Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrei G. Pakhomov is active.

Publication


Featured researches published by Andrei G. Pakhomov.


Bioelectromagnetics | 1998

Current state and implications of research on biological effects of millimeter waves: A review of the literature

Andrei G. Pakhomov; Yahya Akyel; Olga N. Pakhomova; Bruce E. Stuck; Michael R. Murphy

In recent years, research into biological and medical effects of millimeter waves (MMW) has expanded greatly. This paper analyzes general trends in the area and briefly reviews the most significant publications, proceeding from cell-free systems, dosimetry, and spectroscopy issues through cultured cells and isolated organs to animals and humans. The studies reviewed demonstrate effects of low-intensity MMW (10 mW/cm2 and less) on cell growth and proliferation, activity of enzymes, state of cell genetic apparatus, function of excitable membranes, peripheral receptors, and other biological systems. In animals and humans, local MMW exposure stimulated tissue repair and regeneration, alleviated stress reactions, and facilitated recovery in a wide range of diseases (MMW therapy). Many reported MMW effects could not be readily explained by temperature changes during irradiation. The paper outlines some problems and uncertainties in the MMW research area, identifies tasks for future studies, and discusses possible implications for development of exposure safety criteria and guidelines.


International Journal of Cancer | 2009

A new pulsed electric field therapy for melanoma disrupts the tumor's blood supply and causes complete remission without recurrence

Richard Nuccitelli; Xinhua Chen; Andrei G. Pakhomov; Wallace H. Baldwin; Saleh Sheikh; Jennifer L. Pomicter; Wei Ren; Christopher Osgood; R. James Swanson; Juergen F. Kolb; Stephen J. Beebe; Karl H. Schoenbach

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 μsec. 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 Ca2+, DNA fragmentation, disruption of the tumors 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 μsec. 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.


Biochimica et Biophysica Acta | 2011

Manipulation of cell volume and membrane pore comparison following single cell permeabilization with 60- and 600-ns electric pulses

Olena M. Nesin; Olga N. Pakhomova; Shu Xiao; Andrei G. Pakhomov

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.


Bioelectromagnetics | 2009

Plasma membrane permeabilization by 60‐ and 600‐ns electric pulses is determined by the absorbed dose

Bennett L. Ibey; Shu Xiao; Karl H. Schoenbach; Michael R. Murphy; Andrei G. Pakhomov

We explored how the effect of plasma membrane permeabilization by nanosecond-duration electric pulses (nsEP) depends on the physical characteristics of exposure. The resting membrane resistance (R(m)) and membrane potential (MP) were measured in cultured GH3 and CHO cells by conventional whole-cell patch-clamp technique. Intact cells were exposed to a single nsEP (60 or 600 ns duration, 0-22 kV/cm), followed by patch-clamp measurements after a 2-3 min delay. Consistent with earlier findings, nsEP caused long-lasting R(m) decrease, accompanied by the loss of MP. The threshold for these effects was about 6 kV/cm for 60 ns pulses, and about 1 kV/cm for 600 ns pulses. Further analysis established that it was neither pulse duration nor the E-field amplitude per se, but the absorbed dose that determined the magnitude of the biological effect. In other words, exposure to nsEP at either pulse duration caused equal effects if the absorbed doses were equal. The threshold absorbed dose to produce plasma membrane effects in either GH3 or CHO cells at either pulse duration was found to be at or below 10 mJ/g. Despite being determined by the dose, the nsEP effect clearly is not thermal, as the maximum heating at the threshold dose is less than 0.01 degrees C. The use of the absorbed dose as a universal exposure metric may help to compare and quantify nsEP sensitivity of different cell types and of cells in different physiological conditions. The absorbed dose may also prove to be a more useful metric than the incident E-field in determining safety limits for high peak, low average power EMF emissions.


PLOS ONE | 2011

Electroporation-Induced Electrosensitization

Olga N. Pakhomova; Andrei G. Pakhomov

Background Electroporation is a method of disrupting the integrity of cell membrane by electric pulses (EPs). Electrical modeling is widely employed to explain and study electroporation, but even most advanced models show limited predictive power. No studies have accounted for the biological consequences of electroporation as a factor that alters the cells susceptibility to forthcoming EPs. Methodology/Principal Findings We focused first on the role of EP rate for membrane permeabilization and lethal effects in mammalian cells. The rate was varied from 0.001 to 2,000 Hz while keeping other parameters constant (2 to 3,750 pulses of 60-ns to 9-µs duration, 1.8 to 13.3 kV/cm). The efficiency of all EP treatments was minimal at high rates and started to increase gradually when the rate decreased below a certain value. Although this value ranged widely (0.1–500 Hz), it always corresponded to the overall treatment duration near 10 s. We further found that longer exposures were more efficient irrespective of the EP rate, and that splitting a high-rate EP train in two fractions with 1–5 min delay enhanced the effects severalfold. Conclusions/Significance For varied experimental conditions, EPs triggered a delayed and gradual sensitization to EPs. When a portion of a multi-pulse exposure was delivered to already sensitized cells, the overall effect markedly increased. Because of the sensitization, the lethality in EP-treated cells could be increased from 0 to 90% simply by increasing the exposure duration, or the exposure dose could be reduced twofold without reducing the effect. Many applications of electroporation can benefit from accounting for sensitization, by organizing the exposure either to maximize sensitization (e.g., for sterilization) or, for other applications, to completely or partially avoid it. In particular, harmful side effects of electroporation-based therapies (electrochemotherapy, gene therapies, tumor ablation) include convulsions, pain, heart fibrillation, and thermal damage. Sensitization can potentially be employed to reduce these side effects while preserving or increasing therapeutic efficiency.


IEEE Transactions on Plasma Science | 2004

Characterization of the cytotoxic effect of high-intensity, 10-ns duration electrical pulses

Andrei G. Pakhomov; Amy Phinney; John Ashmore; Kerfoot Walker; Juergen F. Kolb; S. Kono; Karl H. Schoenbach; Michael R. Murphy

Cytotoxic effects of 10-ns electrical pulses (EP, 50-380 kV/cm) were analyzed in cultured U937 cells (human lymphoma). Densities of live and dead cells were compared in over 500 samples at intervals from 0.5 to 48 h post exposure. EP trains of 1-20 pulses caused a minor, if any, decrease in cell survival: 24 h post exposure, the density of live cells typically dropped just 10-20% compared with unexposed parallel control. Within studied limits, this effect did not significantly depend on the EP number, voltage, or repetition rate. However, much longer EP trains could cause a sharp survival decline. The transition from plateau to profound cell killing occurred at about 150 pulses at 150 kV/cm, and at over 1000 pulses at 50 kV/cm. Artifact-free thermometry using a fiber optic microprobe established unequivocally that cell killing by extra-long EP trains was not a result of sample heating and has to be explained by other mechanisms. Testing for specific apoptotic cleavage of poly(ADP-ribose) polymerase at scattered time intervals (1-24 h) after EP exposure produced mostly negative results. Overall, 10-ns EP caused far weaker cytotoxic effect than it was reported earlier from experiments in other cell lines and mostly with longer pulses (60 and 300 ns). The survival curve shape (i.e., the shoulder followed by exponential decline) is also characteristic for other cytotoxic factors, such as low-LET ionizing radiation, thereby possibly pointing to common mechanisms or targets.


Biochimica et Biophysica Acta | 2013

Primary pathways of intracellular Ca2+ mobilization by nanosecond pulsed electric field

Iurii Semenov; Shu Xiao; Andrei G. Pakhomov

Permeabilization of cell membranous structures by nanosecond pulsed electric field (nsPEF) triggers transient rise of cytosolic Ca(2+) concentration ([Ca(2+)](i)), which determines multifarious downstream effects. By using fast ratiometric Ca(2+) imaging with Fura-2, we quantified the external Ca(2+) uptake, compared it with Ca(2+) release from the endoplasmic reticulum (ER), and analyzed the interplay of these processes. We utilized CHO cells which lack voltage-gated Ca(2+) channels, so that the nsPEF-induced [Ca(2+)](i) changes could be attributed primarily to electroporation. We found that a single 60-ns pulse caused fast [Ca(2+)](i) increase by Ca(2+) influx from the outside and Ca(2+) efflux from the ER, with the E-field thresholds of about 9 and 19kV/cm, respectively. Above these thresholds, the amplitude of [Ca(2+)](i) response increased linearly by 8-10nM per 1kV/cm until a critical level between 200 and 300nM of [Ca(2+)](i) was reached. If the critical level was reached, the nsPEF-induced Ca(2+) signal was amplified up to 3000nM by engaging the physiological mechanism of Ca(2+)-induced Ca(2+)-release (CICR). The amplification was prevented by depleting Ca(2+) from the ER store with 100nM thapsigargin, as well as by blocking the ER inositol-1,4,5-trisphosphate receptors (IP(3)R) with 50μM of 2-aminoethoxydiphenyl borate (2-APB). Mobilization of [Ca(2+)](i) by nsPEF mimicked native Ca(2+) signaling, but without preceding activation of plasma membrane receptors or channels. NsPEF stimulation may serve as a unique method to mobilize [Ca(2+)](i) and activate downstream cascades while bypassing the plasma membrane receptors.


PLOS ONE | 2011

Dose-Dependent Thresholds of 10-ns Electric Pulse Induced Plasma Membrane Disruption and Cytotoxicity in Multiple Cell Lines

Bennett L. Ibey; Caleb C. Roth; Andrei G. Pakhomov; Joshua A. Bernhard; Gerald J. Wilmink; Olga N. Pakhomova

In this study, we determined the LD50 (50% lethal dose) for cell death, and the ED50 (50% of cell population staining positive) for propidium (Pr) iodide uptake, and phosphatidylserine (PS) externalization for several commonly studied cell lines (HeLa, Jurkat, U937, CHO-K1, and GH3) exposed to 10-ns electric pulses (EP). We found that the LD50 varied substantially across the cell lines studied, increasing from 51 J/g for Jurkat to 1861 J/g for HeLa. PS externalized at doses equal or lower than that required for death in all cell lines ranging from 51 J/g in Jurkat, to 199 J/g in CHO-K1. Pr uptake occurred at doses lower than required for death in three of the cell lines: 656 J/g for CHO-K1, 634 J/g for HeLa, and 142 J/g for GH3. Both Jurkat and U937 had a LD50 lower than the ED50 for Pr uptake at 780 J/g and 1274 J/g, respectively. The mechanism responsible for these differences was explored by evaluating cell size, calcium concentration in the exposure medium, and effect of trypsin treatment prior to exposure. None of the studied parameters correlated with the observed results suggesting that cellular susceptibility to injury and death by 10-ns EP was largely determined by cell physiology. In contrast to previous studies, our findings suggest that permeabilization of internal membranes may not necessarily be responsible for cell death by 10-ns EP. Additionally, a mixture of Jurkat and HeLa cells was exposed to 10-ns EP at a dose of 280 J/g. Death was observed only in Jurkat cells suggesting that 10-ns EP may selectively kill cells within a heterogeneous tissue.


Bioelectrochemistry | 2010

Plasma Membrane Permeabilization by Trains of Ultrashort Electric Pulses

Bennett L. Ibey; Dustin G. Mixon; Jason Payne; Angela M. Bowman; Karl Sickendick; Gerald J. Wilmink; William P. Roach; Andrei G. Pakhomov

Ultrashort electric pulses (USEP) cause long-lasting increase of cell membrane electrical conductance, and that a single USEP increased cell membrane electrical conductance proportionally to the absorbed dose (AD) with a threshold of about 10 mJ/g. The present study extends quantification of the membrane permeabilization effect to multiple USEP and employed a more accurate protocol that identified USEP effect as the difference between post- and pre-exposure conductance values (Deltag) in individual cells. We showed that Deltag can be increased by either increasing the number of pulses at a constant E-field, or by increasing the E-field at a constant number of pulses. For 60-ns pulses, an E-field threshold of 6 kV/cm for a single pulse was lowered to less than 1.7 kV/cm by applying 100-pulse or longer trains. However, the reduction of the E-field threshold was only achieved at the expense of a higher AD compared to a single pulse exposure. Furthermore, the effect of multiple pulses was not fully determined by AD, suggesting that cells permeabilized by the first pulse(s) in the train become less vulnerable to subsequent pulses. This explanation was corroborated by a model that treated multiple-pulse exposures as a series of single-pulse exposures and assumed an exponential decline of cell susceptibility to USEP as Deltag increased after each pulse during the course of the train.


Biochimica et Biophysica Acta | 2015

Multiple nanosecond electric pulses increase the number but not the size of long-lived nanopores in the cell membrane

Andrei G. Pakhomov; Elena C. Gianulis; P. Thomas Vernier; Iurii Semenov; Shu Xiao; Olga N. Pakhomova

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.

Collaboration


Dive into the Andrei G. Pakhomov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shu Xiao

Old Dominion University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bennett L. Ibey

Air Force Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Michael R. Murphy

Air Force Research Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R. P. Joshi

Old Dominion University

View shared research outputs
Researchain Logo
Decentralizing Knowledge