1
|
Sowa PW, Novickij V, Kiełbik A, Kollotzek F, Heinzmann D, Borst O, Gawaz MP. Fractionation of nanosecond pulsed electric fields lowers lethal dose by enhancing cardiomyocyte membrane permeability. Heart Rhythm 2025:S1547-5271(25)02190-3. [PMID: 40107396 DOI: 10.1016/j.hrthm.2025.03.1954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Nanosecond pulsed electric fields (nsPEFs) are a promising method for cardiac pulsed field ablation, currently in early clinical trials. However, effective ablation often requires high voltages, more pulses, and higher frequencies, which can raise tissue temperatures because of Joule heating. Fractionated pulse delivery can help mitigate thermal effects and potentially evoke electrosensitization, increasing cell damage. OBJECTIVE This study evaluates the effects of fractionated nsPEF on treatment efficacy and its selectivity against cardiomyocytes, aiming to determine whether fractionation improves ablation outcomes. METHODS Monolayers of HL-1 murine cardiomyocytes, MHEC5-T murine endothelial cells, AC16 human cardiomyocytes, and human umbilical vein endothelial cells were exposed to pulsed electric fields using a contact electrode operated by a custom robotic system. Cell viability and permeability were measured using wide-field fluorescence microscopy. Stained areas were matched to simulated electric fields for dose-response curves. Fractionation effects were also validated in an ex vivo murine model. RESULTS Fractionation of nsPEF reduced the electric field affecting 50% of cells for plasma membrane permeabilization by 10% compared with a single train of 200 pulses (P < .0001). This translated into enhanced cardiomyocyte ablation, with fractionated exposure lowering the electric field affecting 50% of cells for cell killing by 13% (P < .0001). Ex vivo results further confirmed a larger ablation area with fractionated nsPEF (P < .0001). CONCLUSION Fractionated nsPEF improves cardiac ablation efficiency by enhancing membrane permeability and cell-killing effect. These findings suggest that fractionated delivery could optimize nsPEF therapies, offering a more effective approach for cardiac ablation.
Collapse
Affiliation(s)
- Pamela W Sowa
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany.
| | - Vitalij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius, Lithuania; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | | | - Ferdinand Kollotzek
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany; DFG Heisenberg Group Cardiovascular Thrombo-Inflammation and Translational Thrombocardiology, University of Tübingen, Tübingen, Germany
| | - David Heinzmann
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany; DFG Heisenberg Group Cardiovascular Thrombo-Inflammation and Translational Thrombocardiology, University of Tübingen, Tübingen, Germany
| | - Meinrad P Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
2
|
Jacobs EJ, Rubinsky B, Davalos RV. Pulsed field ablation in medicine: irreversible electroporation and electropermeabilization theory and applications. Radiol Oncol 2025; 59:1-22. [PMID: 40014783 PMCID: PMC11867574 DOI: 10.2478/raon-2025-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/07/2024] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Focal ablation techniques are integral in the surgical intervention of diseased tissue, where it is necessary to minimize damage to the surrounding parenchyma and critical structures. Irreversible electroporation (IRE) and high-frequency IRE (H-FIRE), colloquially called pulsed-field ablation (PFA), utilize high-amplitude, low-energy pulsed electric fields (PEFs) to nonthermally ablate soft tissue. PEFs induce cell death through permeabilization of the cellular membrane, leading to loss of homeostasis. The unique nonthermal nature of PFA allows for selective cell death while minimally affecting surrounding proteinaceous structures, permitting treatment near sensitive anatomy where thermal ablation or surgical resection is contraindicated. Further, PFA is being used to treat tissue when tumor margins are not expected after surgical resection, termed margin accentuation. This review explores both the theoretical foundations of PFA, detailing how PEFs induce cell membrane destabilization and selective tissue ablation, the outcomes following treatment, and its clinical implications across oncology and cardiology. CONCLUSIONS Clinical experience is still progressing, but reports have demonstrated that PFA reduces complications often seen with thermal ablation techniques. Mounting oncology data also support that PFA produces a robust immune response that may prevent local recurrences and attenuate metastatic disease. Despite promising outcomes, challenges such as optimizing field delivery and addressing variations in tissue response require further investigation. Future directions include refining PFA protocols and expanding its application to other therapeutic areas like benign tissue hyperplasia and chronic bronchitis.
Collapse
Affiliation(s)
- Edward J Jacobs
- Wallace H Coulter School of Biomedical Engineering, Georgia Institute of Technology & Emory Medical School, Atlanta, Georgia, USA
| | - Boris Rubinsky
- Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, California, USA
| | - Rafael V Davalos
- Wallace H Coulter School of Biomedical Engineering, Georgia Institute of Technology & Emory Medical School, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Ramirez DA, Garrott K, Garlitski A, Koop B. Coronary Spasm Due to Pulsed Field Ablation: A State-of-the-Art Review. Pacing Clin Electrophysiol 2024. [PMID: 39494719 DOI: 10.1111/pace.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
With the ever-growing population of patients undergoing cardiac ablation with pulsed electric fields, there is a need to understand secondary effects from the therapy. Coronary artery spasm is one such effect that has recently emerged as the subject of further investigation in electrophysiology literature. This review aims to elucidate the basic anatomy underlying vascular spasm due to pulsed electric fields and the effects of irreversible electroporation on coronary arteries. This review also aims to gather the current preclinical and clinical data regarding the physiology and function of coronary arteries following electroporation.
Collapse
Affiliation(s)
- David A Ramirez
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| | - Kara Garrott
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| | - Ann Garlitski
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| | - Brendan Koop
- Electrophysiology Research & Development, Boston Scientific Corporation, Marlborough, Massachusetts, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Silkunas M, Pakhomova ON, Silkuniene G, Pakhomov AG. Dynamics of cell membrane lesions and adaptive conductance under the electrical stress. Cell Stress 2024; 8:69-82. [PMID: 39135750 PMCID: PMC11318148 DOI: 10.15698/cst2024.08.298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
Exceeding physiological limits of the cell membrane potential compromises structural integrity, enabling the passage of normally impermeant solutes and disrupting cell function. Electropermeabilization has been studied extensively at the cellular scale, but not at the individual membrane lesion level. We employed fast total internal reflection fluorescence (TIRF) imaging of Ca2+ entry transients to discern individual lesions in a hyperpolarized cell membrane and characterize their focality, thresholds, electrical conductance, and the lifecycle. A diffuse and momentary membrane permeabilization without a distinct pore formation was observed already at a -100 mV threshold. Polarizing down to -200 mV created focal pores with a low 50- to 300-pS conductance, which disappeared instantly once the hyperpolarization was removed. Charging to -240 mV created high-conductance (> 1 nS) pores which persisted for seconds even at zero membrane potential. With incremental hyperpolarization steps, persistent pores often emerged at locations different from those where the short-lived, low-conductance pores or diffuse permeabilization were previously observed. Attempts to polarize membrane beyond the threshold for the formation of persistent pores increased their conductance adaptively, preventing further potential build-up and "clamping" it at a certain limit (-270 ± 6 mV in HEK cells, -284 ± 5 mV in CHO cells, and -243 ± 9 mV in neurons). The data suggest a previously unknown role of electroporative lesions as a protective mechanism against a potentially fatal membrane overcharging and cell disintegration.
Collapse
Affiliation(s)
- Mantas Silkunas
- Frank Reidy Research Center for Bioelectrics, Old Dominion UniversityVA, NorfolkUSA
| | - Olga N Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion UniversityVA, NorfolkUSA
| | - Giedre Silkuniene
- Frank Reidy Research Center for Bioelectrics, Old Dominion UniversityVA, NorfolkUSA
| | - Andrei G. Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion UniversityVA, NorfolkUSA
| |
Collapse
|
7
|
Urbanskas E, Jakštys B, Venckus J, Malakauskaitė P, Šatkauskienė I, Morkvėnaitė-Vilkončienė I, Šatkauskas S. Interplay between Electric Field Strength and Number of Short-Duration Pulses for Efficient Gene Electrotransfer. Pharmaceuticals (Basel) 2024; 17:825. [PMID: 39065676 PMCID: PMC11279932 DOI: 10.3390/ph17070825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/08/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Electroporation is a method that shows great promise as a non-viral approach for delivering genes by using high-voltage electric pulses to introduce DNA into cells to induce transient gene expression. This research aimed to evaluate the interplay between electric pulse intensity and 100 µs-duration pulse numbers as an outcome of gene electrotransfer efficacy and cell viability. Our results indicated a close relationship between pulse number and electric field strength regarding gene electrotransfer efficacy; higher electric pulse intensity resulted in fewer pulses needed to achieve the same gene electrotransfer efficacy. Subsequently, an increase in pulse number had a more negative impact on overall gene electrotransfer by significantly reducing cell viability. Based on our data, the best pulse parameters to transfect CHO cells with the pMax-GFP plasmid were using 5 HV square wave pulses of 1000 V/cm and 2 HV of 1600 V/cm, correspondingly resulting in 55 and 71% of transfected cells and maintaining 79 and 54% proliferating cells. This shows ESOPE-like 100 µs-duration pulse protocols can be used simultaneously to deliver cytotoxic drugs as well as immune response regulating genetically encoded cytokines.
Collapse
Affiliation(s)
- Ernestas Urbanskas
- Research Institute of Natural and Technological Sciences, Vytautas Magnus University, 44404 Kaunas, Lithuania; (E.U.); (B.J.); (J.V.); (I.Š.)
| | - Baltramiejus Jakštys
- Research Institute of Natural and Technological Sciences, Vytautas Magnus University, 44404 Kaunas, Lithuania; (E.U.); (B.J.); (J.V.); (I.Š.)
- Faculty of Electronics, Vilnius Gediminas Technical University, 10105 Vilnius, Lithuania;
| | - Justinas Venckus
- Research Institute of Natural and Technological Sciences, Vytautas Magnus University, 44404 Kaunas, Lithuania; (E.U.); (B.J.); (J.V.); (I.Š.)
| | - Paulina Malakauskaitė
- Faculty of Electronics, Vilnius Gediminas Technical University, 10105 Vilnius, Lithuania;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Ingrida Šatkauskienė
- Research Institute of Natural and Technological Sciences, Vytautas Magnus University, 44404 Kaunas, Lithuania; (E.U.); (B.J.); (J.V.); (I.Š.)
| | - Inga Morkvėnaitė-Vilkončienė
- Department of Nanotechnology, State Research Institute Centre for Physical Sciences and Technology, 02300 Vilnius, Lithuania;
| | - Saulius Šatkauskas
- Research Institute of Natural and Technological Sciences, Vytautas Magnus University, 44404 Kaunas, Lithuania; (E.U.); (B.J.); (J.V.); (I.Š.)
| |
Collapse
|
8
|
Lisec B, Bozic T, Santek I, Markelc B, Vrecl M, Frangez R, Cemazar M. Characterization of two distinct immortalized endothelial cell lines, EA.hy926 and HMEC-1, for in vitro studies: exploring the impact of calcium electroporation, Ca 2+ signaling and transcriptomic profiles. Cell Commun Signal 2024; 22:118. [PMID: 38347539 PMCID: PMC10863159 DOI: 10.1186/s12964-024-01503-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Disruption of Ca2+ homeostasis after calcium electroporation (CaEP) in tumors has been shown to elicit an enhanced antitumor effect with varying impacts on healthy tissue, such as endothelium. Therefore, our study aimed to determine differences in Ca2+ kinetics and gene expression involved in the regulation of Ca2+ signaling and homeostasis, as well as effects of CaEP on cytoskeleton and adherens junctions of the established endothelial cell lines EA.hy926 and HMEC-1. METHODS CaEP was performed on EA.hy926 and HMEC-1 cells with increasing Ca2+ concentrations. Viability after CaEP was assessed using Presto Blue, while the effect on cytoskeleton and adherens junctions was evaluated via immunofluorescence staining (F-actin, α-tubulin, VE-cadherin). Differences in intracellular Ca2+ regulation ([Ca2+]i) were determined with spectrofluorometric measurements using Fura-2-AM, exposing cells to DPBS, ionomycin, thapsigargin, ATP, bradykinin, angiotensin II, acetylcholine, LaCl3, and GdCl3. Molecular distinctions were identified by analyzing differentially expressed genes and pathways related to the cytoskeleton and Ca2+ signaling through RNA sequencing. RESULTS EA.hy926 cells, at increasing Ca2+ concentrations, displayed higher CaEP susceptibility and lower survival than HMEC-1. Immunofluorescence confirmed CaEP-induced, time- and Ca2+-dependent morphological changes in EA.hy926's actin filaments, microtubules, and cell-cell junctions. Spectrofluorometric Ca2+ kinetics showed higher amplitudes in Ca2+ responses in EA.hy926 exposed to buffer, G protein coupled receptor agonists, bradykinin, and angiotensin II compared to HMEC-1. HMEC-1 exhibited significantly higher [Ca2+]i changes after ionomycin exposure, while responses to thapsigargin, ATP, and acetylcholine were similar in both cell lines. ATP without extracellular Ca2+ ions induced a significantly higher [Ca2+]i rise in EA.hy926, suggesting purinergic ionotropic P2X and metabotropic P2Y receptor activation. RNA-sequencing analysis showed significant differences in cytoskeleton- and Ca2+-related gene expression, highlighting upregulation of ORAI2, TRPC1, TRPM2, CNGA3, TRPM6, and downregulation of TRPV4 and TRPC4 in EA.hy926 versus HMEC-1. Moreover, KEGG analysis showed upregulated Ca2+ import and downregulated export genes in EA.hy926. CONCLUSIONS Our finding show that significant differences in CaEP response and [Ca2+]i regulation exist between EA.hy926 and HMEC-1, which may be attributed to distinct transcriptomic profiles. EA.hy926, compared to HMEC-1, displayed higher susceptibility and sensitivity to [Ca2+]i changes, which may be linked to overexpression of Ca2+-related genes and an inability to mitigate changes in [Ca2+]i. The study offers a bioinformatic basis for selecting EC models based on research objectives.
Collapse
Affiliation(s)
- Barbara Lisec
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000, Ljubljana, Slovenia
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000, Ljubljana, Slovenia
| | - Iva Santek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000, Ljubljana, Slovenia
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbiceva 60, SI-1000, Ljubljana, Slovenia
| | - Robert Frangez
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbiceva 60, SI-1000, Ljubljana, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000, Ljubljana, Slovenia.
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310, Izola, Slovenia.
| |
Collapse
|
9
|
Mazzarda F, Chittams-Miles AE, Pittaluga J, Sözer EB, Vernier PT, Muratori C. Inflammasome Activation and IL-1β Release Triggered by Nanosecond Pulsed Electric Fields in Murine Innate Immune Cells and Skin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:335-345. [PMID: 38047899 PMCID: PMC10752860 DOI: 10.4049/jimmunol.2200881] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 11/08/2023] [Indexed: 12/05/2023]
Abstract
Although electric field-induced cell membrane permeabilization (electroporation) is used in a wide range of clinical applications from cancer therapy to cardiac ablation, the cellular- and molecular-level details of the processes that determine the success or failure of these treatments are poorly understood. Nanosecond pulsed electric field (nsPEF)-based tumor therapies are known to have an immune component, but whether and how immune cells sense the electroporative damage and respond to it have not been demonstrated. Damage- and pathogen-associated stresses drive inflammation via activation of cytosolic multiprotein platforms known as inflammasomes. The assembly of inflammasome complexes triggers caspase-1-dependent secretion of IL-1β and in many settings a form of cell death called pyroptosis. In this study we tested the hypothesis that the nsPEF damage is sensed intracellularly by the NLRP3 inflammasome. We found that 200-ns PEFs induced aggregation of the inflammasome adaptor protein ASC, activation of caspase-1, and triggered IL-1β release in multiple innate immune cell types (J774A.1 macrophages, bone marrow-derived macrophages, and dendritic cells) and in vivo in mouse skin. Efflux of potassium from the permeabilized cell plasma membrane was partially responsible for nsPEF-induced inflammasome activation. Based on results from experiments using both the NRLP3-specific inhibitor MCC950 and NLRP3 knockout cells, we propose that the damage created by nsPEFs generates a set of stimuli for the inflammasome and that more than one sensor can drive IL-1β release in response to electrical pulse stimulation. This study shows, to our knowledge, for the first time, that PEFs activate the inflammasome, suggesting that this pathway alarms the immune system after treatment.
Collapse
Affiliation(s)
- Flavia Mazzarda
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA
| | | | - Julia Pittaluga
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA
| | - Esin B. Sözer
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA
| | - P. Thomas Vernier
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA
| | - Claudia Muratori
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA
| |
Collapse
|
10
|
Tsimpaki T, Anastasova R, Liu H, Seitz B, Bechrakis NE, Berchner-Pfannschmidt U, Kraemer MM, Fiorentzis M. Calcium Electroporation versus Electrochemotherapy with Bleomycin in an In Vivo CAM-Based Uveal Melanoma Xenograft Model. Int J Mol Sci 2024; 25:938. [PMID: 38256012 PMCID: PMC10815639 DOI: 10.3390/ijms25020938] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Despite recent advancements in the diagnosis and treatment of uveal melanoma (UM), its metastatic rate remains high and is accompanied by a highly dismal prognosis, constituting an unmet need for the development of novel adjuvant therapeutic strategies. We established an in vivo chick chorioallantoic membrane (CAM)-based UM xenograft model from UPMD2 and UPMM3 cell lines to examine its feasibility for the improvement of selection of drug candidates. The efficacy of calcium electroporation (CaEP) with 5 or 10 mM calcium chloride (Ca) and electrochemotherapy (ECT) with 1 or 2.5 µg/mL bleomycin in comparison to monotherapy with the tested drug or electroporation (EP) alone was investigated on the generated UM tumors. CaEP and ECT showed a similar reduction of proliferation and melanocytic expansion with a dose-dependent effect for bleomycin, whereas CaEP induced a significant increase of the apoptosis and a reduction of vascularization with varying sensitivity for the two xenograft types. Our in vivo results suggest that CaEP and ECT may facilitate the adequate local tumor control and contribute to the preservation of the bulbus, potentially opening new horizons in the adjuvant treatment of advanced UM.
Collapse
Affiliation(s)
- Theodora Tsimpaki
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (T.T.); (R.A.); (H.L.); (N.E.B.); (U.B.-P.); (M.M.K.)
| | - Ralitsa Anastasova
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (T.T.); (R.A.); (H.L.); (N.E.B.); (U.B.-P.); (M.M.K.)
| | - Hongtao Liu
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (T.T.); (R.A.); (H.L.); (N.E.B.); (U.B.-P.); (M.M.K.)
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Kirrberger Str. 100, 66421 Homburg, Germany;
| | - Nikolaos E. Bechrakis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (T.T.); (R.A.); (H.L.); (N.E.B.); (U.B.-P.); (M.M.K.)
| | - Utta Berchner-Pfannschmidt
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (T.T.); (R.A.); (H.L.); (N.E.B.); (U.B.-P.); (M.M.K.)
| | - Miriam M. Kraemer
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (T.T.); (R.A.); (H.L.); (N.E.B.); (U.B.-P.); (M.M.K.)
| | - Miltiadis Fiorentzis
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Hufeland Str. 55, 45147 Essen, Germany; (T.T.); (R.A.); (H.L.); (N.E.B.); (U.B.-P.); (M.M.K.)
| |
Collapse
|
11
|
Orlacchio R, Kolosnjaj-Tabi J, Mattei N, Lévêque P, Rols MP, Arnaud-Cormos D, Golzio M. Effects of Nanosecond Pulsed Electric Field (nsPEF) on a Multicellular Spheroid Tumor Model: Influence of Pulse Duration, Pulse Repetition Rate, Absorbed Energy, and Temperature. Int J Mol Sci 2023; 24:14999. [PMID: 37834447 PMCID: PMC10573334 DOI: 10.3390/ijms241914999] [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: 09/04/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Cellular response upon nsPEF exposure depends on different parameters, such as pulse number and duration, the intensity of the electric field, pulse repetition rate (PRR), pulsing buffer composition, absorbed energy, and local temperature increase. Therefore, a deep insight into the impact of such parameters on cellular response is paramount to adaptively optimize nsPEF treatment. Herein, we examined the effects of nsPEF ≤ 10 ns on long-term cellular viability and growth as a function of pulse duration (2-10 ns), PRR (20 and 200 Hz), cumulative time duration (1-5 µs), and absorbed electrical energy density (up to 81 mJ/mm3 in sucrose-containing low-conductivity buffer and up to 700 mJ/mm3 in high-conductivity HBSS buffer). Our results show that the effectiveness of nsPEFs in ablating 3D-grown cancer cells depends on the medium to which the cells are exposed and the PRR. When a medium with low-conductivity is used, the pulses do not result in cell ablation. Conversely, when the same pulse parameters are applied in a high-conductivity HBSS buffer and high PRRs are applied, the local temperature rises and yields either cell sensitization to nsPEFs or thermal damage.
Collapse
Affiliation(s)
- Rosa Orlacchio
- University Bordeaux, CNRS, IMS, UMR 5218, 33400 Talence, France;
- École Pratique des Hautes Études, PSL Research University, 75014 Paris, France
| | - Jelena Kolosnjaj-Tabi
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UT3), 31062 Toulouse, France; (J.K.-T.); (N.M.); (M.P.R.)
| | - Nicolas Mattei
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UT3), 31062 Toulouse, France; (J.K.-T.); (N.M.); (M.P.R.)
| | - Philippe Lévêque
- University Limoges, CNRS, XLIM, UMR 7252, 87000 Limoges, France; (P.L.); (D.A.-C.)
| | - Marie Pierre Rols
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UT3), 31062 Toulouse, France; (J.K.-T.); (N.M.); (M.P.R.)
| | - Delia Arnaud-Cormos
- University Limoges, CNRS, XLIM, UMR 7252, 87000 Limoges, France; (P.L.); (D.A.-C.)
- Institut Universitaire de France (IUF), 75005 Paris, France
| | - Muriel Golzio
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UT3), 31062 Toulouse, France; (J.K.-T.); (N.M.); (M.P.R.)
| |
Collapse
|
12
|
Lisec B, Markelc B, Ursic Valentinuzzi K, Sersa G, Cemazar M. The effectiveness of calcium electroporation combined with gene electrotransfer of a plasmid encoding IL-12 is tumor type-dependent. Front Immunol 2023; 14:1189960. [PMID: 37304301 PMCID: PMC10247961 DOI: 10.3389/fimmu.2023.1189960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction In calcium electroporation (CaEP), electroporation enables the cellular uptake of supraphysiological concentrations of Ca2+, causing the induction of cell death. The effectiveness of CaEP has already been evaluated in clinical trials; however, confirmatory preclinical studies are still needed to further elucidate its effectiveness and underlying mechanisms. Here, we tested and compared its efficiency on two different tumor models to electrochemotherapy (ECT) and in combination with gene electrotransfer (GET) of a plasmid encoding interleukin-12 (IL-12). We hypothesized that IL-12 potentiates the antitumor effect of local ablative therapies as CaEP and ECT. Methods The effect of CaEP was tested in vitro as well as in vivo in murine melanoma B16-F10 and murine mammary carcinoma 4T1 in comparison to ECT with bleomycin. Specifically, the treatment efficacy of CaEP with increasing calcium concentrations alone or in combination with IL-12 GET in different treatment protocols was investigated. We closely examined the tumor microenvironment by immunofluorescence staining of immune cells, as well as blood vessels and proliferating cells. Results In vitro, CaEP and ECT with bleomycin reduced cell viability in a dose-dependent manner. We observed no differences in sensitivity between the two cell lines. A dose-dependent response was also observed in vivo; however, the efficacy was better in 4T1 tumors than in B16-F10 tumors. In 4T1 tumors, CaEP with 250 mM Ca resulted in more than 30 days of growth delay, which was comparable to ECT with bleomycin. In contrast, adjuvant peritumoral application of IL-12 GET after CaEP prolonged the survival of B16-F10, but not 4T1-bearing mice. Moreover, CaEP with peritumoral IL-12 GET modified tumor immune cell populations and tumor vasculature. Conclusions Mice bearing 4T1 tumors responded better to CaEP in vivo than mice bearing B16-F10 tumors, even though a similar response was observed in vitro. Namely, one of the most important factors might be involvement of the immune system. This was confirmed by the combination of CaEP or ECT with IL-12 GET, which further enhanced antitumor effectiveness. However, the potentiation of CaEP effectiveness was also highly dependent on tumor type; it was more pronounced in poorly immunogenic B16-F10 tumors compared to moderately immunogenic 4T1 tumors.
Collapse
Affiliation(s)
- Barbara Lisec
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Katja Ursic Valentinuzzi
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| |
Collapse
|
13
|
Silkuniene G, Mangalanathan UM, Rossi A, Mollica PA, Pakhomov AG, Pakhomova O. Identification of Proteins Involved in Cell Membrane Permeabilization by Nanosecond Electric Pulses (nsEP). Int J Mol Sci 2023; 24:ijms24119191. [PMID: 37298142 DOI: 10.3390/ijms24119191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
The study was aimed at identifying endogenous proteins which assist or impede the permeabilized state in the cell membrane disrupted by nsEP (20 or 40 pulses, 300 ns width, 7 kV/cm). We employed a LentiArray CRISPR library to generate knockouts (KOs) of 316 genes encoding for membrane proteins in U937 human monocytes stably expressing Cas9 nuclease. The extent of membrane permeabilization by nsEP was measured by the uptake of Yo-Pro-1 (YP) dye and compared to sham-exposed KOs and control cells transduced with a non-targeting (scrambled) gRNA. Only two KOs, for SCNN1A and CLCA1 genes, showed a statistically significant reduction in YP uptake. The respective proteins could be part of electropermeabilization lesions or increase their lifespan. In contrast, as many as 39 genes were identified as likely hits for the increased YP uptake, meaning that the respective proteins contributed to membrane stability or repair after nsEP. The expression level of eight genes in different types of human cells showed strong correlation (R > 0.9, p < 0.02) with their LD50 for lethal nsEP treatments, and could potentially be used as a criterion for the selectivity and efficiency of hyperplasia ablations with nsEP.
Collapse
Affiliation(s)
- Giedre Silkuniene
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
- Institute for Digestive System Research, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Uma M Mangalanathan
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Alessandra Rossi
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
- Department of Translational Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Peter A Mollica
- College of Health Sciences, Old Dominion University, Norfolk, VA 23508, USA
| | - Andrei G Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Olga Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| |
Collapse
|
14
|
Ferguson CA, Santangelo C, Marramiero L, Farina M, Pietrangelo T, Cheng X. Broadband Electrical Spectroscopy to Distinguish Single-Cell Ca 2+ Changes Due to Ionomycin Treatment in a Skeletal Muscle Cell Line. SENSORS (BASEL, SWITZERLAND) 2023; 23:4358. [PMID: 37177559 PMCID: PMC10181519 DOI: 10.3390/s23094358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/22/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Many skeletal muscle diseases such as muscular dystrophy, myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), and sarcopenia share the dysregulation of calcium (Ca2+) as a key mechanism of disease at a cellular level. Cytosolic concentrations of Ca2+ can signal dysregulation in organelles including the mitochondria, nucleus, and sarcoplasmic reticulum in skeletal muscle. In this work, a treatment is applied to mimic the Ca2+ increase associated with these atrophy-related disease states, and broadband impedance measurements are taken for single cells with and without this treatment using a microfluidic device. The resulting impedance measurements are fitted using a single-shell circuit simulation to show calculated electrical dielectric property contributions based on these Ca2+ changes. From this, similar distributions were seen in the Ca2+ from fluorescence measurements and the distribution of the S-parameter at a single frequency, identifying Ca2+ as the main contributor to the electrical differences being identified. Extracted dielectric parameters also showed different distribution patterns between the untreated and ionomycin-treated groups; however, the overall electrical parameters suggest the impact of Ca2+-induced changes at a wider range of frequencies.
Collapse
Affiliation(s)
- Caroline A. Ferguson
- Department of Bioengineering, P.C. Rossin College of Engineering and Applied Sciences, Lehigh University, Bethlehem, PA 18015, USA
| | - Carmen Santangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Lorenzo Marramiero
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Farina
- Department of Engineering of Information, University Politecnica delle Marche, 60131 Ancona, Italy
| | - Tiziana Pietrangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Xuanhong Cheng
- Department of Bioengineering, P.C. Rossin College of Engineering and Applied Sciences, Lehigh University, Bethlehem, PA 18015, USA
- Department of Materials Science and Engineering, P.C. Rossin College of Engineering and Applied Sciences, Lehigh University, Bethlehem, PA 18015, USA
| |
Collapse
|
15
|
Gudvangen E, Mangalanathan U, Semenov I, Kiester AS, Keppler MA, Ibey BL, Bixler JN, Pakhomov AG. Pulsed Electric Field Ablation of Esophageal Malignancies and Mitigating Damage to Smooth Muscle: An In Vitro Study. Int J Mol Sci 2023; 24:ijms24032854. [PMID: 36769172 PMCID: PMC9917603 DOI: 10.3390/ijms24032854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Cancer ablation therapies aim to be efficient while minimizing damage to healthy tissues. Nanosecond pulsed electric field (nsPEF) is a promising ablation modality because of its selectivity against certain cell types and reduced neuromuscular effects. We compared cell killing efficiency by PEF (100 pulses, 200 ns-10 µs duration, 10 Hz) in a panel of human esophageal cells (normal and pre-malignant epithelial and smooth muscle). Normal epithelial cells were less sensitive than the pre-malignant ones to unipolar PEF (15-20% higher LD50, p < 0.05). Smooth muscle cells (SMC) oriented randomly in the electric field were more sensitive, with 30-40% lower LD50 (p < 0.01). Trains of ten, 300-ns pulses at 10 kV/cm caused twofold weaker electroporative uptake of YO-PRO-1 dye in normal epithelial cells than in either pre-malignant cells or in SMC oriented perpendicularly to the field. Aligning SMC with the field reduced the dye uptake fourfold, along with a twofold reduction in Ca2+ transients. A 300-ns pulse induced a twofold smaller transmembrane potential in cells aligned with the field, making them less vulnerable to electroporation. We infer that damage to SMC from nsPEF ablation of esophageal malignancies can be minimized by applying the electric field parallel to the predominant SMC orientation.
Collapse
Affiliation(s)
- Emily Gudvangen
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Uma Mangalanathan
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Iurii Semenov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Allen S. Kiester
- Bioeffects Division, Airman System Directorate, 711th Human Performance Wing, Air Force Research Laboratory, JBSA Fort Sam Houston, San Antonio, TX 78234, USA
| | | | - Bennett L. Ibey
- Bioeffects Division, Airman System Directorate, 711th Human Performance Wing, Air Force Research Laboratory, JBSA Fort Sam Houston, San Antonio, TX 78234, USA
| | - Joel N. Bixler
- Bioeffects Division, Airman System Directorate, 711th Human Performance Wing, Air Force Research Laboratory, JBSA Fort Sam Houston, San Antonio, TX 78234, USA
| | - Andrei G. Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
- Correspondence:
| |
Collapse
|
16
|
Sowa PW, Kiełbik AS, Pakhomov AG, Gudvangen E, Mangalanathan U, Adams V, Pakhomova ON. How to alleviate cardiac injury from electric shocks at the cellular level. Front Cardiovasc Med 2022; 9:1004024. [PMID: 36620647 PMCID: PMC9812960 DOI: 10.3389/fcvm.2022.1004024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Electric shocks, the only effective therapy for ventricular fibrillation, also electroporate cardiac cells and contribute to the high-mortality post-cardiac arrest syndrome. Copolymers such as Poloxamer 188 (P188) are known to preserve the membrane integrity and viability of electroporated cells, but their utility against cardiac injury from cardiopulmonary resuscitation (CPR) remains to be established. We studied the time course of cell killing, mechanisms of cell death, and protection with P188 in AC16 human cardiomyocytes exposed to micro- or nanosecond pulsed electric field (μsPEF and nsPEF) shocks. A 3D printer was customized with an electrode holder to precisely position electrodes orthogonal to a cell monolayer in a nanofiber multiwell plate. Trains of nsPEF shocks (200, 300-ns pulses at 1.74 kV) or μsPEF shocks (20, 100-μs pulses at 300 V) produced a non-uniform electric field enabling efficient measurements of the lethal effect in a wide range of the electric field strength. Cell viability and caspase 3/7 expression were measured by fluorescent microscopy 2-24 h after the treatment. nsPEF shocks caused little or no caspase 3/7 activation; most of the lethally injured cells were permeable to propidium dye already at 2 h after the exposure. In contrast, μsPEF shocks caused strong activation of caspase 3/7 at 2 h and the number of dead cells grew up to 24 h, indicating the prevalence of the apoptotic death pathway. P188 at 0.2-1% reduced cell death, suggesting its potential utility in vivo to alleviate electric injury from defibrillation.
Collapse
Affiliation(s)
- Pamela W. Sowa
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States,Laboratory of Molecular and Experimental Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany,Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany,*Correspondence: Pamela W. Sowa,
| | - Aleksander S. Kiełbik
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States,Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Wrocław, Poland
| | - Andrei G. Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Emily Gudvangen
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Uma Mangalanathan
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Olga N. Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| |
Collapse
|
17
|
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
|
18
|
Kulbacka J, Rembiałkowska N, Szewczyk A, Rossowska J, Drąg-Zalesińska M, Kulbacki M, Choromańska A. Nanosecond PEF Induces Oxidative Stress and Apoptosis via Proteasomal Activity Inhibition in Gastric Adenocarcinoma Cells with Drug Resistance. Int J Mol Sci 2022; 23:12943. [PMID: 36361727 PMCID: PMC9657809 DOI: 10.3390/ijms232112943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/06/2022] [Accepted: 10/18/2022] [Indexed: 08/01/2023] Open
Abstract
Nanosecond (ns) pulsed electric field (PEF) is a technology in which the application of ultra-short electrical pulses can be used to disrupt the barrier function of cell plasma and internal membranes. Disruptions of the membrane integrity cause a substantial imbalance in cell homeostasis in which oxidative stress is a principal component. In the present study, nsPEF-induced oxidative stress was investigated in two gastric adenocarcinoma cell lines (EPG85-257P and EPG85-257RDB) which differ by their sensitivity to daunorubicin. Cells were exposed to 200 pulses of 10 ns duration, with the amplitude and pulse repetition frequency at 1 kHz, with electric field intensity varying from 12.5 to 50 kV/cm. The electroporation buffer contained either 1 mM or 2 mM calcium chloride. CellMask DeepRed visualized cell plasma permeabilization, Fluo-4 was used to visualize internal calcium ions content, and F-actin was labeled with AlexaFluor®488 for the cytoskeleton. The cellular viability was determined by MTT assay. An alkaline and neutral comet assay was employed to detect apoptotic and necrotic cell death. The luminescent method estimated the modifications in GSSG/GSH redox potential and the imbalance of proteasomal activity (chymotrypsin-, trypsin- and caspase-like). The reactive oxygen species (ROS) level was measured by flow cytometry using dihydroethidium (DHE) dye. Morphological visualization indicated cell shrinkage, affected cell membranes (characteristic bubbles and changed cell shape), and the reorganization of actin fibers with sites of its dense concentration; the effect was more intense with the increasing electric field strength. The most significant decrease in cell viability and GSSG/GSH redox potential was noted at the highest amplitude of 50 kV/cm, and calcium ions amplified this effect. nsPEF, particularly with calcium ions, inhibited proteasomal activities, resulting in increased protein degradation. nsPEF increased the percentage of apoptotic cells and ROS levels. The EPG85-257 RDB cell line, which is resistant to standard chemotherapy, was more sensitive to applied nsPEF protocols. The applied nsPEF method disrupted the metabolism of cancer cells and induced apoptotic cell death. The nsPEF ability to cause apoptosis, oxidative stress, and protein degradation make the nsPEF methodology a suitable alternative to current anticancer pharmacological methods.
Collapse
Affiliation(s)
- Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wroclaw, 50-335 Wroclaw, Poland
| | - Joanna Rossowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-422 Wroclaw, Poland
| | - Małgorzata Drąg-Zalesińska
- Division of Histology and Embryology, Division of Human Morpholog and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Marek Kulbacki
- Polish-Japanese Academy of Information Technology, 02-008 Warsaw, Poland
- DIVE IN AI, 53-307 Wroclaw, Poland
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
| |
Collapse
|
19
|
Electroporation and cell killing by milli- to nanosecond pulses and avoiding neuromuscular stimulation in cancer ablation. Sci Rep 2022; 12:1763. [PMID: 35110567 PMCID: PMC8811018 DOI: 10.1038/s41598-022-04868-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/31/2021] [Indexed: 12/21/2022] Open
Abstract
Ablation therapies aim at eradication of tumors with minimal impact on surrounding healthy tissues. Conventional pulsed electric field (PEF) treatments cause pain and muscle contractions far beyond the ablation area. The ongoing quest is to identify PEF parameters efficient at ablation but not at stimulation. We measured electroporation and cell killing thresholds for 150 ns–1 ms PEF, uni- and bipolar, delivered in 10- to 300-pulse trains at up to 1 MHz rates. Monolayers of murine colon carcinoma cells exposed to PEF were stained with YO-PRO-1 dye to detect electroporation. In 2–4 h, dead cells were labeled with propidium. Electroporation and cell death thresholds determined by matching the stained areas to the electric field intensity were compared to nerve excitation thresholds (Kim et al. in Int J Mol Sci 22(13):7051, 2021). The minimum fourfold ratio of cell killing and stimulation thresholds was achieved with bipolar nanosecond PEF (nsPEF), a sheer benefit over a 500-fold ratio for conventional 100-µs PEF. Increasing the bipolar nsPEF frequency up to 100 kHz within 10-pulse bursts increased ablation thresholds by < 20%. Restricting such bursts to the refractory period after nerve excitation will minimize the number of neuromuscular reactions while maintaining the ablation efficiency and avoiding heating.
Collapse
|
20
|
Steelman ZA, Coker ZN, Kiester A, Noojin G, Ibey BL, Bixler JN. Quantitative phase microscopy monitors subcellular dynamics in single cells exposed to nanosecond pulsed electric fields. JOURNAL OF BIOPHOTONICS 2021; 14:e202100125. [PMID: 34291579 DOI: 10.1002/jbio.202100125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/11/2021] [Accepted: 07/16/2021] [Indexed: 06/13/2023]
Abstract
A substantial body of literature exists to study the dynamics of single cells exposed to short duration (<1 μs), high peak power (~1 MV/m) transient electric fields. Much of this research is limited to traditional fluorescence-based microscopy techniques, which introduce exogenous agents to the culture and are only sensitive to a single molecular target. Quantitative phase imaging (QPI) is a coherent imaging modality which uses optical path length as a label-free contrast mechanism, and has proven highly effective for the study of single-cell dynamics. In this work, we introduce QPI as a useful imaging tool for the study of cells undergoing cytoskeletal remodeling after nanosecond pulsed electric field (nsPEF) exposure. In particular, we use cell swelling, dry mass and disorder strength measurements derived from QPI phase images to monitor the cellular response to nsPEFs. We hope this demonstration of QPI's utility will lead to a further adoption of the technique for the study of directed energy bioeffects.
Collapse
Affiliation(s)
- Zachary A Steelman
- National Research Council Research Associateship Program, Washington, District of Columbia, USA
| | - Zachary N Coker
- Department of Physics and Astronomy, Texas A&M University, College Station, Texas, USA
- SAIC, San Antonio, Texas, USA
| | - Allen Kiester
- 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, JBSA Fort Sam Houston, San Antonio, Texas, USA
| | | | - Bennett L Ibey
- 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, JBSA Fort Sam Houston, San Antonio, Texas, USA
| | - Joel N Bixler
- 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, JBSA Fort Sam Houston, San Antonio, Texas, USA
| |
Collapse
|
21
|
Yang Q, Kajimoto S, Kobayashi Y, Hiramatsu H, Nakabayashi T. Regulation of Cell Volume by Nanosecond Pulsed Electric Fields. J Phys Chem B 2021; 125:10692-10700. [PMID: 34519209 DOI: 10.1021/acs.jpcb.1c06058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Stimulation of cells by nanosecond pulsed electric fields (nsPEFs) has attracted attention as a technology for medical applications such as cancer treatment. nsPEFs have been shown to affect intracellular environments without significant damage to cell membranes; however, the mechanism underlying the effect of nsPEFs on cells remains unclear. In this study, we constructed electrodes for applying nsPEFs and analyzed the change in volume of a single cell due to nsPEFs using fluorescence and Raman microscopy. It was shown that the direction of the change depended on the applied electric field; expansion due to the influx of water was observed at high electric field, and cell shrinkage was observed at low electric field. The change in cell volume was correlated to the change in the intracellular Ca2+ concentration, and nsPEFs-induced shrinking was not observed when the Ca2+-free medium was used. This result suggests that the cell shrinkage is related to the regulatory volume decrease where the cell adjusts the increase in intracellular Ca2+ concentration, inducing the efflux of ions and water from the cell.
Collapse
Affiliation(s)
- Qi Yang
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Shinji Kajimoto
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan.,JST PRESTO, Kawaguchi, Saitama 332-0012, Japan
| | - Yuki Kobayashi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Hirotsugu Hiramatsu
- Department of Applied Chemistry and Institute of Molecular Science, National Yang Ming Chiao Tung University, 1001, Ta-Hsueh Road, Hsinchu 30010, Taiwan.,Center for Emergent Functional Matter Science, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Takakazu Nakabayashi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
22
|
Ca 2+ roles in electroporation-induced changes of cancer cell physiology: From membrane repair to cell death. Bioelectrochemistry 2021; 142:107927. [PMID: 34425390 DOI: 10.1016/j.bioelechem.2021.107927] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022]
Abstract
The combination of Ca2+ ions and electroporation has gained attention as potential alternative to electrochemotherapy. Ca2+ is an important component of the cell membrane repair system and its presence directly influences the dynamics of the pore cycle after electroporation which can be exploited for cancer therapies. Here, the influence of Ca2+ concentration is investigated on small molecule electrotransfer and release of Calcein from 4T1, MX-1, B16F10, U87 cancer cells after cell exposure to microsecond electric pulses. Moreover, we investigated simultaneous molecule electrotransfer and intracellular calcium ion influx when media was supplemented with different Ca2+ concentrations. Results show that increased concentrations of calcium ions reduce the electrotransfer of small molecules to different lines of cancer cells as well as the release of Calcein. These effects are related with an enhanced membrane repair mechanism. Overall, we show that the efficiency of molecular electrotransfer can be controlled by regulating Ca2+ concentration in the electroporation medium. For the first time, the cause of cancer cell death in vitro from 1 mM CaCl2 concentrations is related to the irreversible loss of Ca2+ homeostasis after cell electroporation. Our findings provide fundamental insight on the mechanisms of Ca2+ electroporation that might lead to improved therapeutic outcomes.
Collapse
|
23
|
Kiełbik A, Szlasa W, Novickij V, Szewczyk A, Maciejewska M, Saczko J, Kulbacka J. Effects of high-frequency nanosecond pulses on prostate cancer cells. Sci Rep 2021; 11:15835. [PMID: 34349171 PMCID: PMC8339066 DOI: 10.1038/s41598-021-95180-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023] Open
Abstract
Electroporation with pulsed electric fields show a potential to be applied as an experimental focal therapy of tumors. Sub-microsecond regime of electric pulses displays unique electrophysical features operative in cells and membranes. Recently, MHz compression of nanosecond pulses electric fields (nsPEFs) bursts proved to enhance the effectiveness of the therapy. High morbidity of prostate cancer (PCa) and risk of overtreatment associated with this malignancy call for new minimal-invasive treatment alternative. Herein we present the in vitro study for developing applications based on this new technology. In this study, we used flow cytometric analysis, cell viability assay, caspase activity analysis, wound healing assay, confocal microscopy study, and immunofluorescence to investigate the biological effect of high-frequency nsPEFs on PCa cells. Our results show that high-frequency nsPEFs induces the permeabilization and cell death of PCa cells. The cytotoxicity is significantly enhanced in MHz compression of pulses and with the presence of extracellular Ca2+. High-frequency nsPEFs trigger changes in PCa cells' cytoskeleton and their mobility. The presented data show a therapeutic potential of high-frequency nsPEFs in a PCa setting. The sub-microsecond regime of pulses can potentially be applied in nanosecond electroporation protocols for PCa treatment.
Collapse
Affiliation(s)
- Aleksander Kiełbik
- grid.4495.c0000 0001 1090 049XMedical University Hospital, Borowska 213, 50-556 Wrocław, Poland ,grid.4495.c0000 0001 1090 049XDepartment of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wrocław, Poland
| | - Wojciech Szlasa
- grid.4495.c0000 0001 1090 049XFaculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Vitalij Novickij
- grid.9424.b0000 0004 1937 1776Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Anna Szewczyk
- grid.4495.c0000 0001 1090 049XDepartment of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wrocław, Poland ,grid.8505.80000 0001 1010 5103Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 50-328 Wrocław, Poland
| | - Magdalena Maciejewska
- grid.413454.30000 0001 1958 0162Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
| | - Jolanta Saczko
- grid.4495.c0000 0001 1090 049XDepartment of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wrocław, Poland
| | - Julita Kulbacka
- grid.4495.c0000 0001 1090 049XDepartment of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
24
|
The Impact of Extracellular Ca 2+ and Nanosecond Electric Pulses on Sensitive and Drug-Resistant Human Breast and Colon Cancer Cells. Cancers (Basel) 2021; 13:cancers13133216. [PMID: 34203184 PMCID: PMC8268418 DOI: 10.3390/cancers13133216] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary The drug resistance phenomenon in cancer constantly induces problems in therapeutic protocols. Pulsed electric fields (PEFs) seem to be a promising method in drug molecule delivery. Here we have proved that electroporation supported by calcium ions can alternate the activity of drug resistance proteins. Our results indicated that MDR1 expression is not significantly modified by nanosecond electroporation in multidrug-resistant cells. However, PEF significantly inhibited MDR1 activity and cell viability when combined with calcium ions. Abstract (1) Background: Calcium electroporation (CaEP) is based on the application of electrical pulses to permeabilize cells (electroporation) and allow cytotoxic doses of calcium to enter the cell. (2) Methods: In this work, we have used doxorubicin-resistant (DX) and non-resistant models of human breast cancer (MCF-7/DX, MCF-7/WT) and colon cancer cells (LoVo, LoVo/DX), and investigated the susceptibility of the cells to extracellular Ca2+ and electric fields in the 20 ns–900 ns pulse duration range. (3) Results: We have observed that colon cancer cells were less susceptible to PEF than breast cancer cells. An extracellular Ca2+ (2 mM) with PEF was more disruptive for DX-resistant cells. The expression of glycoprotein P (MDR1, P-gp) as a drug resistance marker was detected by the immunofluorescent (CLSM) method and rhodamine-123 efflux as an MDR1 activity. MDR1 expression was not significantly modified by nanosecond electroporation in multidrug-resistant cells, but a combination with calcium ions significantly inhibited MDR1 activity and cell viability. (4) Conclusions: We believe that PEF with calcium ions can reduce drug resistance by inhibiting drug efflux activity. This phenomenon of MDR mechanism disruption seems promising in anticancer protocols.
Collapse
|
25
|
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
|
26
|
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
|
27
|
Gu Y, Zhang L, Yang H, Zhuang J, Sun Z, Guo J, Guan M. Nanosecond pulsed electric fields impair viability and mucin expression in mucinous colorectal carcinoma cell. Bioelectrochemistry 2021; 141:107844. [PMID: 34052542 DOI: 10.1016/j.bioelechem.2021.107844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/20/2022]
Abstract
Nanosecond pulsed electric fields (nsPEFs) are a non-thermal technology that can induce a myriad of biological responses and changes in cellular physiology. nsPEFs have gained significant attention as a novel cancer therapy. However, studies investigating the application of nsPEF in mucinous carcinomas are scarce. In this study, we explored several biological responses in two mucinous colorectal adenocarcinoma cell lines, LS 174T and HT-29, to nsPEF treatment. We determined the overall cell survival and viability rates following nsPEF treatment using CCK-8 and colony formation assays. We measured the intracellular effects of nsPEF treatment by analyzing cell cycle distribution, cell apoptosis and mitochondrial potential. We also analyzed mucin production at both mRNA and protein levels. Our results showed that nsPEF treatment significantly reduced mucinous cell viability in a dose-dependent manner. nsPEF treatment increased cell cycles arrest at G0/G1 while the proportion of G2/M cells gradually decreased. Cell apoptosis increased following nsPEF treatment with a clear loss in mitochondrial membrane potential. Furthermore, the protein expression of functional mucin family members decreased after nsPEF treatment. In conclusion, nsPEF treatment reduced MCRC cell viability, cell proliferation, and mucin protein production while promoted apoptosis. Our work is a pilot study that projects some insights into the potential clinical applications of nsPEFs in treating mucinous colorectal carcinoma.
Collapse
Affiliation(s)
- Yiran Gu
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, Jiangsu, China; School of Life Science, Shanghai University, Shanghai 200444, China
| | - Long Zhang
- State Key Laboratory of Solid-State Lighting Research Center of Light for Health, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, Jiangsu, China
| | - Hua Yang
- Department of General Surgery, Zhongshan Hospital (South Branch), Fudan University, Shanghai 200083, China
| | - Jie Zhuang
- State Key Laboratory of Solid-State Lighting Research Center of Light for Health, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, Jiangsu, China
| | - Zhenglong Sun
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| | - Jinsong Guo
- State Key Laboratory of Solid-State Lighting Research Center of Light for Health, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, Jiangsu, China
| | - Miao Guan
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
28
|
Poudel A, Oludiran A, Sözer EB, Casciola M, Purcell EB, Muratori C. Growth in a biofilm sensitizes Cutibacterium acnes to nanosecond pulsed electric fields. Bioelectrochemistry 2021; 140:107797. [PMID: 33773215 DOI: 10.1016/j.bioelechem.2021.107797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 10/21/2022]
Abstract
The Gram-positive anaerobic bacterium Cutibacterium acnes (C. acnes) is a commensal of the human skin, but also an opportunistic pathogen that contributes to the pathophysiology of the skin disease acne vulgaris. C. acnes can form biofilms; cells in biofilms are more resilient to antimicrobial stresses. Acne therapeutic options such as topical or systemic antimicrobial treatments often show incomplete responses. In this study we measured the efficacy of nanosecond pulsed electric fields (nsPEF), a new promising cell and tissue ablation technology, to inactivate C. acnes. Our results show that all tested nsPEF doses (250 to 2000 pulses, 280 ns pulses, 28 kV/cm, 5 Hz; 0.5 to 4 kJ/ml) failed to inactivate planktonic C. acnes and that pretreatment with lysozyme, a naturally occurring cell-wall-weakening enzyme, increased C. acnes vulnerability to nsPEF. Surprisingly, growth in a biofilm appears to sensitize C. acnes to nsPEF-induced stress, as C. acnes biofilm-derived cells showed increased cell death after nsPEF treatments that did not affect planktonic cells. Biofilm inactivation by nsPEF was confirmed by treating intact biofilms grown on glass coverslips with an indium oxide conductive layer. Altogether our results show that, contrary to other antimicrobial agents, nsPEF kill more efficiently bacteria in biofilms than planktonic cells.
Collapse
Affiliation(s)
- Asia Poudel
- Old Dominion University, Department of Chemistry and Biochemistry, USA
| | - Adenrele Oludiran
- Old Dominion University, Department of Chemistry and Biochemistry, USA
| | - Esin B Sözer
- Old Dominion University, Frank Reidy Research Center for Bioelectrics, USA
| | - Maura Casciola
- Old Dominion University, Frank Reidy Research Center for Bioelectrics, USA; Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Erin B Purcell
- Old Dominion University, Department of Chemistry and Biochemistry, USA.
| | - Claudia Muratori
- Old Dominion University, Frank Reidy Research Center for Bioelectrics, USA.
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Wasson EM, Alinezhadbalalami N, Brock RM, Allen IC, Verbridge SS, Davalos RV. Understanding the role of calcium-mediated cell death in high-frequency irreversible electroporation. Bioelectrochemistry 2020; 131:107369. [PMID: 31706114 PMCID: PMC10039453 DOI: 10.1016/j.bioelechem.2019.107369] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022]
Abstract
High-frequency irreversible electroporation (H-FIRE) is an emerging electroporation-based therapy used to ablate cancerous tissue. Treatment consists of delivering short, bipolar pulses (1-10μs) in a series of 80-100 bursts (1 burst/s, 100μs on-time). Reducing pulse duration leads to reduced treatment volumes compared to traditional IRE, therefore larger voltages must be applied to generate ablations comparable in size. We show that adjuvant calcium enhances ablation area in vitro for H-FIRE treatments of several pulse durations (1, 2, 5, 10μs). Furthermore, H-FIRE treatment using 10μs pulses delivered with 1mM CaCl2 results in cell death thresholds (771±129V/cm) comparable to IRE thresholds without calcium (698±103V/cm). Quantifying the reversible electroporation threshold revealed that CaCl2 enhances the permeabilization of cells compared to a NaCl control. Gene expression analysis determined that CaCl2 upregulates expression of eIFB5 and 60S ribosomal subunit genes while downregulating NOX1/4, leading to increased signaling in pathways that may cause necroptosis. The opposite was found for control treatment without CaCl2 suggesting cells experience an increase in pro survival signaling. Our study is the first to identify key genes and signaling pathways responsible for differences in cell response to H-FIRE treatment with and without calcium.
Collapse
Affiliation(s)
- Elisa M Wasson
- Department of Mechanical Engineering, Virginia Tech, Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Nastaran Alinezhadbalalami
- Department of Biomedical Engineering and Mechanics, Virginia Tech- Wake Forest University, 325 Stanger Street, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Rebecca M Brock
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, 1 Riverside Circle, Roanoke, VA 24016, United States of America; Department of Biomedical Sciences and Pathobiology, Virginia Tech, 205 Duck Pond Drive, Blacksburg, VA 24061, USA.
| | - Irving C Allen
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, 1 Riverside Circle, Roanoke, VA 24016, United States of America; Department of Biomedical Sciences and Pathobiology, Virginia Tech, 205 Duck Pond Drive, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Scott S Verbridge
- Department of Biomedical Engineering and Mechanics, Virginia Tech- Wake Forest University, 325 Stanger Street, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Rafael V Davalos
- Department of Mechanical Engineering, Virginia Tech, Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA 24061, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech- Wake Forest University, 325 Stanger Street, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| |
Collapse
|
32
|
A Comprehensive Review of Calcium Electroporation -A Novel Cancer Treatment Modality. Cancers (Basel) 2020; 12:cancers12020290. [PMID: 31991784 PMCID: PMC7073222 DOI: 10.3390/cancers12020290] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/17/2020] [Accepted: 01/19/2020] [Indexed: 12/21/2022] Open
Abstract
Calcium electroporation is a potential novel anti-cancer treatment where high calcium concentrations are introduced into cells by electroporation, a method where short, high voltage pulses induce transient permeabilisation of the plasma membrane allowing passage of molecules into the cytosol. Calcium is a tightly regulated, ubiquitous second messenger involved in many cellular processes including cell death. Electroporation increases calcium uptake leading to acute and severe ATP depletion associated with cancer cell death. This comprehensive review describes published data about calcium electroporation applied in vitro, in vivo, and clinically from the first publication in 2012. Calcium electroporation has been shown to be a safe and efficient anti-cancer treatment in clinical studies with cutaneous metastases and recurrent head and neck cancer. Normal cells have been shown to be less affected by calcium electroporation than cancer cells and this difference might be partly induced by differences in membrane repair, expression of calcium transporters, and cellular structural changes. Interestingly, both clinical data and preclinical studies have indicated a systemic immune response induced by calcium electroporation. New cancer treatments are needed, and calcium electroporation represents an inexpensive and efficient treatment with few side effects, that could potentially be used worldwide and for different tumor types.
Collapse
|
33
|
Michel O, Błasiak P, Saczko J, Kulbacka J, Drąg-Zalesińska M, Rzechonek A. Electropermeabilization of metastatic chondrosarcoma cells from primary cell culture. Biotechnol Appl Biochem 2019; 66:945-954. [PMID: 31476023 DOI: 10.1002/bab.1809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 08/20/2019] [Indexed: 11/12/2022]
Abstract
Primary cell cultures are challenging, but reliable model reflecting tumor response in vitro. The study was designed to examine if the increased electropermeabilization can overcame initial drug insensitivity in chondrosarcoma cells from lung metastasis. We established a primary cell culture and evaluated the cytotoxic impact of four drugs-cisplatin (CDDP), camptothecin, 2-methoxyestradiol, and leucovorin calcium (LeuCa). After determination of parameters allowing for electropermeabilization, we performed electrochemotherapy in vitro with the least toxic drugs-CDDP and LeuCa. Although combining CDDP and leucovorin together increased their toxicity and supported apoptosis, application of pulsed electric fields (PEFs) brought no advantage for their efficacy. The study emphasizes the need for introduction of primary cell cultures into studies on pulse electric fields as model frequently less sensitive to PEF-based treatments than continuous cell lines.
Collapse
Affiliation(s)
- Olga Michel
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Błasiak
- Department of Thoracic Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Wroclaw, Poland
| | - Małgorzata Drąg-Zalesińska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Adam Rzechonek
- Department of Thoracic Surgery, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
34
|
Nanosecond pulsed electric fields induce extracellular release of chromosomal DNA and histone citrullination in neutrophil-differentiated HL-60 cells. Sci Rep 2019; 9:8451. [PMID: 31186478 PMCID: PMC6559984 DOI: 10.1038/s41598-019-44817-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022] Open
Abstract
Nanosecond pulsed electric fields (nsPEFs) have gained attention as a novel physical stimulus for life sciences. Although cancer therapy is currently their promising application, nsPEFs have further potential owing to their ability to elicit various cellular responses. This study aimed to explore stimulatory actions of nsPEFs, and we used HL-60 cells that were differentiated into neutrophils under cultured conditions. Exposure of neutrophil-differentiated HL-60 cells to nsPEFs led to the extracellular release of chromosomal DNA, which appears to be equivalent to neutrophil extracellular traps (NETs) that serve as a host defense mechanism against pathogens. Fluorometric measurement of extracellular DNA showed that DNA extrusion was rapidly induced after nsPEF exposure and increased over time. Western blot analysis demonstrated that nsPEFs induced histone citrullination that is the hydrolytic conversion of arginine to citrulline on histones and facilitates chromatin decondensation. DNA extrusion and histone citrullination by nsPEFs were cell type-specific and Ca2+-dependent events. Taken together, these observations suggest that nsPEFs drive the mechanism for neutrophil-specific immune response without infection, highlighting a novel aspect of nsPEFs as a physical stimulus.
Collapse
|
35
|
Chiapperino MA, Bia P, Caratelli D, Gielis J, Mescia L, Dermol‐Černe J, Miklavčič D. Nonlinear Dispersive Model of Electroporation for Irregular Nucleated Cells. Bioelectromagnetics 2019; 40:331-342. [DOI: 10.1002/bem.22197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/03/2019] [Indexed: 12/15/2022]
Affiliation(s)
| | - Pietro Bia
- Department of Design SolutionElettronica S.p.ARome Italy
| | - Diego Caratelli
- Antenna Division, the Antenna CompanyHigh Tech CampusEindhoven The Netherlands
| | - Johan Gielis
- Department of BioengineeringUniversity of AntwerpAntwerp Belgium
| | - Luciano Mescia
- Department of Electrical and Information EngineeringPolytechnic University of BariBari Italy
| | - Janja Dermol‐Černe
- Department of Biocybernetics, Faculty of Electrical EngineeringUniversity of LjubljanaLjubljana Slovenia
| | - Damijan Miklavčič
- Department of Biocybernetics, Faculty of Electrical EngineeringUniversity of LjubljanaLjubljana Slovenia
| |
Collapse
|
36
|
Plaschke CC, Gehl J, Johannesen HH, Fischer BM, Kjaer A, Lomholt AF, Wessel I. Calcium electroporation for recurrent head and neck cancer: A clinical phase I study. Laryngoscope Investig Otolaryngol 2019; 4:49-56. [PMID: 30828619 PMCID: PMC6383303 DOI: 10.1002/lio2.233] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/24/2018] [Indexed: 12/17/2022] Open
Abstract
Background Calcium electroporation is a novel cancer treatment, which combines temporary cell permeability from electroporation with a high influx of calcium intracellularly resulting in cancer cell necrosis. Methods A phase I trial performing calcium electroporation on 6 patients suffering from recurrent head and neck cancer. In general anesthesia, intratumoral calcium injections were followed by electroporation. Safety was monitored by adverse events registration, serum Ca2+, ECG, and pain scores. Tumor response was measured on PET/MRI scans. Results Procedures were performed without complications. No serious adverse events, signs of hypercalcemia, or cardiac arrhythmias were observed. Two months post‐treatment tumor responses on MRI: three partial responses, one stable disease, and two progression. Responses on PET: one partial metabolic disease, four with stable metabolic disease, and one not evaluable. One patient was without clinical evidence of disease after 12 months of observation. Conclusion Calcium electroporation is feasible and safe in head and neck tumors. Clinical responses were observed in three of six patients, warranting further studies. Level of Evidence Level 4
Collapse
Affiliation(s)
- Christina Caroline Plaschke
- Department of Otorhinolaryngology, Head & Neck Surgery and Audiology Copenhagen University Hospital Rigshospitalet Copenhagen Denmark
| | - Julie Gehl
- Department of Clinical Medicine Copenhagen University Hospital Rigshospitalet Copenhagen Denmark.,Department of Clinical Oncology and Palliative Care, Center for Experimental Drug and Gene Electrotransfer (CEDGE) Zealand University Hospital Roskilde Denmark.,Department of Oncology Herlev and Gentofte Hospital, University of Copenhagen Herlev Denmark
| | - Helle Hjorth Johannesen
- Department of Clinical Physiology, Nuclear Medicine & PET, and Cluster for Molecular Imaging Copenhagen University Hospital Rigshospitalet Copenhagen Denmark
| | - Barbara Malene Fischer
- Department of Clinical Medicine Copenhagen University Hospital Rigshospitalet Copenhagen Denmark
| | - Andreas Kjaer
- Department of Clinical Medicine Copenhagen University Hospital Rigshospitalet Copenhagen Denmark
| | - Anne Fog Lomholt
- Department of Otorhinolaryngology, Head & Neck Surgery and Audiology Copenhagen University Hospital Rigshospitalet Copenhagen Denmark
| | - Irene Wessel
- Department of Otorhinolaryngology, Head & Neck Surgery and Audiology Copenhagen University Hospital Rigshospitalet Copenhagen Denmark
| |
Collapse
|
37
|
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
|
38
|
Calabrò E, Magazù S. Resonant interaction between electromagnetic fields and proteins: A possible starting point for the treatment of cancer. Electromagn Biol Med 2018; 37:155-168. [DOI: 10.1080/15368378.2018.1499031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Emanuele Calabrò
- Department of Mathematical and Informatics Sciences, Physical Sciences and Earth Sciences of Messina University, Messina, Italy
- CISFA - Interuniversity Consortium of Applied Physical Sciences (Consorzio Interuniversitario di Scienze Fisiche Applicate), Messina, Italy
| | - Salvatore Magazù
- Department of Mathematical and Informatics Sciences, Physical Sciences and Earth Sciences of Messina University, Messina, Italy
- Le Studium, Loire Valley Institute for Advanced Studies, Orléans & Tours, Orléans, France
- Centre de Biophysique Moleculaire (CBM), rue Charles Sadron, Laboratoire Interfaces, Confinement, Matériaux et Nanostructures (ICMN) – UMR 7374 CNRS, Université d’Orléans, Orleans, France
- Istituto Nazionale di Alta Matematica “F. Severi” – INDAM – Gruppo Nazionale per la Fisica Matematica – GNFM, Rome, Italy
| |
Collapse
|
39
|
Abstract
Calcium electroporation (CaEP) is a novel anti-tumour treatment that induces cell death by internalization of large quantities of calcium. The anti-tumour effectiveness of CaEP has been demonstrated in vitro, in vivo, and in preliminary clinical trials; however, its effects on the vasculature have not been previously investigated. Using a dorsal window chamber tumour model, we observed that CaEP affected to the same degree normal and tumour blood vessels in vivo, as it disrupted the vessels and caused tumour eradication by necrosis. In all cases, the effect was more pronounced in small vessels, similar to electrochemotherapy (ECT) with bleomycin. In vitro studies in four different cell lines (the B16F1 melanoma, HUVEC endothelial, FADU squamous cell carcinoma, and CHO cell lines) confirmed that CaEP causes necrosis associated with acute and severe ATP depletion, a picture different from bleomycin with electroporation. Furthermore, CaEP considerably inhibited cell migratory capabilities of endothelial cells and their potential to form capillary-like structures. The finding that CaEP has anti-vascular effects and inhibits cell migration capabilities may contribute to the explanation of the high efficacy observed in preclinical and clinical studies.
Collapse
|
40
|
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: 113] [Impact Index Per Article: 16.1] [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]
|
41
|
The second phase of bipolar, nanosecond-range electric pulses determines the electroporation efficiency. Bioelectrochemistry 2018; 122:123-133. [PMID: 29627664 DOI: 10.1016/j.bioelechem.2018.03.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/21/2018] [Accepted: 03/28/2018] [Indexed: 12/31/2022]
Abstract
Bipolar cancellation refers to a phenomenon when applying a second electric pulse reduces ("cancels") cell membrane damage by a preceding electric pulse of the opposite polarity. Bipolar cancellation is a reason why bipolar nanosecond electric pulses (nsEP) cause weaker electroporation than just a single unipolar phase of the same pulse. This study was undertaken to explore the dependence of bipolar cancellation on nsEP parameters, with emphasis on the amplitude ratio of two opposite polarity phases of a bipolar pulse. Individual cells (CHO, U937, or adult mouse ventricular cardiomyocytes (VCM)) were exposed to either uni- or bipolar trapezoidal nsEP, or to nanosecond electric field oscillations (NEFO). The membrane injury was evaluated by time-lapse confocal imaging of the uptake of propidium (Pr) or YO-PRO-1 (YP) dyes and by phosphatidylserine (PS) externalization. Within studied limits, bipolar cancellation showed little or no dependence on the electric field intensity, pulse repetition rate, chosen endpoint, or cell type. However, cancellation could increase for larger pulse numbers and/or for longer pulses. The sole most critical parameter which determines bipolar cancellation was the phase ratio: maximum cancellation was observed with the 2nd phase of about 50% of the first one, whereas a larger 2nd phase could add a damaging effect of its own. "Swapping" the two phases, i.e., delivering the smaller phase before the larger one, reduced or eliminated cancellation. These findings are discussed in the context of hypothetical mechanisms of bipolar cancellation and electroporation by nsEP.
Collapse
|
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
|
Semenov I, Casciola M, Ibey BL, Xiao S, Pakhomov AG. Electropermeabilization of cells by closely spaced paired nanosecond-range pulses. Bioelectrochemistry 2018; 121:135-141. [PMID: 29413863 DOI: 10.1016/j.bioelechem.2018.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/26/2018] [Accepted: 01/26/2018] [Indexed: 01/10/2023]
Abstract
Decreasing the time gap between two identical electric pulses is expected to render bioeffects similar to those of a single pulse of equivalent total duration. In this study, we show that it is not necessarily true, and that the effects vary for different permeabilization markers. We exposed individual CHO or NG108 cells to one 300-ns pulse (3.7-11.6 kV/cm), or a pair of such pulses (0.4-1000 μs interval), or to a single 600-ns pulse of the same amplitude. Electropermeabilization was evaluated (a) by the uptake of YO-PRO-1 (YP) dye; (b) by the amplitude of elicited Ca2+ transients, and (c) by the entry of Tl+ ions. For YP uptake, applying a 600-ns pulse or a pair of 300-ns pulses doubled the effect of a single 300-ns pulse; this additive effect did not depend on the time interval between pulses or the electric field, indicating that already permeabilized cells are as susceptible to electropermeabilization as naïve cells. In contrast, Ca2+ transients and Tl+ uptake increased in a supra-additive fashion when two pulses were delivered instead of one. Paired pulses at 3.7 kV/cm with minimal separation (0.4 and 1 μs) elicited 50-100% larger Ca2+ transients than either a single 600-ns pulse or paired pulses with longer separation (10-1000 μs). This paradoxically high efficiency of the closest spaced pulses was emphasized when Ca2+ transients were elicited in a Ca2+-free solution (when the endoplasmic reticulum (ER) was the sole significant source of Ca2+), but was eliminated by Ca2+ depletion from the ER and was not observed for Tl+ entry through the electropermeabilized membrane. We conclude that closely spaced paired pulses specifically target ER, by either permeabilizing it to a greater extent than a single double-duration pulse thus causing more Ca2+ leak, or by amplifying Ca2+-induced Ca2+ release by an unknown mechanism.
Collapse
Affiliation(s)
- Iurii Semenov
- 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
| | - Bennet L Ibey
- Radio Frequency Bioeffects Branch, Air Force Research Laboratories, Ft. Sam Houston, San Antonio, TX, 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
| | - Andrei G Pakhomov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA.
| |
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
|
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
|
46
|
Ciobanu F, Golzio M, Kovacs E, Teissié J. Control by Low Levels of Calcium of Mammalian Cell Membrane Electropermeabilization. J Membr Biol 2017; 251:221-228. [PMID: 28823021 DOI: 10.1007/s00232-017-9981-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 08/15/2017] [Indexed: 01/12/2023]
Abstract
Electric pulses, when applied to a cell suspension, induce a reversible permeabilization of the plasma membrane. This permeabilized state is a long-lived process (minutes). The biophysical molecular mechanisms supporting the membrane reorganization associated to its permeabilization remain poorly understood. Modeling the transmembrane structures as toroidal lipidic pores cannot explain why they are long-lived and why their resealing is under the control of the ATP level. Our results describe the effect of the level of free Calcium ions. Permeabilization induces a Ca2+ burst as previously shown by imaging of cells loaded with Fluo-3. But this sharp increase is reversible even when Calcium is present at a millimolar concentration. Viability is preserved to a larger extent when submillimolar concentrations are used. The effect of calcium ions is occurring during the resealing step not during the creation of permeabilization as the same effect is observed if Ca2+ is added in the few seconds following the pulses. The resealing time is faster when Ca2+ is present in a dose-dependent manner. Mg2+ is observed to play a competitive role. These observations suggest that Ca2+ is acting not on the external leaflet of the plasma membrane but due to its increased concentration in the cytoplasm. Exocytosis will be enhanced by this Ca2+ burst (but hindered by Mg2+) and occurs in the electropermeabilized part of the cell surface. This description is supported by previous theoretical and experimental results. The associated fusion of vesicles will be the support of resealing.
Collapse
Affiliation(s)
- Florin Ciobanu
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France.,University Carol Davila, Bucarest, Romania
| | - Muriel Golzio
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Justin Teissié
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
47
|
Frandsen SK, Krüger MB, Mangalanathan UM, Tramm T, Mahmood F, Novak I, Gehl J. Normal and Malignant Cells Exhibit Differential Responses to Calcium Electroporation. Cancer Res 2017; 77:4389-4401. [DOI: 10.1158/0008-5472.can-16-1611] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/28/2016] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
|
48
|
Frandsen SK, Gehl J. Effect of calcium electroporation in combination with metformin in vivo and correlation between viability and intracellular ATP level after calcium electroporation in vitro. PLoS One 2017; 12:e0181839. [PMID: 28742810 PMCID: PMC5526525 DOI: 10.1371/journal.pone.0181839] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 07/08/2017] [Indexed: 02/07/2023] Open
Abstract
Background Calcium electroporation is a new experimental anti-cancer treatment where calcium is internalized into cells by application of short, high voltage pulses. Calcium electroporation has been shown to induce tumor necrosis associated with ATP depletion while the effect on normal fibroblasts was limited when investigated in a 3D in vitro spheroid model. We aimed to investigate the effect of calcium electroporation in combination with metformin, a drug that affects intracellular ATP level. We also aimed to study the relationship between the viability and intracellular ATP levels after calcium electroporation in vitro. Methods In this study, we investigated the effect of calcium electroporation with metformin on NMRI-Foxn1nu mice in vivo on tumor size, survival, and intracellular ATP. We further investigated viability and intracellular ATP level in vitro after calcium electroporation in two human cancer cell lines: Breast (MDA-MB231) and colon (HT29), and in normal human fibroblasts (HDF-n), as well as investigating viability in human bladder cancer cells (SW780) and human small cell lung cancer cells (H69) where we have previously published intracellular ATP levels. Results Calcium electroporation significantly reduced the size and ATP level of bladder cancer tumors treated in vivo but no increased effect of metformin combined with calcium electroporation was shown on neither tumor size, survival, nor ATP level. Calcium electroporation in vitro significantly decreased viability compared with calcium alone (p<0.0001 for calcium concentrations from 0.5 mM for H69, HDF-n, and MDA-MB231; p<0.0001 for calcium concentrations from 1 mM for HT29 and SW780). Intracellular ATP levels decreased significantly after calcium electroporation (p<0.05), however no correlation between intracellular ATP level and viability after treatment was observed. Conclusion Calcium electroporation caused reduced tumor size, increased survival, and acute ATP depletion in vivo. This effect was not augmented by metformin. Calcium electroporation is a possible novel anti-cancer treatment that has been shown to cause cell death associated with acute ATP depletion in vitro and in vivo.
Collapse
Affiliation(s)
- Stine Krog Frandsen
- Center for Experimental Drug and Gene Electrotransfer, Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Julie Gehl
- Center for Experimental Drug and Gene Electrotransfer, Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
- * E-mail:
| |
Collapse
|
49
|
Delivery devices for exposure of biological cells to nanosecond pulsed electric fields. Med Biol Eng Comput 2017; 56:85-97. [PMID: 28674780 DOI: 10.1007/s11517-017-1676-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 06/25/2017] [Indexed: 12/11/2022]
Abstract
In this paper, delivery devices for nanosecond pulsed electric field exposure of biological samples in direct contact with electrodes or isolated are presented and characterized. They are based on a modified electroporation cuvette and two transverse electromagnetic cells (TEM cells). The devices were used to apply pulses with high intensity (4.5 kV) and short durations (3 and 13 ns). The delivery devices were electromagnetically characterized in the frequency and time domains. Field intensities of around 5, 0.5, and 12 MV m-1 were obtained by numerical simulations of the biological sample positioned in the three delivery devices. Two delivery systems had a homogenous electric field spatial distribution, and one was adapted to permit a highly localized exposure in the vicinity of a needle. Experimental biological investigations were carried out at different field intensities for five cancer cell lines. The results using flow cytometry showed that cells kept polarized mitochondrial membrane but lost plasma membrane integrity following a dose-response trend after exposure to different electric field intensities. Certain cell types (U87, MCF7) showed higher sensitivities to nsPEFs than other lines tested.
Collapse
|
50
|
Morotomi-Yano K, Yano KI. Calcium-dependent activation of transglutaminase 2 by nanosecond pulsed electric fields. FEBS Open Bio 2017; 7:934-943. [PMID: 28680807 PMCID: PMC5494297 DOI: 10.1002/2211-5463.12227] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022] Open
Abstract
Exposure of cultured human cells to nanosecond pulsed electric fields (nsPEFs) elicits various cellular events, including Ca2+ influx and cell death. Recently, nsPEFs have been regarded as a novel physical treatment useful for biology and medicine, but the underlying mechanism of action remains to be fully elucidated. In this study, we investigated the effect of nsPEFs on transglutaminases (TGs), enzymes that catalyze covalent protein modifications such as protein-protein crosslinking. Cellular TG activity was monitored by conjugation of cellular proteins with biotin-cadaverine, a cell-permeable pseudosubstrate for TGs. We applied nsPEFs to HeLa S3 cells and found that overall catalytic activity of cellular TGs was greatly increased in a Ca2+-dependent manner. The Ca2+ ionophore ionomycin significantly augmented nsPEF-induced TG activation, further supporting the importance of Ca2+. Among human TG family members, TG2 is known to be the most ubiquitously expressed, and its catalytic activity requires elevated intracellular Ca2+. Given the requirement of Ca2+ for TG activation by nsPEFs, we performed depletion of TG2 by RNA interference (RNAi). We observed that TG2 RNAi suppressed the nsPEF-induced TG activation and partially alleviated the cytotoxic effects of nsPEFs. These findings demonstrate that TG2 activation is a Ca2+-dependent event in nsPEF-exposed cells and exerts negative effects on cell physiology.
Collapse
Affiliation(s)
- Keiko Morotomi-Yano
- Department of Bioelectrics Institute of Pulsed Power Science Kumamoto University Japan
| | - Ken-Ichi Yano
- Department of Bioelectrics Institute of Pulsed Power Science Kumamoto University Japan
| |
Collapse
|