Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 695: 149408, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38157631

RESUMEN

With over 50 years of electroporation research, the nature of cell membrane permeabilization remains elusive. The lifetime of electropores in molecular models is limited to nano- or microseconds, whereas the permeabilization of electroporated cells can last minutes. This study aimed at resolving a longstanding debate on whether the prolonged permeabilization is due to the formation of long-lived pores in cells. We developed a method for dynamic monitoring and conductance measurements of individual electropores. This was accomplished by time-lapse total internal reflection fluorescence (TIRF) imaging in HEK cells loaded with CAL-520 dye and placed on an indium tin oxide (ITO) surface. Applying a 1-ms, 0 to -400 mV pulse between the patch pipette and ITO evoked focal Ca2+ transients that identified individual electropores. Some transients disappeared in milliseconds but others persisted for over a minute. Persistent transients ("Ca2+ plumes") faded over time to a stable or a randomly fluctuating level that could include periods of full quiescence. Single pore conductance, measured by 0 to -50 mV, 50 ms steps at 30 and 60 s after the electroporation, ranged from 80 to 200 pS. These experiments proved electropore longevity in cells, in stark contrast to molecular simulations and many findings in lipid bilayers.


Asunto(s)
Membrana Dobles de Lípidos , Longevidad , Membrana Dobles de Lípidos/metabolismo , Membrana Celular/metabolismo , Electroporación/métodos , División Celular
2.
Biochem Biophys Res Commun ; 677: 93-97, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37566922

RESUMEN

This study explored the role of the Na/K-ATPase (NKA) in membrane permeabilization induced by nanosecond electric pulses. Using CRISPR/Cas9 and shRNA, we silenced the ATP1A1 gene, which encodes α1 NKA subunit in U937 human monocytes. Silencing reduced the rate and the cumulative uptake of YoPro-1 dye after electroporation by 300-ns, 7-10 kV/cm pulses, while ouabain, a specific NKA inhibitor, enhanced YoPro-1 entry. We conclude that the α1 subunit supports the electropermeabilized membrane state, by forming or stabilizing electropores or by hindering repair mechanisms, and this role is independent of NKA's ion pump function.


Asunto(s)
Electricidad , Electroporación , Humanos , Permeabilidad de la Membrana Celular , Membrana Celular/metabolismo , ARN Interferente Pequeño/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
3.
Int J Mol Sci ; 24(11)2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37298142

RESUMEN

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.


Asunto(s)
Electricidad , Electroporación , Cricetinae , Animales , Humanos , Cricetulus , Permeabilidad de la Membrana Celular , Membrana Celular/metabolismo , Transporte Biológico
4.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36769172

RESUMEN

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.


Asunto(s)
Carcinoma , Neoplasias Esofágicas , Humanos , Electricidad , Potenciales de la Membrana , Electroporación , Músculo Liso , Neoplasias Esofágicas/terapia
5.
Int J Mol Sci ; 24(13)2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-37446096

RESUMEN

Reversing the pulse polarity, i.e., changing the electric field direction by 180°, inhibits electroporation and electrostimulation by nanosecond electric pulses (nsEPs). This feature, known as "bipolar cancellation," enables selective remote targeting with nsEPs and reduces the neuromuscular side effects of ablation therapies. We analyzed the biophysical mechanisms and measured how cancellation weakens and is replaced by facilitation when nsEPs are applied from different directions at angles from 0 to 180°. Monolayers of endothelial cells were electroporated by a train of five pulses (600 ns) or five paired pulses (600 + 600 ns) applied at 1 Hz or 833 kHz. Reversing the electric field in the pairs (180° direction change) caused 2-fold (1 Hz) or 20-fold (833 kHz) weaker electroporation than the train of single nsEPs. Reducing the angle between pulse directions in the pairs weakened cancellation and replaced it with facilitation at angles <160° (1 Hz) and <130° (833 kHz). Facilitation plateaued at about three-fold stronger electroporation compared to single pulses at 90-100° angle for both nsEP frequencies. The profound dependence of the efficiency on the angle enables novel protocols for highly selective focal electroporation at one electrode in a three-electrode array while avoiding effects at the other electrodes. Nanosecond-resolution imaging of cell membrane potential was used to link the selectivity to charging kinetics by co- and counter-directional nsEPs.


Asunto(s)
Electroporación , Células Endoteliales , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Electroporación/métodos , Terapia de Electroporación
6.
Int J Mol Sci ; 22(13)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34208945

RESUMEN

Intense pulsed electric fields (PEF) are a novel modality for the efficient and targeted ablation of tumors by electroporation. The major adverse side effects of PEF therapies are strong involuntary muscle contractions and pain. Nanosecond-range PEF (nsPEF) are less efficient at neurostimulation and can be employed to minimize such side effects. We quantified the impact of the electrode configuration, PEF strength (up to 20 kV/cm), repetition rate (up to 3 MHz), bi- and triphasic pulse shapes, and pulse duration (down to 10 ns) on eliciting compound action potentials (CAPs) in nerve fibers. The excitation thresholds for single unipolar but not bipolar stimuli followed the classic strength-duration dependence. The addition of the opposite polarity phase for nsPEF increased the excitation threshold, with symmetrical bipolar nsPEF being the least efficient. Stimulation by nsPEF bursts decreased the excitation threshold as a power function above a critical duty cycle of 0.1%. The threshold reduction was much weaker for symmetrical bipolar nsPEF. Supramaximal stimulation by high-rate nsPEF bursts elicited only a single CAP as long as the burst duration did not exceed the nerve refractory period. Such brief bursts of bipolar nsPEF could be the best choice to minimize neuromuscular stimulation in ablation therapies.


Asunto(s)
Electroporación/instrumentación , Fibras Nerviosas/fisiología , Potenciales de Acción , Animales , Anuros , Técnicas Electroquímicas , Electrodos
7.
Cell Mol Life Sci ; 76(22): 4539-4550, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31055644

RESUMEN

The initiation of action potentials (APs) by membrane depolarization occurs after a brief vulnerability period, during which excitation can be abolished by the reversal of the stimulus polarity. This vulnerability period is determined by the time needed for gating of voltage-gated sodium channels (VGSC). We compared nerve excitation by ultra-short uni- and bipolar stimuli to define the time frame of bipolar cancellation and of AP initiation. Propagating APs in isolated frog sciatic nerve were elicited by cathodic pulses (200 ns-300 µs), followed by an anodic (canceling) pulse of the same duration after a 0-200-µs delay. We found that the earliest and the latest boundaries for opening the critical number of VGSC needed to initiate AP are, respectively, between 11 and 20 µs and between 100 and 200 µs after the onset of depolarization. Stronger depolarization accelerated AP initiation, apparently due to faster VGSC opening, but not beyond the 11-µs limit. Bipolar cancellation was augmented by reducing pulse duration, shortening the delay between pulses, decreasing the amplitude of the cathodic pulse, and increasing the amplitude of the anodic one. Some of these characteristics contrasted the bipolar cancellation of cell membrane electroporation (Pakhomov et al. in Bioelectrochemistry 122:123-133, 2018; Gianulis et al. in Bioelectrochemistry 119:10-19, 2017), suggesting different mechanisms. The ratio of nerve excitation thresholds for a unipolar cathodic pulse and a symmetrical bipolar pulse increased as a power function as the pulse duration decreased, in remarkable agreement with the predictions of SENN model of nerve excitation (Reilly and Diamant in Health Phys 83(3):356-365, 2002).


Asunto(s)
Potenciales de Acción/fisiología , Activación del Canal Iónico/fisiología , Sistema Nervioso/metabolismo , Canales de Sodio/metabolismo , Animales , Anuros/metabolismo , Anuros/fisiología , Membrana Celular/metabolismo , Membrana Celular/fisiología , Permeabilidad de la Membrana Celular/fisiología , Electroporación/métodos , Femenino
8.
Int J Mol Sci ; 21(9)2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403282

RESUMEN

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.


Asunto(s)
Bario/metabolismo , Calcio/metabolismo , Permeabilidad de la Membrana Celular , Membrana Celular/metabolismo , Animales , Células CHO , Cationes/metabolismo , Línea Celular , Cricetinae , Cricetulus , Citosol/metabolismo , Electroporación/métodos , Colorantes Fluorescentes/química , Células HEK293 , Humanos , Microscopía Fluorescente/métodos , Nanotecnología/métodos , Imagen de Lapso de Tiempo/métodos
9.
Biochem Biophys Res Commun ; 518(4): 759-764, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31472962

RESUMEN

Intense nanosecond pulsed electric field (nsPEF) is a novel modality for cell activation and nanoelectroporation. Applications of nsPEF in research and therapy are hindered by a high electric field requirement, typically from 1 to over 50 kV/cm to elicit any bioeffects. We show how this requirement can be overcome by engaging temporal summation when pulses are compressed into high-rate bursts (up to several MHz). This approach was tested for excitation of ventricular cardiomyocytes and peripheral nerve fibers; for membrane electroporation of cardiomyocytes, CHO, and HEK cells; and for killing EL-4 cells. MHz compression of nsPEF bursts (100-1000 pulses) enables excitation at only 0.01-0.15 kV/cm and electroporation already at 0.4-0.6 kV/cm. Clear separation of excitation and electroporation thresholds allows for multiple excitation cycles without membrane disruption. The efficiency of nsPEF bursts increases with the duty cycle (by increasing either pulse duration or repetition rate) and with increasing the total time "on" (by increasing either pulse duration or number). For some endpoints, the efficiency of nsPEF bursts matches a single "long" pulse whose amplitude and duration equal the time-average amplitude and duration of the bursts. For other endpoints this rule is not valid, presumably because of nsPEF-specific bioeffects and/or possible modification of targets already during the burst. MHz compression of nsPEF bursts is a universal and efficient way to lower excitation thresholds and facilitate electroporation.


Asunto(s)
Potenciales de Acción/fisiología , Permeabilidad de la Membrana Celular/fisiología , Electroporación/métodos , Miocitos Cardíacos/fisiología , Fibras Nerviosas/fisiología , Animales , Células CHO , Calcio , Línea Celular Tumoral , Células Cultivadas , Cricetulus , Estimulación Eléctrica/métodos , Células HEK293 , Humanos , Ratones Endogámicos DBA , Miocitos Cardíacos/citología , Rana catesbeiana/fisiología , Factores de Tiempo
10.
J Cardiovasc Electrophysiol ; 30(3): 392-401, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30582656

RESUMEN

INTRODUCTION: Opening of voltage-gated sodium channels takes tens to hundreds of microseconds, and mechanisms of their opening by nanosecond pulsed electric field (nsPEF) stimuli remain elusive. This study was aimed at uncovering the mechanisms of how nsPEF elicits action potentials (APs) in cardiomyocytes. METHODS AND RESULTS: Fluorescent imaging of optical APs (FluoVolt) and Ca2+ -transients (Fluo-4) was performed in enzymatically isolated murine ventricular cardiomyocytes stimulated by 200-nanosecond trapezoidal pulses. nsPEF stimulation evoked tetrodotoxin-sensitive APs accompanied or preceded by slow sustained depolarization (SSD) and, in most cells, by transient afterdepolarization waves. SSD threshold was lower than the AP threshold (1.26 ± 0.03 vs 1.34 ± 0.03 kV/cm, respectively, P < 0.001). Inhibition of l-type calcium and sodium-calcium exchanger currents reduced the SSD amplitude and increased the AP threshold ( P < 0.05). The threshold for Ca 2+ -transients (1.40 ± 0.04 kV/cm) was not significantly affected by a tetrodotoxin-verapamil cocktail, suggesting the activation of a Ca 2+ entry pathway independent from the opening of Na + or Ca 2+ voltage-gated channels. Removal of external Ca 2+ decreased the SSD amplitude ( P = 0.004) and blocked Ca 2+ -transients but not APs. The incidence of transient afterdepolarization waves was decreased by verapamil and by removal of external Ca 2+ ( P = 0.002). CONCLUSIONS: The study established that nsPEF stimulation caused calcium entry into cardiac myocytes (including routes other than voltage-gated calcium channels) and SSD. Tetrodotoxin-sensitive APs were mediated by SSD, whose amplitude depended on the calcium entry. Plasma membrane electroporation was the most likely primary mechanism of SSD with additional contribution from l-type calcium and sodium-calcium exchanger currents.


Asunto(s)
Potenciales de Acción , Señalización del Calcio , Calcio/metabolismo , Estimulación Eléctrica , Miocitos Cardíacos/metabolismo , Sodio/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Ratones Endogámicos DBA , Intercambiador de Sodio-Calcio/metabolismo , Factores de Tiempo
11.
J Biol Chem ; 292(47): 19381-19391, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-28982976

RESUMEN

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.


Asunto(s)
Anoctaminas/metabolismo , Apoptosis/efectos de la radiación , Calcio/metabolismo , Membrana Celular/fisiología , Electricidad , Nanotecnología , Fosfatidilserinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Relación Dosis-Respuesta en la Radiación , Células HEK293 , Humanos , Canales Iónicos/metabolismo
12.
Cell Mol Life Sci ; 74(9): 1741-1754, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27986976

RESUMEN

Tumor ablation by nanosecond pulsed electric fields (nsPEF) is an emerging therapeutic modality. We compared nsPEF cytotoxicity for human cell lines of cancerous (IMR-32, Hep G2, HT-1080, and HPAF-II) and non-cancerous origin (BJ and MRC-5) under strictly controlled and identical conditions. Adherent cells were uniformly treated by 300-ns PEF (0-2000 pulses, 1.8 kV/cm, 50 Hz) on indium tin oxide-covered glass coverslips, using the same media and serum. Cell survival plotted against the number of pulses displayed three distinct regions (initial resistivity, logarithmic survival decline, and residual resistivity) for all tested cell types, but with differences in LD50 spanning as much as nearly 80-fold. The non-cancerous cells were less sensitive than IMR-32 neuroblastoma cells but more vulnerable than the other cancers tested. The cytotoxic efficiency showed no apparent correlation with cell or nuclear size, cell morphology, metabolism level, or the extent of membrane disruption by nsPEF. Increasing pulse duration to 9 µs (0.75 kV/cm, 5 Hz) produced a different selectivity pattern, suggesting that manipulation of PEF parameters can, at least for certain cancers, overcome their resistance to nsPEF ablation. Identifying mechanisms and cell markers of differential nsPEF susceptibility will critically contribute to the proper choice and outcome of nsPEF ablation therapies.


Asunto(s)
Electricidad , Electroporación/métodos , Muerte Celular , Línea Celular , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Núcleo Celular/metabolismo , Proliferación Celular , Forma de la Célula , Tamaño de la Célula , Supervivencia Celular , Humanos , Factores de Tiempo
13.
Biochim Biophys Acta Biomembr ; 1859(7): 1273-1281, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28432032

RESUMEN

Electric field pulses of nano- and picosecond duration are a novel modality for neurostimulation, activation of Ca2+ signaling, and tissue ablation. However it is not known how such brief pulses activate voltage-gated ion channels. We studied excitation and electroporation of hippocampal neurons by 200-ns pulsed electric field (nsPEF), by means of time-lapse imaging of the optical membrane potential (OMP) with FluoVolt dye. Electroporation abruptly shifted OMP to a more depolarized level, which was reached within <1ms. The OMP recovery started rapidly (τ=8-12ms) but gradually slowed down (to τ>10s), so cells remained above the resting OMP level for at least 20-30s. Activation of voltage-gated sodium channels (VGSC) enhanced the depolarizing effect of electroporation, resulting in an additional tetrodotoxin-sensitive OMP peak in 4-5ms after nsPEF. Omitting Ca2+ in the extracellular solution did not reduce the depolarization, suggesting no contribution of voltage-gated calcium channels (VGCC). In 40% of neurons, nsPEF triggered a single action potential (AP), with the median threshold of 3kV/cm (range: 1.9-4kV/cm); no APs could be evoked by stimuli below the electroporation threshold (1.5-1.9kV/cm). VGSC opening could already be detected in 0.5ms after nsPEF, which is too fast to be mediated by the depolarizing effect of electroporation. The overlap of electroporation and AP thresholds does not necessarily reflect the causal relation, but suggests a low potency of nsPEF, as compared to conventional electrostimulation, for VGSC activation and AP induction.


Asunto(s)
Electricidad , Colorantes Fluorescentes/química , Potenciales de la Membrana , Neuronas/fisiología , Potenciales de Acción , Animales , Permeabilidad de la Membrana Celular , Electroporación , Óptica y Fotónica , Ratas
14.
Biochim Biophys Acta Biomembr ; 1859(7): 1282-1290, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28432034

RESUMEN

In this paper a simple prediction method for the bipolar pulse cancellation effect is proposed, based on the frequency analysis of the TMP spectra of a single cell and the computed relative global spectral content up to a defined frequency threshold. We present a spectral analysis of pulses applied in experiments, and we extract the induced TMP from a microdosimetric model of the cell. The induced TMP computation is carried out on a hemispherical multi-layered cell model in the time domain. The analysis is presented for a variety of unipolar and bipolar input signals in the nanosecond and the microsecond time scales. Our evaluations are in good agreement with experimental results for bipolar pulse cancellation of electropermeabilization-induced Ca2+ influx using 300ns, 750kV/m pulses and with other results reported in recent literature.


Asunto(s)
Permeabilidad de la Membrana Celular , Potenciales de la Membrana , Animales , Células CHO , Cricetinae , Cricetulus , Medios de Cultivo , Análisis de Fourier , Modelos Biológicos
15.
J Membr Biol ; 250(2): 217-224, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28243693

RESUMEN

Electric pulses of nanosecond duration (nsEP) are emerging as a new modality for tissue ablation. Plasma membrane permeabilization by nsEP may cause osmotic imbalance, water uptake, cell swelling, and eventual membrane rupture. The present study was aimed to increase the cytotoxicity of nsEP by fostering water uptake and cell swelling. This aim was accomplished by lowering temperature after nsEP application, which delayed the membrane resealing and/or suppressed the cell volume mechanisms. The cell diameter in U-937 monocytes exposed to a train of 50, 300-ns pulses (100 Hz, 7 kV/cm) at room temperature and then incubated on ice for 30 min increased by 5.6 +/- 0.7 µm (40-50%), which contrasted little or no changes (1 +/- 0.3 µm, <10%) if the incubation was at 37 °C. Neither this nsEP dose nor the 30-min cooling caused cell death when applied separately; however, their combination reduced cell survival to about 60% in 1.5-3 h. Isosmotic addition of a pore-impermeable solute (sucrose) to the extracellular medium blocked cell swelling and rescued the cells, thereby pointing to swelling as a primary cause of membrane rupture and cell death. Cooling after nsEP exposure can potentially be employed in medical practice to assist tissue and tumor ablation.


Asunto(s)
Frío , Electroporación , Muerte Celular/fisiología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/fisiología , Tamaño de la Célula , Supervivencia Celular/fisiología , Humanos
16.
Biochim Biophys Acta ; 1848(10 Pt A): 2118-25, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26112464

RESUMEN

Ca2+ activation and membrane electroporation by 10-ns and 4-ms electric pulses (nsEP and msEP) were compared in rat embryonic cardiomyocytes. The lowest electric field which triggered Ca2+ transients was expectedly higher for nsEP (36 kV/cm) than for msEP (0.09 kV/cm) but the respective doses were similar (190 and 460 mJ/g). At higher intensities, both stimuli triggered prolonged firing in quiescent cells. An increase of basal Ca2+ level by >10 nM in cells with blocked voltage-gated Ca2+ channels and depleted Ca2+ depot occurred at 63 kV/cm (nsEP) or 0.14 kV/cm (msEP) and was regarded as electroporation threshold. These electric field values were at 150-230% of stimulation thresholds for both msEP and nsEP, notwithstanding a 400,000-fold difference in pulse duration. For comparable levels of electroporative Ca2+ uptake, msEP caused at least 10-fold greater uptake of propidium than nsEP, suggesting increased yield of larger pores. Electroporation by msEP started Ca2+ entry abruptly and locally at the electrode-facing poles of cell, followed by a slow diffusion to the center. In a stark contrast, nsEP evoked a "supra-electroporation" pattern of slower but spatially uniform Ca2+ entry. Thus nsEP and msEP had comparable dose efficiency, but differed profoundly in the size and localization of electropores.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Electroporación/métodos , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/efectos de la radiación , Propidio/farmacocinética , Animales , Permeabilidad de la Membrana Celular/efectos de la radiación , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Tasa de Depuración Metabólica/efectos de la radiación , Dosis de Radiación , Ratas , Electricidad Estática
17.
Biochim Biophys Acta ; 1848(4): 958-66, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25585279

RESUMEN

Exposure to intense, nanosecond-duration electric pulses (nsEP) opens small but long-lived pores in the plasma membrane. We quantified the cell uptake of two membrane integrity marker dyes, YO-PRO-1 (YP) and propidium (Pr) in order to test whether the pore size is affected by the number of nsEP. The fluorescence of the dyes was calibrated against their concentrations by confocal imaging of stained homogenates of the cells. The calibrations revealed a two-phase dependence of Pr emission on the concentration (with a slower rise at<4µM) and a linear dependence for YP. CHO cells were exposed to nsEP trains (1 to 100 pulses, 60ns, 13.2kV/cm, 10Hz) with Pr and YP in the medium, and the uptake of the dyes was monitored by time-lapse imaging for 3min. Even a single nsEP triggered a modest but detectable entry of both dyes, which increased linearly when more pulses were applied. The influx of Pr per pulse was constant and independent of the pulse number. The influx of YP per pulse was highest with 1- and 2-pulse exposures, decreasing to about twice the Pr level for trains from 5 to 100 pulses. The constant YP/Pr influx ratio for trains of 5 to 100 pulses suggests that increasing the number of pulses permeabilizes cells to a greater extent by increasing the pore number and not the pore diameter.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Membrana Celular/metabolismo , Estimulación Eléctrica/métodos , Campos Electromagnéticos , Electroporación/métodos , Nanoporos , Animales , Benzoxazoles/química , Células CHO , Membrana Celular/efectos de la radiación , Permeabilidad de la Membrana Celular/efectos de la radiación , Cricetinae , Cricetulus , Propidio/química , Pulso Arterial , Compuestos de Quinolinio/química , Imagen de Lapso de Tiempo
18.
Biochim Biophys Acta ; 1838(10): 2547-54, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24978108

RESUMEN

Opening of long-lived pores in the cell membrane is the principal primary effect of intense, nanosecond pulsed electric field (nsPEF). Here we demonstrate that the evolution of pores, cell survival, the time and the mode of cell death (necrotic or apoptotic) are determined by the level of external Ca(2+) after nsPEF. We also introduce a novel, minimally disruptive technique for nsEP exposure of adherent cells on indium tin oxide (ITO)-coated glass coverslips, which does not require cell detachment and enables fast exchanges of bath media. Increasing the Ca(2+) level from the nominal 2-5µM to 2mM for the first 60-90min after permeabilization by 300-nsPEF increased the early (necrotic) death in U937, CHO, and BPAE cells. With nominal Ca(2+), the inhibition of osmotic swelling rescued cells from the early necrosis and increased caspase 3/7 activation later on. However, the inhibition of swelling had a modest or no protective effect with 2mM Ca(2+) in the medium. With the nominal Ca(2+), most cells displayed gradual increase in YO-PRO-1 and propidium (Pr) uptake. With 2mM Ca(2+), the initially lower Pr uptake was eventually replaced by a massive and abrupt Pr entry (necrotic death). It was accompanied by a transient acceleration of the growth of membrane blebs due to the increase of the intracellular osmotic pressure. We conclude that the high-Ca(2+)-dependent necrotic death in nsPEF-treated cells is effected by a delayed, sudden, and osmotically-independent pore expansion (or de novo formation of larger pores), but not by the membrane rupture.


Asunto(s)
Calcio/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Electroporación , Presión Osmótica , Animales , Células CHO , Bovinos , Cricetinae , Cricetulus , Humanos , Necrosis/metabolismo , Células U937
19.
J Membr Biol ; 248(5): 837-47, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25796485

RESUMEN

Non-thermal probing and stimulation with subnanosecond electric pulses and terahertz electromagnetic radiation may lead to new, minimally invasive diagnostic and therapeutic procedures and to methods for remote monitoring and analysis of biological systems, including plants, animals, and humans. To effectively engineer these still-emerging tools, we need an understanding of the biophysical mechanisms underlying the responses that have been reported to these novel stimuli. We show here that subnanosecond (≤500 ps) electric pulses induce action potentials in neurons and cause calcium transients in neuroblastoma-glioma hybrid cells, and we report complementary molecular dynamics simulations of phospholipid bilayers in electric fields in which membrane permeabilization occurs in less than 1 ns. Water dipoles in the interior of these model membranes respond in less than 1 ps to permeabilizing electric potentials by aligning in the direction of the field, and they re-orient at terahertz frequencies to field reversals. The mechanism for subnanosecond lipid electropore formation is similar to that observed on longer time scales-energy-minimizing intrusions of interfacial water into the membrane interior and subsequent reorganization of the bilayer into hydrophilic, conductive structures.


Asunto(s)
Membrana Celular/química , Electroporación/métodos , Glioma/patología , Membrana Dobles de Lípidos/química , Neuroblastoma/patología , Neuronas/fisiología , Agua/química , Animales , Calcio/metabolismo , Campos Electromagnéticos , Simulación de Dinámica Molecular , Fosfolípidos/química , Ratas , Células Tumorales Cultivadas
20.
Arch Biochem Biophys ; 570: 1-7, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25707556

RESUMEN

Lanthanide ions are the only known blockers of permeabilization by electric pulses of nanosecond duration (nsEP), but the underlying mechanisms are unknown. We employed timed applications of Gd(3+) before or after nsEP (600-ns, 20 kV/cm) to investigate the mechanism of inhibition, and measured the uptake of the membrane-impermeable YO-PRO-1 (YP) and propidium (Pr) dyes. Gd(3+) inhibited dye uptake in a concentration-dependent manner. The inhibition of Pr uptake was always about 2-fold stronger. Gd(3+) was effective when added after nsEP, as well as when it was present during nsEP exposure and removed afterward. Pores formed by nsEP in the presence of Gd(3+) remained quiescent unless Gd(3+) was promptly washed away. Such pores resealed (or shrunk) shortly after the wash despite the absence of Gd(3+). Finally, a brief (3s) Gd(3+) perfusion was equally potent at inhibiting dye uptake when performed either immediately before or after nsEP, or early before nsEP. The persistent protective effect of Gd(3+) even in its absence proves that inhibition by Gd(3+) does not result from simple pore obstruction. Instead, Gd(3+) causes lasting modification of the membrane, occurring promptly and irrespective of pore presence; it makes the membrane less prone to permeabilization and/or reduces the stability of electropores.


Asunto(s)
Membrana Celular/efectos de los fármacos , Gadolinio/química , Nanotecnología/métodos , Animales , Células CHO , Cationes , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Colorantes/química , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Electroquímica/métodos , Electroporación , Colorantes Fluorescentes/química , Porosidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA