (DIET), University of Rome "La Sapienza", Rome, 00184, Italy
| | - Micaela Liberti
- Department of Information Engineering, Electronics and Telecommunication (DIET), University of Rome "La Sapienza", Rome, 00184, Italy
| | - Franck M André
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94 805, Villejuif, France
| | - Lluis M Mir
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94 805, Villejuif, France.
| |
Collapse
|
46
|
Burke RC, Bardet SM, Carr L, Romanenko S, Arnaud-Cormos D, Leveque P, O'Connor RP. Nanosecond pulsed electric fields depolarize transmembrane potential via voltage-gated K+, Ca2+ and TRPM8 channels in U87 glioblastoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2040-2050. [DOI: 10.1016/j.bbamem.2017.07.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022]
|
47
|
Benign Effect of Extremely Low-Frequency Electromagnetic Field on Brain Plasticity Assessed by Nitric Oxide Metabolism during Poststroke Rehabilitation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2181942. [PMID: 29138675 PMCID: PMC5613626 DOI: 10.1155/2017/2181942] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/02/2017] [Accepted: 08/14/2017] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is one of the most important signal molecules, involved in both physiological and pathological processes. As a neurotransmitter in the central nervous system, NO regulates cerebral blood flow, neurogenesis, and synaptic plasticity. The aim of our study was to investigate the effect of the extremely low-frequency electromagnetic field (ELF-EMF) on generation and metabolism of NO, as a neurotransmitter, in the rehabilitation of poststroke patients. Forty-eight patients were divided into two groups: ELF-EMF and non-ELF-EMF. Both groups underwent the same 4-week rehabilitation program. Additionally, the ELF-EMF group was exposed to an extremely low-frequency electromagnetic field of 40 Hz, 7 mT, for 15 min/day. Levels of 3-nitrotyrosine, nitrate/nitrite, and TNFα in plasma samples were measured, and NOS2 expression was determined in whole blood samples. Functional status was evaluated before and after a series of treatments, using the Activity Daily Living, Geriatric Depression Scale, and Mini-Mental State Examination. We observed that application of ELF-EMF significantly increased 3-nitrotyrosine and nitrate/nitrite levels, while expression of NOS2 was insignificantly decreased in both groups. The results also show that ELF-EMF treatments improved functional and mental status. We conclude that ELF-EMF therapy is capable of promoting recovery in poststroke patients.
Collapse
|
48
|
Damage-free peripheral nerve stimulation by 12-ns pulsed electric field. Sci Rep 2017; 7:10453. [PMID: 28874684 PMCID: PMC5585227 DOI: 10.1038/s41598-017-10282-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/07/2017] [Indexed: 12/15/2022] Open
Abstract
Modern technologies enable deep tissue focusing of nanosecond pulsed electric field (nsPEF) for non-invasive nerve and muscle stimulation. However, it is not known if PEF orders of magnitude shorter than the activation time of voltage-gated sodium channels (VGSC) would evoke action potentials (APs). One plausible scenario requires the loss of membrane integrity (electroporation) and resulting depolarization as an intermediate step. We report, for the first time, that the excitation of a peripheral nerve can be accomplished by 12-ns PEF without electroporation. 12-ns stimuli at 4.1-11 kV (3.3-8.8 kV/cm) evoked APs similarly to conventional stimuli (100-250 μs, 1-5 V, 103-515 V/m), except for having higher selectivity for the faster nerve fibers. Nerves sustained repeated tetanic stimulations (50 Hz or 100 Hz for 1 min) alternately by 12-ns PEF and by conventional pulses. Such tetani caused a modest AP decrease, to a similar extent for both types of stimuli. Nerve refractory properties were not affected. The lack of cumulative damages even from tens of thousands of 12-ns stimuli and the similarities with the conventional stimulation prove VGSC activation by nsPEF without nerve membrane damage.
Collapse
|
49
|
Doyon P, Johansson O. Electromagnetic fields may act via calcineurin inhibition to suppress immunity, thereby increasing risk for opportunistic infection: Conceivable mechanisms of action. Med Hypotheses 2017; 106:71-87. [PMID: 28818275 DOI: 10.1016/j.mehy.2017.06.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/25/2017] [Accepted: 06/30/2017] [Indexed: 11/26/2022]
|
50
|
Gianulis EC, Casciola M, Xiao S, Pakhomova ON, Pakhomov AG. Electropermeabilization by uni- or bipolar nanosecond electric pulses: The impact of extracellular conductivity. Bioelectrochemistry 2017; 119:10-19. [PMID: 28865240 DOI: 10.1016/j.bioelechem.2017.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/05/2017] [Accepted: 08/15/2017] [Indexed: 02/03/2023]
Abstract
Cellular effects caused by nanosecond electric pulses (nsEP) can be reduced by an electric field reversal, a phenomenon known as bipolar cancellation. The reason for this cancellation effect remains unknown. We hypothesized that assisted membrane discharge is the mechanism for bipolar cancellation. CHO-K1 cells bathed in high (16.1mS/cm; HCS) or low (1.8mS/cm; LCS) conductivity solutions were exposed to either one unipolar (300-ns) or two opposite polarity (300+300-ns; bipolar) nsEP (4-40kV/cm) with increasing interpulse intervals (0.1-50μs). Time-lapse YO-PRO-1 (YP) uptake revealed enhanced membrane permeabilization in LCS compared to HCS at all tested voltages. The time-dependence of bipolar cancellation was similar in both solutions, using either identical (22kV/cm) or isoeffective nsEP treatments (12 and 32kV/cm for LCS and HCS, respectively). However, cancellation was significantly stronger in LCS when the bipolar nsEP had no, or very short (<1μs), interpulse intervals. Finally, bipolar cancellation was still present with interpulse intervals as long as 50μs, beyond the time expected for membrane discharge. Our findings do not support assisted membrane discharge as the mechanism for bipolar cancellation. Instead they exemplify the sustained action of nsEP that can be reversed long after the initial stimulus.
Collapse
Affiliation(s)
- Elena C Gianulis
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA.
| | - Maura Casciola
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Shu Xiao
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA 23508, USA
| | - Olga N Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Andrei G Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| |
Collapse
|