Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
AMB Express ; 10(1): 55, 2020 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-32189137

RESUMO

Cell suspensions of Escherichia coli and Lactobacillus acidophilus were exposed to 600-ns pulsed electric fields (nsPEFs) at varying amplitudes (Low-13.5, Mid-18.5 or High-23.5 kV cm-1) and pulse numbers (0 (sham), 1, 5, 10, 100 or 1000) at a 1 hertz (Hz) repetition rate. The induced temperature rise generated at these exposure parameters, hereafter termed thermal gradient, was measured and applied independently to cell suspensions in order to differentiate inactivation triggered by electric field (E-field) from heating. Treated cell suspensions were plated and cellular inactivation was quantified by colony counts after a 24-hour (h) incubation period. Additionally, cells from both exposure conditions were incubated with various antibiotic-soaked discs to determine if nsPEF exposure would induce changes in antibiotic susceptibility. Results indicate that, for both species, the total delivered energy (amplitude, pulse number and pulse duration) determined the magnitude of cell inactivation. Specifically, for 18.5 and 23.5 kV cm-1 exposures, L. acidophilus was more sensitive to the inactivation effects of nsPEF than E. coli, however, for the 13.5 kV cm-1 exposures E. coli was more sensitive, suggesting that L. acidophilus may need to meet an E-field threshold before significant inactivation can occur. Results also indicate that antibiotic susceptibility was enhanced by multiple nsPEF exposures, as observed by increased zones of growth inhibition. Moreover, for both species, a temperature increase of ≤ 20 °C (89% of exposures) was not sufficient to significantly alter cell inactivation, whereas none of the thermal equivalent exposures were sufficient to change antibiotic susceptibility categories.

2.
Biophys J ; 116(1): 120-126, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30579565

RESUMO

Direct observation of rapid membrane potential changes is critical to understand how complex neurological systems function. This knowledge is especially important when stimulation is achieved through an external stimulus meant to mimic a naturally occurring process. To enable exploration of this dynamic space, we developed an all-optical method for observing rapid changes in membrane potential at temporal resolutions of ∼25 ns. By applying a single 600-ns electric pulse, we observed sub-microsecond, continuous membrane charging and discharging dynamics. Close agreement between the acquired results and an analytical membrane-charging model validates the utility of this technique. This tool will deepen our understanding of the role of membrane potential dynamics in the regulation of many biological and chemical processes within living systems.


Assuntos
Membrana Celular/ultraestrutura , Potenciais da Membrana , Animais , Células CHO , Membrana Celular/química , Membrana Celular/fisiologia , Cricetinae , Cricetulus , Imagem Óptica/métodos
3.
Bioelectromagnetics ; 39(6): 491-499, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29984845

RESUMO

Cell-circuit models have suggested that nanosecond pulsed electric fields (nsPEFs) can disrupt intracellular membranes including endoplasmic reticulum (ER), mitochondria, and/or nucleus thereby inducing intrinsic apoptotic pathways. Therefore, we hypothesized that the unfolded protein response (UPR) would be activated, due to the fluctuations of ionic concentrations, upon poration of the ER membrane. Quantitative real-time polymerase chain reaction was utilized to measure changes in messenger RNA (mRNA) expression of specific ER stress genes in adult human dermal fibroblast (HDFa) cells treated with tunicamycin (TM) (known ER stress inducer) and cells exposed to nsPEFs (100, 10-ns pulses at 150 kV/cm delivered at a repetition rate of 1 Hz). For HDFa cells, results showed time-dependent UPR activation to TM; however, when HDFa cells were exposed to nsPEFs, no significant changes in mRNA expression of ER stress genes, and/or caspase gene were observed. These results indicate that although cell death can be observed under these exposure parameters, it is most likely not initiated through activation of the UPR. Bioelectromagnetics. 2018;39:491-499, 2018. Published 2018. This article is a U.S. Government work and is in the public domain in the USA.


Assuntos
Campos Eletromagnéticos , Fibroblastos/metabolismo , Resposta a Proteínas não Dobradas , Linhagem Celular , Campos Eletromagnéticos/efeitos adversos , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Humanos , Íons/metabolismo , Análise em Microsséries , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Pele/citologia , Pele/metabolismo , Fatores de Tempo
4.
Bioelectromagnetics ; 39(6): 441-450, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29984850

RESUMO

Nanosecond electric pulse (nsEP) exposure generates an array of physiological effects. The extent of these effects is impacted by whether the nsEP is a unipolar (UP) or bipolar (BP) exposure. A 600 ns pulse can generate 71% more YO-PRO-1 uptake compared to a 600 ns + 600 ns pulse exposure. This observation is termed "bipolar cancellation" (BPC) because despite the BP nsEP consisting of an additional 600 ns pulse, it generates reduced membrane perturbation. BPC is achieved by varying pulse amplitudes, and symmetrical and asymmetric pulse widths. The effect appears to reverse by increasing the interphase interval between symmetric BP pulses, suggesting membrane recovery is a BPC factor. To date, the impact of the interphase interval between asymmetrical BP and other BPC-inducing symmetrical BP nsEPs has not been fully explored. Additionally, interpulse intervals beyond 50 µs have not been explored to understand the impact of time between the BP nsEP phases. Here, we surveyed different interphase intervals among symmetrical and asymmetrical BP nsEPs to monitor their impact on BPC of YO-PRO-1 uptake. We identified that a 10 microsecond (ms) interphase interval within a symmetrical 600 ns + 600 ns, and 900 ns + 900 ns pulse can resolve BPC. Furthermore, the interphase interval to resolve asymmetric BPC from a 300 ns + 900 ns pulse versus 600 ns pulse exposure is greater (<10 ms) compared to symmetrical BP nsEPs. From these findings, we extended on our conceptual model that BPC is balanced by localized charging and discharging events across the membrane. Bioelectromagnetics. 39:441-450, 2018. Published 2018. This article is a U.S. Government work and is in the public domain in the USA.


Assuntos
Membrana Celular/metabolismo , Estimulação Elétrica/métodos , Animais , Benzoxazóis/farmacocinética , Células CHO , Permeabilidade da Membrana Celular , Cricetulus , Corantes Fluorescentes/farmacocinética , Microscopia Confocal , Compostos de Quinolínio/farmacocinética , Fatores de Tempo
5.
Sci Rep ; 7(1): 16372, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29180756

RESUMO

A bipolar (BP) nanosecond electric pulse (nsEP) exposure generates reduced calcium influx compared to a unipolar (UP) nsEP. This attenuated physiological response from a BP nsEP exposure is termed "bipolar cancellation" (BPC). The predominant BP nsEP parameters that induce BPC consist of a positive polarity (↑) front pulse followed by the delivery of a negative polarity (↓) back pulse of equal voltage and width; thereby the duration is twice a UP nsEP exposure. We tested these BPC parameters, and discovered that a BP nsEP with symmetrical pulse widths is not required to generate BPC. For example, our data revealed the physiological response initiated by a ↑900 nsEP exposure can be cancelled by a second pulse that is a third of its duration.  However, we observed a complete loss of BPC from a ↑300 nsEP followed by a ↓900 nsEP exposure. Spatiotemporal analysis revealed these asymmetrical BP nsEP exposures generate distinct local YO-PRO®-1 uptake patterns across the plasma membrane. From these findings, we generated a conceptual model that suggests BPC is a phenomenon balanced by localized charging and discharging events across the membrane.

6.
Biochem Biophys Rep ; 9: 302-309, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28956017

RESUMO

BACKGROUND: Exposure of cells to very short (<1 µs) electric pulses in the megavolt/meter range have been shown to cause a multitude of effects, both physical and molecular in nature. Physically, nanosecond electrical pulses (nsEP) can cause disruption of the plasma membrane, cellular swelling, shrinking and blebbing. Molecularly, nsEP have been shown to activate signaling pathways, produce oxidative stress, stimulate hormone secretion and induce both apoptotic and necrotic death. We hypothesize that studying the genetic response of primary human dermal fibroblasts exposed to nsEP, will gain insight into the molecular mechanism(s) either activated directly by nsEP, or indirectly through electrophysiology interactions. METHODS: Microarray analysis in conjunction with quantitative real time polymerase chain reaction (qRT-PCR) was used to screen and validate genes selectively upregulated in response to nsEP exposure. RESULTS: Expression profiles of 486 genes were found to be significantly changed by nsEP exposure. 50% of the top 20 responding genes coded for proteins located in two distinct cellular locations, the plasma membrane and the nucleus. Further analysis of five of the top 20 upregulated genes indicated that the HDFa cells' response to nsEP exposure included many elements of a mechanical stress response. CONCLUSIONS: We found that several genes, some of which are mechanosensitive, were selectively upregulated due to nsEP exposure. This genetic response appears to be a primary response to the stimuli and not a secondary response to cellular swelling. GENERAL SIGNIFICANCE: This work provides strong evidence that cells exposed to nsEP interpret the insult as a mechanical stress.

7.
Opt Express ; 25(6): 6621-6643, 2017 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-28381008

RESUMO

Electric-field induced physical phenomena, such as thermal, mechanical and electrochemical dynamics, may be the driving mechanism behind bioeffects observed in mammalian cells during exposure to nanosecond-duration electric pulses (nsEP) in-vitro. Correlating a driving mechanism to a biological response requires the experimental measurement and quantification of all physical dynamics resulting from the nsEP stimulus. A passive and electromagnetic interference (EMI) immune sensor is required to resolve these dynamics in high strength electric fields. The probe beam deflection technique (PBDT) is a passive and EMI immune optical method for quantifying and imaging refractive index gradients in liquids and gases, both dynamic and static, with nanosecond temporal resolution. In this work, a probe beam deflection imaging system was designed to acquire 2-D time-lapse images of thermal/mechanical dynamics resulting from monopolar and bipolar nsEP stimulus.

8.
Biochim Biophys Acta Biomembr ; 1859(3): 438-445, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28064021

RESUMO

Previously, it was demonstrated that nanometer-sized pores (nanopores) are formed in outer cellular membranes after exposure to nanosecond electric pulses (nsEPs). We reported that plasma membrane nanoporation affects phospholipids of the cell membrane, culminating in cascading phosphoinositide phosphatidylinositol-4,5-bisphosphate (PIP2) intracellular signaling. In the current study, we show that nsEPs initiated electric field (EF) dose-dependent PIP2 hydrolysis and/or depletion from the plasma membrane. This process was confirmed using fluorescent optical probes of PIP2 hydrolysis: PLCδ-PH-EGFP and GFP-C1-PKCγ-C1a. The 50% maximum response occurs with a single 600ns pulse achieving an effective dose (ED50) of EF~8kV/cm within our model cell system. At 16.2kV/cm, the ED50 for the pulse width was 484ns. Reduction of the pulse width or EF amplitude gradually reduced the observed effect, but twenty 60ns 16.2kV/cm pulses produced an effect similar to a single 600ns pulse of the same amplitude. Propidium iodide (PI) uptake after the nsEP exposure confirmed a strong relationship between EF-induced plasma membrane impact and PIP2 depletion. These results have expanded our current knowledge of nsEPs dependent cell physiological effects, and serve as a basis for model development of new exposure standards, providing novel tools for drug independent stimulation and approaches to differential modulation of key cellular functions.


Assuntos
Eletricidade , Fosfatidilinositol 4,5-Difosfato/metabolismo , Animais , Células CHO , Cálcio/metabolismo , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Citoplasma/metabolismo , Hidrólise , Inositol 1,4,5-Trifosfato/metabolismo , Fosfolipase C delta/genética , Fosfolipase C delta/metabolismo , Fosfolipase C gama/genética , Fosfolipase C gama/metabolismo , Transdução de Sinais , Fatores de Tempo
9.
Photoacoustics ; 4(3): 91-101, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27761408

RESUMO

Optoacoustic (OA) microscopy using an all-optical system based on the probe beam deflection technique (PBDT) for detection of laser-induced acoustic signals was investigated as an alternative to conventional piezoelectric transducers. PBDT provides a number of advantages for OA microscopy including (i) efficient coupling of laser excitation energy to the samples being imaged through the probing laser beam, (ii) undistorted coupling of acoustic waves to the detector without the need for separation of the optical and acoustic paths, (iii) high sensitivity and (iv) ultrawide bandwidth. Because of the unimpeded optical path in PBDT, diffraction-limited lateral resolution can be readily achieved. The sensitivity of the current PBDT sensor of 22 µV/Pa and its noise equivalent pressure (NEP) of 11.4 Pa are comparable with these parameters of the optical micro-ring resonator and commercial piezoelectric ultrasonic transducers. Benefits of the present prototype OA microscope were demonstrated by successfully resolving micron-size details in histological sections of cardiac muscle.

10.
PLoS One ; 11(5): e0154555, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27135944

RESUMO

Nanosecond electrical pulse (nsEP) exposure activates signaling pathways, produces oxidative stress, stimulates hormone secretion, causes cell swelling and induces apoptotic and necrotic death. The underlying biophysical connection(s) between these diverse cellular reactions and nsEP has yet to be elucidated. Using global genetic analysis, we evaluated how two commonly studied cell types, U937 and Jurkat, respond to nsEP exposure. We hypothesized that by studying the genetic response of the cells following exposure, we would gain direct insight into the stresses experienced by the cell and in turn better understand the biophysical interaction taking place during the exposure. Using Ingenuity Systems software, we found genes associated with cell growth, movement and development to be significantly up-regulated in both cell types 4 h post exposure to nsEP. In agreement with our hypothesis, we also found that both cell lines exhibit significant biological changes consistent with mechanical stress induction. These results advance nsEP research by providing strong evidence that the interaction of nsEPs with cells involves mechanical stress.


Assuntos
Eletricidade , Nanotecnologia/métodos , Linhagem Celular Tumoral , Membrana Celular/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Eletroquímica , Regulação da Expressão Gênica/fisiologia , Humanos , Células Jurkat , Estresse Mecânico
11.
Bioelectromagnetics ; 37(3): 141-151, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26991689

RESUMO

Electric fields produced by advanced pulsed microwave transmitter technology now readily exceed the Institute of Electrical and Electronic Engineers (IEEE) C.95.1 peak E-field limit of 100 kV/m, highlighting a need for scientific validation of such a specific limit. Toward this goal, we exposed Jurkat Clone E-6 human lymphocyte preparations to 20 high peak power microwave (HPPM) pulses (120 ns duration) with a mean peak amplitude of 2.3 MV/m and standard deviation of 0.1 with the electric field at cells predicted to range from 0.46 to 2.7 MV/m, well in excess of current standard limit. We observed that membrane integrity and cell morphology remained unchanged 4 h after exposure and cell survival 24 h after exposure was not statistically different from sham exposure or control samples. Using flow cytometry to analyze membrane disruption and morphological changes per exposed cell, no changes were observed in HPPM-exposed samples. Current IEEE C95.1-2005 standards for pulsed radiofrequency exposure limits peak electric field to 100 kV/m for pulses shorter than 100 ms [IEEE (1995) PC95.1-Standard for Safety Levels with Respect to Human Exposure to Electric, Magnetic and Electromagnetic Fields, 0 Hz to 300 GHz, Institute of Electrical and Electronic Engineers: Piscataway, NJ, USA]. This may impose large exclusion zones that limit HPPM technology use. In this study, we offer evidence that maximum permissible exposure of 100 kV/m for peak electric field may be unnecessarily restrictive for HPPM devices. Bioelectromagnetics. 37:141-151, 2016. © 2016 Wiley Periodicals, Inc.

12.
Biochem Biophys Res Commun ; 470(1): 35-40, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26721436

RESUMO

Permeabilization of cell membranes occurs upon exposure to a threshold absorbed dose (AD) of nanosecond pulsed electric fields (nsPEF). The ultimate, physiological bioeffect of this exposure depends on the type of cultured cell and environment, indicating that cell-specific pathways and structures are stimulated. Here we investigate 10 and 600 ns duration PEF effects on Chinese hamster ovary (CHO) cell nuclei, where our hypothesis is that pulse disruption of the nuclear envelope membrane leads to observed cell death and decreased viability 24 h post-exposure. To observe short-term responses to nsPEF exposure, CHO cells have been stably transfected with two fluorescently-labeled proteins known to be sequestered for cellular chromosomal function within the nucleus - histone-2b (H2B) and proliferating cell nuclear antigen (PCNA). H2B remains associated with chromatin after nsPEF exposure, whereas PCNA leaks out of nuclei permeabilized by a threshold AD of 10 and 600 ns PEF. A downturn in 24 h viability, measured by MTT assay, is observed at the number of pulses required to induce permeabilization of the nucleus.


Assuntos
Apoptose/efeitos da radiação , Permeabilidade da Membrana Celular/fisiologia , Permeabilidade da Membrana Celular/efeitos da radiação , Eletroporação/métodos , Membrana Nuclear/fisiologia , Membrana Nuclear/efeitos da radiação , Animais , Apoptose/fisiologia , Células CHO , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Cricetinae , Cricetulus , Relação Dose-Resposta à Radiação , Campos Eletromagnéticos , Doses de Radiação
13.
Sci Rep ; 5: 15063, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26450165

RESUMO

The mechanism(s) responsible for the breakdown (nanoporation) of cell plasma membranes after nanosecond pulse (nsEP) exposure remains poorly understood. Current theories focus exclusively on the electrical field, citing electrostriction, water dipole alignment and/or electrodeformation as the primary mechanisms for pore formation. However, the delivery of a high-voltage nsEP to cells by tungsten electrodes creates a multitude of biophysical phenomena, including electrohydraulic cavitation, electrochemical interactions, thermoelastic expansion, and others. To date, very limited research has investigated non-electric phenomena occurring during nsEP exposures and their potential effect on cell nanoporation. Of primary interest is the production of acoustic shock waves during nsEP exposure, as it is known that acoustic shock waves can cause membrane poration (sonoporation). Based on these observations, our group characterized the acoustic pressure transients generated by nsEP and determined if such transients played any role in nanoporation. In this paper, we show that nsEP exposures, equivalent to those used in cellular studies, are capable of generating high-frequency (2.5 MHz), high-intensity (>13 kPa) pressure transients. Using confocal microscopy to measure cell uptake of YO-PRO®-1 (indicator of nanoporation of the plasma membrane) and changing the electrode geometry, we determined that acoustic waves alone are not responsible for poration of the membrane.


Assuntos
Permeabilidade da Membrana Celular , Membrana Celular/metabolismo , Eletroporação/instrumentação , Eletroporação/métodos , Animais , Benzoxazóis/metabolismo , Benzoxazóis/farmacocinética , Células CHO , Membrana Celular/química , Cricetinae , Cricetulus , Eletricidade , Corantes Fluorescentes/metabolismo , Corantes Fluorescentes/farmacocinética , Análise de Fourier , Microscopia Confocal , Porosidade , Pressão , Compostos de Quinolínio/metabolismo , Compostos de Quinolínio/farmacocinética , Fatores de Tempo
14.
Biochem Biophys Res Commun ; 458(2): 411-7, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25660455

RESUMO

Previous work demonstrated significant changes in cellular membranes following exposure of cells to nanosecond pulsed electric fields (nsPEF), including nanoporation and increases in intracellular calcium concentration. While it is known that nsPEF exposure can cause cell death, how cells repair and survive nsPEF-induced cellular damage is not well understood. In this paper, we investigated whether autophagy is stimulated following nsPEF exposure to repair damaged membranes, proteins, and/or organelles in a pro-survival response. We hypothesized that autophagy is activated to repair nsPEF-induced plasma membrane damage and overwhelming this compensatory mechanism results in cell death. Activation of autophagy and subsequent cell death pathways were assessed measuring toxicity, gene and protein expression of autophagy markers, and by monitoring autophagosome formation and maturation using fluorescent microscopy. Results show that autophagy is activated at subtoxic nsPEF doses, as a compensatory mechanism to repair membrane damage. However, prolonged exposure results in increased cell death and a concomitant decrease in autophagic markers. These results suggest that cells take an active role in membrane repair, through autophagy, following exposure to nsPEF.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Apoptose/efeitos da radiação , Autofagia/fisiologia , Autofagia/efeitos da radiação , Animais , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta à Radiação , Humanos , Doses de Radiação
15.
Apoptosis ; 19(12): 1755-68, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25331537

RESUMO

In this publication, we demonstrate that exposure of Jurkat and U937 cells to nanosecond pulsed electrical fields (nsPEF) can modulate the extrinsic-mediated apoptotic pathway via the Fas/CD95 death receptor. An inherent difference in survival between these two cell lines in response to 10 ns exposures has been previously reported (Jurkat being more sensitive to nsPEF than U937), but the reason for this sensitivity difference remains unknown. We found that exposure of each cell line to 100, 10 ns pulses at 50 kV/cm caused a marked increase in expression of cFLIP (extrinsic apoptosis inhibitor) in U937 and FasL (extrinsic apoptosis activator) in Jurkat, respectively. Measurement of basal expression levels revealed an inherent difference between U937 cells, having a higher expression of cFLIP, and Jurkat cells, having a higher expression of FasL. From these data, we hypothesize that the sensitivity difference between the cells to nsPEF exposure may be directly related to expression of extrinsic apoptotic regulators. To validate this hypothesis, we used siRNA to knockdown cFLAR (coding for cFLIP protein) expression in U937, and FasL expression in Jurkat and challenged them to 100, 10 ns pulses at 150 kV/cm, a typical lethal dose. We observed that U937 survival was reduced nearly 60% in the knockdown population while Jurkat survival improved ~40%. These findings support the hypothesis that cell survival following 10 ns pulse exposures depends on extrinsic apoptotic regulators. Interestingly, pretreatment of U937 with a 100-pulse, 50 kV/cm exposure (to amplify cFLAR expression) significantly reduced the lethality of a 150 kV/cm, 100-pulse exposure applied 24 h later. From these data, we conclude that the observed survival differences between cells, exposed to 10 ns pulsed electric fields, is due to inherent cell biochemistry rather than the biophysics of the exposure itself. Understanding cell sensitivity to nsPEF may provide researchers/clinicians with a predicable way to control or avoid unintended cell death during nsPEF exposure.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Eletricidade , Proteína Ligante Fas/metabolismo , Transdução de Sinais , Receptor fas/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Humanos , Células Jurkat , RNA Interferente Pequeno/genética , Células U937
16.
J Biomed Opt ; 19(5): 055005, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24825506

RESUMO

The cellular response to subtle membrane damage following exposure to nanosecond pulsed electric fields (nsPEF) is not well understood. Recent work has shown that when cells are exposed to nsPEF, ion permeable nanopores (<2 nm) are created in the plasma membrane in contrast to larger diameter pores (>2 nm) created by longer micro- and millisecond duration pulses. Nanoporation of the plasma membrane by nsPEF has been shown to cause a transient increase in intracellular calcium concentration within milliseconds after exposure. Our research objective is to determine the impact of nsPEF on calcium-dependent structural and repair systems in mammalian cells. Chinese hamster ovary (CHO-K1) cells were exposed in the presence and absence of calcium ions in the outside buffer to either 1 or 20, 600-ns duration electrical pulses at 16.2 kV/cm, and pore size was determined using propidium iodide and calcium green. Membrane organization was observed with morphological changes and increases in FM1-43 fluorescence. Migration of lysosomes, implicated in membrane repair, was followed using confocal microscopy of red fluorescent protein-tagged LAMP1. Microtubule structure was imaged using mEmerald-tubulin. We found that at high 600-ns PEF dosage, calcium-induced membrane restructuring and microtubule depolymerization coincide with interruption of membrane repair via lysosomal exocytosis.


Assuntos
Transporte Biológico/fisiologia , Transporte Biológico/efeitos da radiação , Cálcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/efeitos da radiação , Eletricidade , Animais , Células CHO , Cricetinae , Cricetulus , Corantes Fluorescentes , Lisossomos/metabolismo , Microscopia Confocal , Microtúbulos/metabolismo , Microtúbulos/efeitos da radiação , Nanotecnologia , Porosidade , Compostos de Piridínio , Compostos de Amônio Quaternário
17.
Bioelectrochemistry ; 100: 80-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24530104

RESUMO

The interaction between nsPEF-induced Ca(2+) release and nsPEF-induced phosphatidylinositol4,5-bisphosphate (PIP2) hydrolysis is not well understood. To better understand this interrelation we monitored intracellular calcium changes, in cells loaded with Calcium Green-1 AM, and generation of PIP2 hydrolysis byproducts (inositol-1,4,5-trisphosphate (IP3) and diacylglycerol (DAG)) in cells transfected with one of two fluorescent reporter genes: PLCδ-PH-EGFP or GFP-C1-PKCγ-C1a. The percentage fluorescence differences (ΔF %) after exposures were determined. Upon nsPEF impact, we found that in the absence of extracellular Ca(2+) the population of IP3 liberated during nsPEF exposure (ΔF 6%±3, n=22), is diminished compared to the response in the presence of calcium (ΔF 84%±15, n=20). The production of DAG in the absence of extracellular Ca(2+) (ΔF 29%±5, n=25), as well as in cells exposed to thapsigargin (ΔF 40%±12, n=15), was not statistically different from cells exposed in the presence of extracellular calcium (ΔF 22±6%, n=18). This finding suggests that the change in intracellular calcium concentration is not solely driving the observed response. Interestingly, the DAG produced in the absence of Ca(2+) is the strongest near the membrane regions facing the electrodes, whereas the presence of extracellular Ca(2+) leads to a whole cell response. The reported observations of Ca(2+) dynamics combined with IP3 and DAG production suggest that nsPEF may cause a direct effect on the phospholipids within the plasma membrane.


Assuntos
Eletricidade , Eletroporação/métodos , Fosfatos de Fosfatidilinositol/deficiência , Animais , Células CHO , Cálcio/metabolismo , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Diglicerídeos/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hidrólise , Espaço Intracelular/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fatores de Tempo
18.
Biochem Biophys Res Commun ; 443(2): 568-73, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24332942

RESUMO

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.


Assuntos
Apoptose/efeitos da radiação , Permeabilidade da Membrana Celular/fisiologia , Permeabilidade da Membrana Celular/efeitos da radiação , Estimulação Elétrica/métodos , Eletroporação/métodos , Animais , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta à Radiação , Campos Eletromagnéticos , Humanos , Células Jurkat , Doses de Radiação
19.
Bioelectrochemistry ; 94: 23-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23747521

RESUMO

Exposure to nanosecond pulsed electrical fields (nsPEFs) results in a myriad of observable effects in mammalian cells. While these effects are often attributed to the direct permeabilization of both the plasma and organelle membranes, the underlying mechanism(s) are not well understood. We hypothesize that nsPEF-induced membrane disturbance will initiate complex intracellular lipid signaling pathways, which ultimately lead to the observed multifarious effects. In this article, we show activation of one of these pathways--phosphoinositide signaling cascade. Here we demonstrate that nsPEF initiates phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) hydrolysis or depletion from the plasma membrane, accumulation of inositol-1,4,5-trisphosphate (IP3) in the cytoplasm and increase of diacylglycerol (DAG) on the inner surface of the plasma membrane. All of these events are initiated by a single 16.2 kV/cm, 600 ns pulse exposure. To further this claim, we show that the nsPEF-induced activation mirrors the response of M1-acetylcholine Gq/11-coupled metabotropic receptor (hM1). This demonstration of PIP2 hydrolysis by nsPEF exposure is an important step toward understanding the mechanisms underlying this unique stimulus for activation of lipid signaling pathways and is critical for determining the potential for nsPEFs to modulate mammalian cell functions.


Assuntos
Membrana Celular/metabolismo , Eletricidade , Campos Eletromagnéticos , Fosfatidilinositóis/metabolismo , Animais , Cálcio/química , Cálcio/metabolismo , Caspases/metabolismo , Membrana Celular/efeitos da radiação , Citoplasma/efeitos da radiação , Humanos , Células Jurkat , Metabolismo dos Lipídeos/efeitos da radiação , Fosfatidilinositóis/química , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação
20.
PLoS One ; 8(4): e63122, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23658665

RESUMO

High-amplitude, MV/m, nanosecond pulsed electric fields (nsPEF) have been hypothesized to cause nanoporation of the plasma membrane. Phosphatidylserine (PS) externalization has been observed on the outer leaflet of the membrane shortly after nsPEF exposure, suggesting local structural changes in the membrane. In this study, we utilized fluorescently-tagged Annexin V to observe the externalization of PS on the plasma membrane of isolated Chinese Hamster Ovary (CHO) cells following exposure to nsPEF. A series of experiments were performed to determine the dosimetric trends of PS expression caused by nsPEF as a function of pulse duration, τ, delivered field strength, ED, and pulse number, n. To accurately estimate dose thresholds for cellular response, data were reduced to a set of binary responses and ED50s were estimated using Probit analysis. Probit analysis results revealed that PS externalization followed the non-linear trend of (τ*ED (2))(-1) for high amplitudes, but failed to predict low amplitude responses. A second set of experiments was performed to determine the nsPEF parameters necessary to cause observable calcium uptake, using cells preloaded with calcium green (CaGr), and membrane permeability, using FM1-43 dye. Calcium influx and FM1-43 uptake were found to always be observed at lower nsPEF exposure parameters compared to PS externalization. These findings suggest that multiple, higher amplitude and longer pulse exposures may generate pores of larger diameter enabling lateral diffusion of PS; whereas, smaller pores induced by fewer, lower amplitude and short pulse width exposures may only allow extracellular calcium and FM1-43 uptake.


Assuntos
Células CHO/efeitos da radiação , Permeabilidade da Membrana Celular/efeitos da radiação , Membrana Celular/efeitos da radiação , Fosfatidilserinas/metabolismo , Animais , Anexina A5 , Apoptose/efeitos da radiação , Células CHO/citologia , Células CHO/metabolismo , Cálcio/metabolismo , Membrana Celular/metabolismo , Cricetulus , Relação Dose-Resposta à Radiação , Campos Eletromagnéticos , Radiação Eletromagnética , Eletroporação/métodos , Corantes Fluorescentes , Compostos Orgânicos , Compostos de Piridínio , Compostos de Amônio Quaternário
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...