Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Bioelectromagnetics ; 39(4): 257-276, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29528124

RESUMEN

This article is based on my presentation at the D'Arsonval Ceremony at the Joint Annual Meeting of the Bioelectromagnetics Society and the European BioElectromagnetics Association in Hangzhou, China, in June of 2017. It describes the pathway from the first studies on the effects of intense, nanosecond pulses on biological cells to the development of medical therapies based on these effects. The motivation for the initial studies of the effects of high voltage, nanosecond pulses on mammalian cells was based on a simple electrical circuit model, which predicted that such pulses allow us to affect not just the plasma membrane but also the subcellular structures. The first experimental study that confirmed this hypothesis was published in 2001 in the Bioelectromagnetics journal. It was followed by a large number of publications that showed that such ultrashort pulses affect cell functions, such as programmed cell death, and, at lower intensity, calcium mobilization from intracellular structures. These basic studies were leading to novel cancer treatments, treatments of cardiac arrhythmia, and advanced wound healing. Further, by reducing the pulse duration into the picosecond range, antenna-based neural stimulation seems to be possible. This manuscript gives an overview of the progress in this field of research in the decade after the initial bioelectric studies with high-voltage, nanosecond pulses, particularly the research performed at the Frank Reidy Research Center for Bioelectrics. It also tells you about my journey and that of my colleagues at the Center for Bioelectrics into and through this fascinating bioelectromagnetics research area. Bioelectromagnetics. 39:257-276, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Terapia por Estimulación Eléctrica/métodos , Animales , Calcio/metabolismo , Muerte Celular , Humanos , Factores de Tiempo
2.
Bioelectrochemistry ; 105: 65-71, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26011130

RESUMEN

We tested if picosecond electric pulses (psEP; 190 kV/cm, 500 ps at 50% height), which are much shorter than channel activation time, can activate voltage-gated (VG) channels. Cytosolic Ca(2+) was monitored by Fura-2 ratiometric imaging in GH3 and NG108 cells (which express multiple types of VG calcium channels, VGCC), and in CHO cells (which express no VGCC). Trains of up to 100 psEP at 1 kHz elicited no response in CHO cells. However, even a single psEP significantly increased Ca(2+) in both GH3 (by 114 ± 48 nM) and NG108 cells (by 6 ± 1.1 nM). Trains of 100 psEP amplified the response to 379 ± 33 nM and 719 ± 315 nM, respectively. Ca(2+) responses peaked within 2-15s and recovered for over 100 s; they were 80-100% inhibited by verapamil and ω-conotoxin, but not by the substitution of Na(+) with N-methyl-D-glucamine. There was no response to psEP in Ca(2+)-free medium, but adding external Ca(2+) even 10s later evoked Ca(2+) response. We conclude that electrical stimuli as short as 500 ps can cause long-lasting opening of VGCC by a mechanism which does not involve conventional electroporation, heating (which was under 0.06 K per psEP), or membrane depolarization by opening of VG Na(+) channels.


Asunto(s)
Calcio/metabolismo , Estimulación Eléctrica , Animales , Células CHO , Cricetinae , Cricetulus , Ratones
3.
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
4.
In Vivo ; 25(1): 23-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21282730

RESUMEN

A mouse melanoma model was set up with green fluorescent protein (GFP) expression in vivo. With the same energy, long- (1 ms) and short- (300 ns) pulsed electric fields were delivered to two melanomas injected into the same mouse. The tumor growth and green fluorescence were followed up to compare the different treatment efficacy of long and short pulses. After two days post treatment, short pulse-treated tumors showed a significantly lower tumor volume compared with long pulse-treated tumors (n=8, p<0.05). On 8 experimental animals, a short nanosecond pulsed electric field (nsPEF) had lesser or delayed effects on GFP quenching and greater effects in reducing tumor size. Short pulses produced by nsPEFs can cause melanoma regression with less effect on the plasma membrane.


Asunto(s)
Terapia por Estimulación Eléctrica/métodos , Melanoma Experimental/terapia , Modalidades de Fisioterapia , Neoplasias Cutáneas/terapia , Animales , Membrana Celular/fisiología , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Melanoma Experimental/patología , Ratones , Neoplasias Cutáneas/patología , Resultado del Tratamiento
5.
J Dermatolog Treat ; 22(3): 153-61, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20666667

RESUMEN

A recent study has shown that nanosecond pulsed electric fields (nsPEF) can affect the intracellular structures of melanoma within weeks. nsPEF is a non-drug, non-thermal treatment using ultrashort, intense pulsed electric fields with nanosecond durations. In the current study we followed up melanoma histopathology and metastasis with tissue micro-array 5 months post-nsPEF. After nsPEF treatment, tumor growth, tumor histology, metastasis, peri-tumor vessel and micro-vessel density were examined for the effect of nsPEF treatment on melanoma in vivo. The 17 nsPEF-treated mice were tumor-free for 169 days, significantly longer than those 19 control mice bearing melanoma without nsPEF. Histopathology follow-up showed that melanoma did not recur to the primary injection place after complete elimination. Compared with the control tumor, nsPEF-treated tumors present decreased micro-vessel density in a time-course manner in this survival study. Treatment with nsPEF caused continuous histopathological changes in melanomas, eliminated melanoma without recurrence at the primary site and prolonged animal survival time by inhibiting tumor blood supply and leading to tumor infarction. Thus, nsPEF could be applied in a non-ionizing therapeutic approach, without other agents, to locally treat tumors within a defined boundary.


Asunto(s)
Terapia por Estimulación Eléctrica , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Animales , Femenino , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Melanoma Experimental/secundario , Ratones , Ratones Pelados , Trasplante de Neoplasias , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología
6.
Pigment Cell Melanoma Res ; 23(4): 554-63, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20370854

RESUMEN

Many effective anti-cancer strategies target apoptosis and angiogenesis mechanisms. Applications of non-ionizing, nanosecond pulsed electric fields (nsPEFs) induce apoptosis in vitro and eliminate cancer in vivo; however in vivo mechanisms require closer analysis. These studies investigate nsPEF-induced apoptosis and anti-angiogenesis examined by fluorescent microscopy, immunoblots, and morphology. Six hours after treatment with one hundred 300 ns pulses at 40 kV/cm, cells transiently expressed active caspases indicating that caspase-mediated mechanisms. Three hours after treatment transient peaks in Histone 2AX phosphorylation coincided with terminal deoxynucleotidyl transferase dUTP nick end labeling positive cells and pyknotic nuclei, suggesting caspase-independent mechanisms on nuclei/DNA. Large DNA fragments, but not 180 bp fragmentation ladders, were observed, suggesting incomplete apoptosis. Nevertheless, tumor weight and volume decreased and tumors disappeared. One week after treatment, vessel numbers, vascular endothelial growth factor (VEGF), platelet derived endothelial cell growth factor (PD-ECGF), CD31, CD35 and CD105 were decreased, indicating anti-angiogenesis. The nsPEFs activate multiple melanoma therapeutic targets, which is consistent with successes of nsPEF applications for tumor treatment in vivo as a new cancer therapeutic modality.


Asunto(s)
Apoptosis , Terapia por Estimulación Eléctrica/métodos , Melanoma Experimental/patología , Melanoma Experimental/terapia , Neovascularización Patológica , Animales , Caspasas/metabolismo , Línea Celular Tumoral , Forma del Núcleo Celular , Roturas del ADN de Doble Cadena , Etiquetado Corte-Fin in Situ , Melanoma Experimental/irrigación sanguínea , Ratones , Timidina Fosforilasa/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis
7.
Artículo en Inglés | MEDLINE | ID: mdl-19964169

RESUMEN

Cancer continues to be a major risk to the health and well being among populations around the world. A new method using ion-ionizing radiation with nanosecond pulsed electric fields (nsPEFs) provides a novel means to treat cancer at local sites. NsPEFs promote cell death in several cell types and here we investigated mechanisms for cell death induction. In murine B16f10 melanoma, murine E4 squamous carcinoma, murine Hep1-6 and human HepG2 hepatocellular carcinoma, nsPEFs induced cell death in 90-95% of cells. Cell death coincides with decreases in the mitochondria membrane potential, increases in YO-PRO-1 uptake and active caspases in the presence or absence of cytochrome c release. The results indicate that nsPEFs induced cell death by multiple apoptosis mechanisms that involve mitochondrial responses, but not necessarily through cytochrome c release. Further, these in vitro studies suggest a potential to induce cell death that bypasses cancer mechanisms that evade apoptosis.


Asunto(s)
Apoptosis/efectos de la radiación , Terapia por Estimulación Eléctrica/métodos , Nanomedicina/métodos , Neoplasias/fisiopatología , Neoplasias/terapia , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Ratones , Dosis de Radiación , Radiación no Ionizante
8.
Melanoma Res ; 19(6): 361-71, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19730404

RESUMEN

Nanosecond pulsed electric fields (nsPEFs) can affect the intracellular structures of cells in vitro. This study shows the direct effects of nsPEFs on tumor growth, tumor volume, and histological characteristics of normal skin and B16-F10 melanoma in SKH-1 mice. A melanoma model was set up by injecting B16-F10 into female SKH-1 mice. After a 100-pulse treatment with an nsPEF (40-kV/cm field strength; 300-ns duration; 30-ns rise time; 2-Hz repetition rate), tumor growth and histology were studied using transillumination, light microscopy with hematoxylin and eosin stain and transmission electron microscopy. Melanin and iron within the melanoma tumor were also detected with specific stains. After nsPEF treatment, tumor development was inhibited with decreased volumes post-nsPEF treatment compared with control tumors (P<0.05). The nsPEF-treated tumor volume was reduced significantly compared with the control group (P<0.01). Hematoxylin and eosin stain and transmission electron microscopy showed morphological changes and nuclear shrinkage in the tumor. Fontana-Masson stain indicates that nsPEF can externalize the melanin. Iron stain suggested nsPEF caused slight hemorrhage in the treated tissue. Histology confirmed that repeated applications of nsPEF disrupted the vascular network. nsPEF treatment can significantly disrupt the vasculature, reduce subcutaneous murine melanoma development, and produce tumor cell contraction and nuclear shrinkage while concurrently, but not permanently, damaging peripheral healthy skin tissue in the treated area, which we attribute to the highly localized electric fields surrounding the needle electrodes.


Asunto(s)
Terapia por Estimulación Eléctrica , Melanoma Experimental/patología , Melanoma Experimental/terapia , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Piel/química , Animales , Núcleo Celular , Electricidad , Femenino , Hierro/análisis , Melaninas/análisis , Melanoma Experimental/irrigación sanguínea , Ratones , Microscopía Electrónica de Transmisión , Neoplasias Cutáneas/irrigación sanguínea , Células Tumorales Cultivadas
9.
Int J Cancer ; 125(2): 438-45, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19408306

RESUMEN

We have discovered a new, ultrafast therapy for treating skin cancer that is extremely effective with a total electric field exposure time of only 180 microsec. The application of 300 high-voltage (40 kV/cm), ultrashort (300 nsec) electrical pulses to murine melanomas in vivo triggers both necrosis and apoptosis, resulting in complete tumor remission within an average of 47 days in the 17 animals treated. None of these melanomas recurred during a 4-month period after the initial melanoma had disappeared. These pulses generate small, long-lasting, rectifying nanopores in the plasma membrane of exposed cells, resulting in increased membrane permeability to small molecules and ions, as well as an increase in intracellular Ca(2+), DNA fragmentation, disruption of the tumor's blood supply and the initiation of apoptosis. Apoptosis was indicated by a 3-fold increase in Bad labeling and a 72% decrease in Bcl-2 labeling. In addition, microvessel density within the treated tumors fell by 93%. This new therapy utilizing nanosecond pulsed electric fields has the advantages of highly localized targeting of tumor cells and a total exposure time of only 180 microsec. These pulses penetrate into the interior of every tumor cell and initiate DNA fragmentation and apoptosis while at the same time reducing blood flow to the tumor. This new physical tumor therapy is drug free, highly localized, uses low energy, has no significant side effects and results in very little scarring.


Asunto(s)
Terapia por Estimulación Eléctrica , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Animales , Calcio/metabolismo , Femenino , Inmunohistoquímica , Melanoma Experimental/irrigación sanguínea , Ratones , Ratones Desnudos , Técnicas de Placa-Clamp , Recurrencia , Inducción de Remisión , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo
10.
Biochem Biophys Res Commun ; 343(2): 351-60, 2006 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-16545779

RESUMEN

We have discovered a new, drug-free therapy for treating solid skin tumors. Pulsed electric fields greater than 20 kV/cm with rise times of 30 ns and durations of 300 ns penetrate into the interior of tumor cells and cause tumor cell nuclei to rapidly shrink and tumor blood flow to stop. Melanomas shrink by 90% within two weeks following a cumulative field exposure time of 120 micros. A second treatment at this time can result in complete remission. This new technique provides a highly localized targeting of tumor cells with only minor effects on overlying skin. Each pulse deposits 0.2 J and 100 pulses increase the temperature of the treated region by only 3 degrees C, ten degrees lower than the minimum temperature for hyperthermia effects.


Asunto(s)
Terapia por Estimulación Eléctrica/métodos , Campos Electromagnéticos , Melanoma/patología , Melanoma/radioterapia , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/radioterapia , Animales , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Ratones , Nanotecnología/métodos , Dosis de Radiación , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA