Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Proc Natl Acad Sci U S A ; 112(16): 5165-70, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25848012

RESUMEN

Mycoplasma pneumoniae (Mp) infections cause tracheobronchitis and "walking" pneumonia, and are linked to asthma and other reactive airway diseases. As part of the infectious process, the bacterium expresses a 591-aa virulence factor with both mono-ADP ribosyltransferase (mART) and vacuolating activities known as Community-Acquired Respiratory Distress Syndrome Toxin (CARDS TX). CARDS TX binds to human surfactant protein A and annexin A2 on airway epithelial cells and is internalized, leading to a range of pathogenetic events. Here we present the structure of CARDS TX, a triangular molecule in which N-terminal mART and C-terminal tandem ß-trefoil domains associate to form an overall architecture distinct from other well-recognized ADP-ribosylating bacterial toxins. We demonstrate that CARDS TX binds phosphatidylcholine and sphingomyelin specifically over other membrane lipids, and that cell surface binding and internalization activities are housed within the C-terminal ß-trefoil domain. The results enhance our understanding of Mp pathogenicity and suggest a novel avenue for the development of therapies to treat Mp-associated asthma and other acute and chronic airway diseases.


Asunto(s)
Proteínas Bacterianas/química , Toxinas Bacterianas/química , Citotoxinas/química , Mycoplasma pneumoniae/metabolismo , Vacuolas/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/metabolismo , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Dominio Catalítico , Citotoxinas/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Fosfatidilcolinas/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Esfingomielinas/metabolismo , Relación Estructura-Actividad
7.
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
8.
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
9.
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
10.
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
11.
Mol Microbiol ; 93(3): 568-81, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24948331

RESUMEN

Community-acquired respiratory distress syndrome (CARDS) toxin from Mycoplasma pneumoniae is a 591-amino-acid virulence factor with ADP-ribosyltransferase (ADPRT) and vacuolating activities. It is expressed at low levels during in vitro growth and at high levels during colonization of the lung. Exposure of experimental animals to purified recombinant CARDS toxin alone is sufficient to recapitulate the cytopathology and inflammatory responses associated with M. pneumoniae infection in humans and animals. Here, by molecular modelling, serial truncations and site-directed mutagenesis, we show that the N-terminal region is essential for ADP-ribosylating activity. Also, by systematic truncation and limited proteolysis experiments we identified a portion of the C-terminal region that mediates toxin binding to mammalian cell surfaces and subsequent internalization. In addition, the C-terminal region alone induces vacuolization in a manner similar to full-length toxin. Together, these data suggest that CARDS toxin has a unique architecture with functionally separable N-terminal and C-terminal domains.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Mycoplasma pneumoniae , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/genética , Secuencias de Aminoácidos , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Células HeLa , Humanos , Modelos Moleculares , NAD/metabolismo , Estructura Terciaria de Proteína , Proteolisis , Factores de Virulencia/química , Factores de Virulencia/metabolismo
12.
Cell Mol Life Sci ; 71(22): 4431-41, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24748074

RESUMEN

Nanoelectroporation of biomembranes is an effect of high-voltage, nanosecond-duration electric pulses (nsEP). It occurs both in the plasma membrane and inside the cell, and nanoporated membranes are distinguished by ion-selective and potential-sensitive permeability. Here we report a novel phenomenon of bioeffects cancellation that puts nsEP cardinally apart from the conventional electroporation and electrostimulation by milli- and microsecond pulses. We compared the effects of 60- and 300-ns monopolar, nearly rectangular nsEP on intracellular Ca(2+) mobilization and cell survival with those of bipolar 60 + 60 and 300 + 300 ns pulses. For diverse endpoints, exposure conditions, pulse numbers (1-60), and amplitudes (15-60 kV/cm), the addition of the second phase cancelled the effects of the first phase. The overall effect of bipolar pulses was profoundly reduced, despite delivering twofold more energy. Cancellation also took place when two phases were separated into two independent nsEP of opposite polarities; it gradually tapered out as the interval between two nsEP increased, but was still present even at a 10-µs interval. The phenomenon of cancellation is unique for nsEP and has not been predicted by the equivalent circuit, transport lattice, and molecular dynamics models of electroporation. The existing paradigms of membrane permeabilization by nsEP will need to be modified. Here we discuss the possible involvement of the assisted membrane discharge, two-step oxidation of membrane phospholipids, and reverse transmembrane ion transport mechanisms. Cancellation impacts nsEP applications in cancer therapy, electrostimulation, and biotechnology, and provides new insights into effects of more complex waveforms, including pulsed electromagnetic emissions.


Asunto(s)
Polaridad Celular/fisiología , Electroporación , Nanotecnología , Animales , Células CHO , Calcio/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Cricetinae , Cricetulus , Humanos , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
13.
Biochem Biophys Res Commun ; 443(2): 568-73, 2014 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-24332942

RESUMEN

Multiple studies have shown that bipolar (BP) electric pulses in the microsecond range are more effective at permeabilizing cells while maintaining similar cell survival rates as compared to monopolar (MP) pulse equivalents. In this paper, we investigated whether the same advantage existed for BP nanosecond-pulsed electric fields (nsPEF) as compared to MP nsPEF. To study permeabilization effectiveness, MP or BP pulses were delivered to single Chinese hamster ovary (CHO) cells and the response of three dyes, Calcium Green-1, propidium iodide (PI), and FM1-43, was measured by confocal microscopy. Results show that BP pulses were less effective at increasing intracellular calcium concentration or PI uptake and cause less membrane reorganization (FM1-43) than MP pulses. Twenty-four hour survival was measured in three cell lines (Jurkat, U937, CHO) and over ten times more BP pulses were required to induce death as compared to MP pulses of similar magnitude and duration. Flow cytometry analysis of CHO cells after exposure (at 15 min) revealed that to achieve positive FITC-Annexin V and PI expression, ten times more BP pulses were required than MP pulses. Overall, unlike longer pulse exposures, BP nsPEF exposures proved far less effective at both membrane permeabilization and cell killing than MP nsPEF.


Asunto(s)
Apoptosis/efectos de la radiación , Permeabilidad de la Membrana Celular/fisiología , Permeabilidad de la Membrana Celular/efectos de la radiación , Estimulación Eléctrica/métodos , Electroporación/métodos , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta en la Radiación , Campos Electromagnéticos , Humanos , Células Jurkat , Dosis de Radiación
14.
J Cell Mol Med ; 17(1): 154-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23305510

RESUMEN

Cell permeabilization by electric pulses (EP), or electroporation, is widely used for intracellular delivery of drugs and plasmids, as well as for tumour and tissue ablation. We found that cells pre-treated with 100-µs EP develop delayed hypersensitivity to subsequent EP applications. Sensitizing B16 and CHO cells by splitting a single train of eight 100-µs EP into two trains of four EP each (with 5-min. interval) decreased the LD(50) 1.5-2 times. Sensitization profoundly enhanced the electroporation-assisted uptake of bleomycin, a cell-impermeable cytotoxic agent accepted for killing tumours by electrochemotherapy. EP exposures that were not lethal per se caused cell death in the presence of bleomycin and proportionally to its concentration. Sensitizing cells by a split-dose EP exposure increased bleomycin-mediated lethality to the same extent as a 10-fold increase in bleomycin concentration when using a single EP dose. Likewise, sensitization by a split-dose EP exposure (without changing the overall dose, pulse number, or amplitude) enhanced the electroporative uptake of propidium up to fivefold. Enhancement of the electroporative uptake appears a key mechanism of electrosensitization and may benefit electrochemotherapy and numerous applications that employ EP for cell permeabilization.


Asunto(s)
Bleomicina/metabolismo , Permeabilidad de la Membrana Celular , Citotoxinas/metabolismo , Electroporación/métodos , Propidio/metabolismo , Animales , Transporte Biológico , Bleomicina/farmacología , Células CHO , Supervivencia Celular/efectos de los fármacos , Cricetinae , Cricetulus , Citotoxinas/farmacología , Melanoma Experimental , Ratones , Propidio/farmacología
15.
Bioelectrochemistry ; 149: 108289, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36270049

RESUMEN

The quest for safe and effective ablation resulted in the development of nanosecond pulsed electric fields (nsPEF) technology for tumor treatment. For future applications of nsPEF in urothelial cancer treatment, we evaluated the effect of urine presence at the ablation site. We prepared artificial urine (AU) with compounds commonly present in the healthy human urine at physiological concentrations. We compared nsPEF cytotoxicity for cancerous (T24) and non-cancerous (SV-HUC-1) human urothelial cell lines pulsed either in the AU or in a physiological solution (PS). Cell monolayers were exposed to trains of 300-ns, 10-Hz pulses using a two-needle electrode assembly placed orthogonal to the monolayer. The assembly produced the electric field gradually weakening with the distance from the electrodes. The electric field which killed 50 % of cells (LD50) was measured by staining with propidium iodide and matching the stained area with the simulated electric field strength. nsPEF exposure in PS was more cytotoxic to cancer cells. The AU protected both healthy and cancer urothelial cells, increasing their LD50 1.4 and 1.6 times, respectively. Omitting urea from the AU reduced the LD50 for healthy and cancer urothelial cells. Testing the role of other AU components, we found that it was the high concentration of phosphates what also rendered the protective effect of the AU. Our findings suggest that the nsPEF ablation of bladder cancer will be less efficient if the bladder is filled with urine.


Asunto(s)
Electricidad , Humanos , Línea Celular , Propidio
16.
Biochim Biophys Acta ; 1808(3): 792-801, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21182825

RESUMEN

Intense nanosecond-duration electric pulses (nsEP) open stable nanopores in the cell membrane, followed by cell volume changes due to water uptake or expulsion, as regulated by the osmolality balance of pore-impermeable solutes inside and outside the cell. The size of pores opened by either fifty 60-ns EP (~13 kV/cm) or five, 600-ns EP (~6 kV/cm) in GH3 cells was estimated by isoosmotic replacement of bath NaCl with polyethylene glycols and sugars. Such replacement reduced cell swelling or resulted in transient or sustained cell shrinking in response to EP. depending on the availability of pores permeable to the test solute. Unexpectedly, solute substitutions showed that for the same integral area of pores opened by 60- and 600-ns treatments (as estimated by cell volume changes), the pore sizes were similar. However, the 600-ns exposure triggered significantly higher cell uptake of propidium. We concluded that 600-ns EP opened a greater number of larger (propidium-permeable pores), but the fraction of the larger pores in the entire pore population was insufficient to contribute to cell volume changes. For both the 60- and 600-ns exposures, cell volume changes were determined by pores smaller than 0.9 nm in diameter; however, the diameter increased with increasing the nsEP intensity.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de la radiación , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Tamaño de la Célula/efectos de la radiación , Electricidad , Animales , Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Electroporación , Hipófisis/citología , Hipófisis/efectos de los fármacos , Polietilenglicoles/farmacología , Propidio/metabolismo , Ratas , Tensoactivos/farmacología
17.
Arch Biochem Biophys ; 527(1): 55-64, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22910297

RESUMEN

Nanosecond pulsed electric field (nsPEF) is a novel modality for permeabilization of membranous structures and intracellular delivery of xenobiotics. We hypothesized that oxidative effects of nsPEF could be a separate primary mechanism responsible for bioeffects. ROS production in cultured cells and media exposed to 300-ns PEF (1-13 kV/cm) was assessed by oxidation of 2',7'-dichlorodihydrofluoresein (H(2)DCF), dihidroethidium (DHE), or Amplex Red. When a suspension of H(2)DCF-loaded cells was subjected to nsPEF, the yield of fluorescent 2',7'-dichlorofluorescein (DCF) increased proportionally to the pulse number and cell density. DCF emission increased with time after exposure in nsPEF-sensitive Jurkat cells, but remained stable in nsPEF-resistant U937 cells. In cell-free media, nsPEF facilitated the conversion of H(2)DCF into DCF. This effect was not related to heating and was reduced by catalase, but not by mannitol or superoxide dismutase. Formation of H(2)O(2) in nsPEF-treated media was confirmed by increased oxidation of Amplex Red. ROS increase within individual cells exposed to nsPEF was visualized by oxidation of DHE. We conclude that nsPEF can generate both extracellular (electrochemical) and intracellular ROS, including H(2)O(2) and possibly other species. Therefore, bioeffects of nsPEF are not limited to electropermeabilization; concurrent ROS formation may lead to cell stimulation and/or oxidative cell damage.


Asunto(s)
Permeabilidad de la Membrana Celular , Electroporación , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células CHO , Línea Celular , Membrana Celular/metabolismo , Supervivencia Celular , Sistema Libre de Células/metabolismo , Cricetinae , Electroporación/métodos , Fluoresceínas/análisis , Fluoresceínas/metabolismo , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Células Jurkat , Oxidación-Reducción
18.
Biochim Biophys Acta Biomembr ; 1864(2): 183823, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838875

RESUMEN

Electroporation, in particular with nanosecond pulses, is an efficient technique to generate nanometer-size membrane lesions without the use of toxins or other chemicals. The restoration of the membrane integrity takes minutes and is only partially dependent on [Ca2+]. We explored the impact of Ca2+ on the kinetics of membrane resealing by monitoring the entry of a YO-PRO-1 dye (YP) in BPAE and HEK cells. Ca2+ was promptly removed or added after the electric pulse (EP) by a fast-step perfusion. YP entry increased sharply after the EP and gradually slowed down following either a single- or a double-exponential function. In BPAE cells permeabilized by a single 300- or 600-ns EP at 14 kV/cm in a Ca2+-free medium, perfusion with 2 mM of external Ca2+ advanced the 90% resealing and reduced the dye uptake about twofold. Membrane restoration was accomplished by a combination of fast, Ca2+-independent resealing (τ = 13-15 s) and slow, Ca2+-dependent processes (τ ~70 s with Ca2+ and ~ 110 s or more without it). These time constants did not change when the membrane damage was doubled by increasing EP duration from 300 to 600 ns. However, injury by microsecond-range EP (300 and 600 µs) took longer to recover even when the membrane initially was less damaged, presumably because of the larger size of pores made in the membrane. Full membrane recovery was not prevented by blocking both extra- and intracellular Ca2+ (by loading cells with BAPTA or after Ca2+ depletion from the reticulum), suggesting the recruitment of unknown Ca2+-independent repair mechanisms.


Asunto(s)
Calcio/metabolismo , Permeabilidad de la Membrana Celular , Membrana Celular/fisiología , Electroporación/métodos , Riñón/metabolismo , Electricidad , Células HEK293 , Humanos , Cinética
19.
Front Cardiovasc Med ; 9: 1004024, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36620647

RESUMEN

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.

20.
Biochim Biophys Acta ; 1800(11): 1210-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20691249

RESUMEN

BACKGROUND: Nanosecond electric pulses (EP) disrupt cell membrane and organelles and cause cell death in a manner different from the conventional irreversible electroporation. We explored the cytotoxic effect of 10-ns EP (quantitation, mechanisms, efficiency, and specificity) in comparison with 300-ns, 1.8- and 9-µs EP. METHODS: Effects in Jurkat and U937 cells were characterized by survival assays, DNA electrophoresis and flow cytometry. RESULTS: 10-ns EP caused apoptotic or necrotic death within 2-20 h. Survival (S, %) followed the absorbed dose (D, J/g) as: S=alphaD((-K)), where coefficients K and alpha determined the slope and the "shoulder" of the survival curve. K was similar in all groups, whereas alpha was cell type- and pulse duration-dependent. Long pulses caused immediate propidium uptake and phosphatidylserine (PS) externalization, whereas 10-ns pulses caused PS externalization only. CONCLUSIONS: 1.8- and 9-µs EP cause cell death efficiently and indiscriminately (LD50 1-3 J/g in both cell lines); 10-ns EP are less efficient, but very selective (LD50 50-80 J/g for Jurkat and 400-500 J/g for U937); 300-ns EP show intermediate effects. Shorter EP open propidium-impermeable, small membrane pores ("nanopores"), triggering different cell death mechanisms. GENERAL SIGNIFICANCE: Nanosecond EP can selectively target certain cells in medical applications like tumor ablation.


Asunto(s)
Apoptosis/efectos de la radiación , Permeabilidad de la Membrana Celular/efectos de la radiación , Membrana Celular/patología , Membrana Celular/efectos de la radiación , Campos Electromagnéticos , Membrana Celular/metabolismo , Daño del ADN , Electroporación , Citometría de Flujo , Humanos , Células Jurkat , Orgánulos/metabolismo , Orgánulos/patología , Orgánulos/efectos de la radiación , Fosfatidilserinas/metabolismo , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA