Network


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

Hotspot


Dive into the research topics where Gleb P. Tolstykh is active.

Publication


Featured researches published by Gleb P. Tolstykh.


Bioelectrochemistry | 2014

600 ns pulse electric field-induced phosphatidylinositol4,5-bisphosphate depletion

Gleb P. Tolstykh; Hope T. Beier; Caleb C. Roth; Gary L. Thompson; Bennett L. Ibey

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.


Journal of Neural Engineering | 2014

Plasma membrane nanoporation as a possible mechanism behind infrared excitation of cells

Hope T. Beier; Gleb P. Tolstykh; Joshua D. Musick; Robert J. Thomas; Bennett L. Ibey

OBJECTIVE Short infrared (IR) laser pulses have been used to stimulate action potentials in neurons both in vivo and in vitro. However, the mechanism(s) underlying this phenomenon has remained elusive. In vitro studies have found that pulsed IR exposure generates a nearly instant change in capacitance in the plasma membrane, characterized by inward rectification, a common feature in pore-forming exposures, such as electrical pulses and acoustic shock waves. Based on this similarity, we hypothesize that the mechanism of IR stimulation is the formation of short-lived nanopores in the plasma membrane. These transient, small-diameter pores allow the influx of extracellular ions that lead to action potential generation, possibly through activation of secondary messenger pathways or depolarization of the cell membrane resulting in activation of voltage-gated ion channels. APPROACH A variety of fluorescent markers are used to observe the cell response to IR stimulation to monitor for effects indicative of nanoporation in other modalities. MAIN RESULTS We observe rapid, transient rises in intracellular Ca(2+), influx of YO-PRO-1 and propidium iodide into the cell signifying membrane permeabilization, cellular blebbing and swelling, and activation of the intracellular phosphoinositides lipid signaling pathway. SIGNIFICANCE This conclusion better explains the experimental observations and limitations of IR-induced neurological stimulation and represents a distinct theoretical shift in the understanding of the mechanism of IR-induced stimulation.


Biochimica et Biophysica Acta | 2017

Nanosecond pulsed electric field induced dose dependent phosphatidylinositol-4,5-bisphosphate signaling and intracellular electro-sensitization

Gleb P. Tolstykh; Melissa Tarango; Caleb C. Roth; Bennett L. Ibey

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.


Proceedings of SPIE | 2015

The role of PIP2 and the IP3/DAG pathway in intracellular calcium release and cell survival during nanosecond electric pulse exposures

Zachary A. Steelman; Gleb P. Tolstykh; Larry E. Estlack; Caleb C. Roth; Bennett L. Ibey

Phosphatidylinositol4,5-biphosphate (PIP2) is a membrane phospholipid of particular importance in cell-signaling pathways. Hydrolysis of PIP2 releases inositol-1,4,5-triphosphate (IP3) from the membrane, activating IP3 receptors on the smooth endoplasmic reticulum (ER) and facilitating a release of intracellular calcium stores and activation of protein kinase C (PKC). Recent studies suggest that nanosecond pulsed electric fields (nsPEF) cause depletion of PIP2 in the cellular membrane, activating the IP3 signaling pathway. However, the exact mechanism(s) causing this observed depletion of PIP2 are unknown. Complicating the matter, nsPEF create nanopores in the plasma membrane, allowing calcium to enter the cell and thus causing an increase in intracellular calcium. While elevated intracellular calcium can cause activation of phospholipase C (PLC) (a known catalyst of PIP2 hydrolysis), PIP2 depletion has been shown to occur in the absence of both extracellular and intracellular calcium. These observations have led to the hypothesis that the high electric field itself may be playing a direct role in the hydrolysis of PIP2 from the plasma membrane. To support this hypothesis, we used edelfosine to block PLC and prevent activation of the IP3/DAG pathway in Chinese Hamster Ovarian (CHO) cells prior to applying nsPEF. Fluorescence microscopy was used to monitor intracellular calcium bursts during nsPEF, while MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) survivability assays were utilized to determine whether edelfosine improved cell survival during nsPEF exposure. This work is critical to refine the role of PIP2 in the cellular response to nsPEF, and also to determine the fundamental biological effects of high electric field exposures.


PLOS ONE | 2016

Evaluation of the Genetic Response of U937 and Jurkat Cells to 10-Nanosecond Electrical Pulses (nsEP)

Caleb C. Roth; Randolph D. Glickman; Gleb P. Tolstykh; Larry E. Estlack; Erick K. Moen; Ibtissam Echchgadda; Hope T. Beier; Ronald A. Barnes; Bennett L. Ibey

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.


Biochemistry and biophysics reports | 2017

nsPEF-induced PIP2 Depletion, PLC Activity and Actin Cytoskeletal Cortex Remodeling Are Responsible for Post-exposure Cellular Swelling and Blebbing

Gleb P. Tolstykh; Gary L. Thompson; Hope T. Beier; Zachary A. Steelman; Bennett L. Ibey

Cell swelling and blebbing has been commonly observed following nanosecond pulsed electric field (nsPEF) exposure. The hypothesized origin of these effects is nanoporation of the plasma membrane (PM) followed by transmembrane diffusion of extracellular fluid and disassembly of cortical actin structures. This investigation will provide evidence that shows passive movement of fluid into the cell through nanopores and increase of intracellular osmotic pressure are not solely responsible for this observed phenomena. We demonstrate that phosphatidylinositol-4,5-bisphosphate (PIP2) depletion and hydrolysis are critical steps in the chain reaction leading to cellular blebbing and swelling. PIP2 is heavily involved in osmoregulation by modulation of ion channels and also serves as an intracellular membrane anchor to cortical actin and phospholipase C (PLC). Given the rather critical role that PIP2 depletion appears to play in the response of cells to nsPEF exposure, it remains unclear how its downstream effects and, specifically, ion channel regulation may contribute to cellular swelling, blebbing, and unknown mechanisms of the lasting “permeabilization” of the PM.


Proceedings of SPIE | 2014

Dose dependent translocations of fluorescent probes of PIP2 hydrolysis in cells exposed to nanosecond pulsed electric fields

Gleb P. Tolstykh; Melissa Tarango; Caleb C. Roth; Bennett L. Ibey

Previously, it was demonstrated that small nanometer-sized pores (nanopores) are preferentially formed after exposure to nanosecond pulsed electric fields (nsPEF). We have reported that nanoporation of the plasma membrane directly affects the phospholipids of the cell membrane, ultimately culminating in phosphatidylinositol4,5- bisphosphate (PIP2) intracellular signaling. PIP2, located within the internal layer of the plasma membrane, plays a critical role as a regulator of ion transport proteins, a source of second messenger compounds, and an anchor for cytoskeletal elements. In this proceeding, we present data that demonstrates that nsPEFs initiate electric field dose-dependent PIP2 hydrolysis and/or depletion from the plasma membrane through the observation of the accumulation of inositol1,4,5-trisphosphate (IP3) in the cytoplasm and the increase of diacylglycerol (DAG) on the inner surface of the plasma membrane. The phosphoinositide signaling cascade presented here involves activation of phospholipase C (PLC) and protein kinase C (PKC), which are responsible for a multitude of biological effects after nsPEF exposure. These results expand our current knowledge of nsPEF induced physiological effects, and serve as a basis for development of novel tools for drug independent stimulation or modulation of different cellular functions.


Spie Newsroom | 2013

Nanosecond pulsed electric fields activate intracellular signaling pathways

Gleb P. Tolstykh; Thompson Gary L. Thompson; Hope T. Beier; Caleb C. Roth; Bennett L. Ibey

In cellular electrochemistry, ions respond to stimuli by constantly shuffling across cellular membranes to perform their physiological roles. This flow of ions, the electromotive force, leaves cells vulnerable to exogenous electromagnetic fields that can stimulate and/or modulate cellular activity. An irreparable link exists between changes in ionic concentration and the electric gradient of the cell (or its potential energy). Consequently, we can manipulate the physiology of the cell by altering its permeability to various ions, thereby modulating its electrical gradient. Only a few millivolts in excess of the resting membrane potential can stimulate a dramatic change in ion distribution within the cellular microenvironment. In excitable neural-type cells, electrical-stimulation-induced changes in membrane potential lead to the generation or inactivation of action potentials (AP). These AP trigger activities, such as nerve impulses in


Proceedings of SPIE | 2017

Fluorescence lifetime imaging of calcium flux in neurons in response to pulsed infrared light

Alex J. Walsh; Anna Sedelnikova; Gleb P. Tolstykh; Bennett L. Ibey; Hope T. Beier

Pulsed infrared light can excite action potentials in neurons; yet, the fundamental mechanism underlying this phenomenon is unknown. Previous work has observed a rise in intracellular calcium concentration following infrared exposure, but the source of the calcium and mechanism of release is unknown. Here, we used fluorescence lifetime imaging of Oregon Green BAPTA-1 to study intracellular calcium dynamics in primary rat hippocampal neurons in response to infrared light exposure. The fluorescence lifetime of Oregon Green BAPTA-1 is longer when bound to calcium, and allows robust measurement of intracellular free calcium concentrations. First, a fluorescence lifetime calcium calibration curve for Oregon Green BAPTA-1 was determined in solutions. The normalized amplitude of the short and long lifetimes was calibrated to calcium concentration. Then, neurons were incubated in Oregon Green BAPTA-1 and exposed to pulses of infrared light (0-1 J/cm2; 0-5 ms; 1869 nm). Fluorescence lifetime images were acquired prior to, during, and after the infrared exposure. Fluorescence lifetime images, 64x64 pixels, were acquired at 12 or 24 ms for frame rates of 83 and 42 Hz, respectively. Accurate α1 approximations were achieved in images with low photon counts by computing an α1 index value from the relative probability of the observed decay events. Results show infrared light exposure increases intracellular calcium in neurons. Altogether, this study demonstrates accurate fluorescence lifetime component analysis from low-photon count data for improved imaging speed.


Proceedings of SPIE | 2016

High frequency application of nanosecond pulsed electric fields alters cellular membrane disruption and fluorescent dye uptake

Zachary A. Steelman; Gleb P. Tolstykh; Hope T. Beier; Bennett L. Ibey

Cells exposed to nanosecond-pulsed electric fields (nsPEF) exhibit a wide variety of nonspecific effects, including blebbing, swelling, intracellular calcium bursts, apoptotic and necrotic cell death, formation of nanopores, and depletion of phosphatidylinositol 4,5-biphosphate (PIP2) to induce activation of the inositol trisphosphate/diacylglycerol pathway. While several studies have taken place in which multiple pulses were delivered to cells, the effect of pulse repetition rate (PRR) is not well understood. To better understand the effects of PRR, a laser scanning confocal microscope was used to observe CHO-K1 cells exposed to ten 600ns, 200V pulses at varying repetition rates (5Hz up to 500KHz) in the presence of either FM 1-43, YO-PRO-1, or Propidium Iodide (PI) fluorescent dyes, probes frequently used to indicate nanoporation or permeabilization of the plasma membrane. Dye uptake was monitored for 30 seconds after pulse application at a rate of 1 image/second. In addition, a single long pulse of equivalent energy (200V, 6 μs duration) was applied to test the hypothesis that very fast PRR will approximate the biological effects of a single long pulse of equal energy. Upon examination of the data, we found strong variation in the relationship between PRR and uptake in each of the three dyes. In particular, PI uptake showed little frequency dependence, FM 1-43 showed a strong inverse relationship between frequency and internal cell fluorescence, and YO-PRO-1 exhibited a “threshold” point of around 50 KHz, after which the inverse trend observed in FM 1-43 was seen to reverse itself. Further, a very high PRR of 500 KHz only approximated the biological effects of a single 6 μs pulse in cells stained with YO-PRO-1, suggesting that uptake of different dyes may proceed by different physical mechanisms.

Collaboration


Dive into the Gleb P. Tolstykh's collaboration.

Top Co-Authors

Avatar

Bennett L. Ibey

Air Force Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Hope T. Beier

Air Force Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Caleb C. Roth

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Sedelnikova

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Gary L. Thompson

Oak Ridge Institute for Science and Education

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stacey L. Martens

Air Force Research Laboratory

View shared research outputs
Researchain Logo
Decentralizing Knowledge