Network


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

Hotspot


Dive into the research topics where Marina V. Shirmanova is active.

Publication


Featured researches published by Marina V. Shirmanova.


Physics in Medicine and Biology | 2008

Contrasting properties of gold nanoparticles for optical coherence tomography: phantom, in vivo studies and Monte Carlo simulation.

Elena V. Zagaynova; Marina V. Shirmanova; M Yu Kirillin; Boris N. Khlebtsov; Anna G. Orlova; I.V. Balalaeva; Marina A. Sirotkina; Marina L. Bugrova; Pavel Agrba; Vladislav A. Kamensky

The possibility of using silica-gold nanoshells with 150 nm silica core size and 25 nm thick gold shell as contrasting agents for optical coherence tomography (OCT) is analyzed. Experiments on agar biotissue phantoms showed that the penetration of nanoshells into the phantoms increases the intensity of the optical coherence tomography (OCT) signal and the brightness of the corresponding areas of the OCT image. In vivo experiments on rabbit skin demonstrated that the application of nanoshells onto the skin provides significant contrasting of the borders between the areas containing nanoshells and those without. This effect of nanoshells on skin in vivo is manifested by the increase in intensity of the OCT signal in superficial parts of the skin, boundary contrast between superficial and deep dermis and contrast of hair follicles and glands. The presence of nanoshells in the skin was confirmed by electron microscopy. Monte Carlo simulations of OCT images confirmed the possibility of contrasting skin-layer borders and structures by the application of gold nanoshells. The Monte Carlo simulations were performed for two skin models and exhibit effects of nanoparticles similar to those obtained in the experimental part of the study, thus proving that the effects originate exactly from the presence of nanoparticles.


Journal of Biomedical Optics | 2009

Contrasting properties of gold nanoshells and titanium dioxide nanoparticles for optical coherence tomography imaging of skin: Monte Carlo simulations and in vivo study

Mikhail Kirillin; Marina V. Shirmanova; Marina A. Sirotkina; Marina L. Bugrova; Boris N. Khlebtsov; Elena V. Zagaynova

The effect of silica/gold nanoshells and titanium dioxide nanoparticles on the optical properties of skin is studied. By implementing in vivo measurements and Monte Carlo simulations, we analyze the efficiency of using these nanoparticles as contrasting agents for optical coherence tomography (OCT) imaging of skin. In vivo measurements are performed on pig skin, where nanoparticle suspension drops have been applied. The identification of skin layers is performed by comparison with corresponding histology images. Experimental results exhibit an increase in contrast of the obtained OCT images after a single nanoparticles application. Multiple applications do not lead to increase in the obtained contrast. To interpret the obtained experimental OCT images of skin and understand the mechanisms of contrasting, a set of Monte Carlo calculations is performed. The results of the simulations exhibit good qualitative agreement with the experimental images, and prove that the contrasting originates from the nanoparticles added, while the contrast of inclusion originates from the absence of nanoparticles within it and their presence in the surrounding area.


Journal of Biophotonics | 2013

Phototoxic effects of fluorescent protein KillerRed on tumor cells in mice

Marina V. Shirmanova; Ekaterina O. Serebrovskaya; Konstantin A. Lukyanov; Ludmila B. Snopova; Marina A. Sirotkina; Natalia N. Prodanetz; Marina L. Bugrova; Ekaterina A. Minakova; Ilya V. Turchin; Vladislav A. Kamensky; Sergey Lukyanov; Elena V. Zagaynova

KillerRed is known to be a unique red fluorescent protein displaying strong phototoxic properties. Its effectiveness has been shown previously for killing bacterial and cancer cells in vitro. Here, we investigated the photototoxicity of the protein on tumor xenografts in mice. HeLa Kyoto cell line stably expressing KillerRed in mitochondria and in fusion with histone H2B was used. Irradiation of the tumors with 593 nm laser led to photobleaching of KillerRed indicating photosensitization reaction and caused significant destruction of the cells and activation of apoptosis. The portion of the dystrophically changed cells increased from 9.9% to 63.7%, and the cells with apoptosis hallmarks from 6.3% to 14%. The results of this study suggest KillerRed as a potential genetically encoded photosensitizer for photodynamic therapy of cancer.


Biochimica et Biophysica Acta | 2013

Flavoprotein miniSOG as a genetically encoded photosensitizer for cancer cells

Alina P. Ryumina; Ekaterina O. Serebrovskaya; Marina V. Shirmanova; Ludmila B. Snopova; Maria M. Kuznetsova; Ilya V. Turchin; Nadezhda I. Ignatova; Natalia V. Klementieva; Arkady F. Fradkov; Boris E. Shakhov; Elena V. Zagaynova; Konstantin A. Lukyanov; Sergey Lukyanov

BACKGROUND Genetically encoded photosensitizers are a promising optogenetic instrument for light-induced production of reactive oxygen species in desired locations within cells in vitro or whole body in vivo. Only two such photosensitizers are currently known, GFP-like protein KillerRed and FMN-binding protein miniSOG. In this work we studied phototoxic effects of miniSOG in cancer cells. METHODS HeLa Kyoto cell lines stably expressing miniSOG in different localizations, namely, plasma membrane, mitochondria or chromatin (fused with histone H2B) were created. Phototoxicity of miniSOG was tested on the cells in vitro and tumor xenografts in vivo. RESULTS Blue light induced pronounced cell death in all three cell lines in a dose-dependent manner. Caspase 3 activation was characteristic of illuminated cells with mitochondria- and chromatin-localized miniSOG, but not with miniSOG in the plasma membrane. In addition, H2B-miniSOG-expressing cells demonstrated light-induced activation of DNA repair machinery, which indicates massive damage of genomic DNA. In contrast to these in vitro data, no detectable phototoxicity was observed on tumor xenografts with HeLa Kyoto cell lines expressing mitochondria- or chromatin-localized miniSOG. CONCLUSIONS miniSOG is an excellent genetically encoded photosensitizer for mammalian cells in vitro, but it is inferior to KillerRed in the HeLa tumor. GENERAL SIGNIFICANCE This is the first study to assess phototoxicity of miniSOG in cancer cells. The results suggest an effective ontogenetic tool and may be of interest for molecular and cell biology and biomedical applications.


Biochimica et Biophysica Acta | 2015

Intracellular pH imaging in cancer cells in vitro and tumors in vivo using the new genetically encoded sensor SypHer2.

Marina V. Shirmanova; Irina N. Druzhkova; Maria M. Lukina; Mikhail E. Matlashov; Vsevolod V. Belousov; Ludmila B. Snopova; Natalia N. Prodanetz; Varvara V. Dudenkova; Sergey Lukyanov; Elena V. Zagaynova

BACKGROUND Measuring intracellular pH (pHi) in tumors is essential for the monitoring of cancer progression and the response of cancer cells to various treatments. The purpose of the study was to develop a method for pHi mapping in living cancer cells in vitro and in tumors in vivo, using the novel genetically encoded indicator, SypHer2. METHODS A HeLa Kyoto cell line stably expressing SypHer2 in the cytoplasm was used, to perform ratiometric (dual excitation) imaging of the probe in cell culture, in 3D tumor spheroids and in tumor xenografts in living mice. RESULTS Using SypHer2, pHi was demonstrated to be 7.34±0.11 in monolayer HeLa cells in vitro under standard cultivation conditions. An increasing pHi gradient from the center to the periphery of the spheroids was displayed. We obtained fluorescence ratio maps for HeLa tumors in vivo and ex vivo. Comparison of the map with the pathomorphology and with hypoxia staining of the tumors revealed a correspondence of the zones with higher pHi to the necrotic and hypoxic areas. CONCLUSIONS Our results demonstrate that pHi imaging with the genetically encoded pHi indicator, SypHer2, can be a valuable tool for evaluating tumor progression in xenograft models. GENERAL SIGNIFICANCE We have demonstrated, for the first time, the possibility of using the genetically encoded sensor SypHer2 for ratiometric pH imaging in cancer cells in vitro and in tumors in vivo. SypHer2 shows great promise as an instrument for pHi monitoring able to provide high accuracy and spatiotemporal resolution.


Methods in Enzymology | 2013

Visualization of intracellular hydrogen peroxide with HyPer, a genetically encoded fluorescent probe

Nataliya M. Mishina; Kseniya N. Markvicheva; Dmitry S. Bilan; Mikhail E. Matlashov; Marina V. Shirmanova; David Liebl; Carsten Schultz; Sergey Lukyanov; Vsevolod V. Belousov

The fluorescent sensor HyPer allows monitoring of intracellular H2O2 levels with a high degree of sensitivity and specificity. Here, we provide a detailed protocol of ratiometric imaging of H2O2 produced by cells during phagocytosis, including instructions for experiments on different commercial confocal systems, namely, Leica SP2, Leica SP5, and Carl Zeiss LSM, as well as wide-field Leica 6000 microscope. The general experimental scheme is easily adaptable for imaging H2O2 production by various cell types under a variety of conditions.


Journal of Biomedical Optics | 2008

Fluorescence diffuse tomography for detection of red fluorescent protein expressed tumors in small animals.

Ilya V. Turchin; Vladislav A. Kamensky; Vladimir I. Plehanov; Anna G. Orlova; Mikhail Kleshnin; Ilya I. Fiks; Marina V. Shirmanova; Irina G. Meerovich; Lyaisan R. Arslanbaeva; Viktoria V. Jerdeva; Alexander P. Savitsky

A fluorescence diffuse tomography (FDT) setup for monitoring tumor growth in small animals has been created. In this setup an animal is scanned in the transilluminative configuration by a single source and detector pair. To remove stray light in the detection system, we used a combination of interferometric and absorption filters. To reduce the scanning time, an experimental animal was scanned using the following algorithm: (1) large-step scanning to obtain a general view of the animal (source and detector move synchronously); (2) selection of the fluorescing region; and (3) small-step scanning of the selected region and different relative shifts between the source and detector to obtain sufficient information for 3D reconstruction. We created a reconstruction algorithm based on the Holder norm to estimate the fluorophore distribution. This algorithm converges to the solution with a minimum number of fluorescing zones. The use of tumor cell lines transfected with fluorescent proteins allowed us to conduct intravital monitoring studies. Cell lines of human melanomas Mel-P, Mel-Ibr, Mel-Kor, and human embryonic kidney HEK293 Phoenix were transfected with DsRed-Express and Turbo-RFP genes. The emission of red fluorescent proteins (RFPs) in the long-wave optical range permits detection of deep-seated tumors. In vivo experiments were conducted immediately after subcutaneous injection of fluorescing cells into small animals.


Biomedical Optics Express | 2015

Simultaneous photoacoustic and optically mediated ultrasound microscopy: an in vivo study.

Pavel Subochev; Anna Orlova; Marina V. Shirmanova; Anna S. Postnikova; Ilya V. Turchin

We propose the use of thermoelastic (TE) excitation of an ultrasonic (US) detector by backscattered laser radiation as a means of upgrading a single-modality photoacoustic (PA) microscope to dual-modality PA/US imaging at minimal cost. The upgraded scanning head of our dual-modality microscope consists of a fiber bundle with 14 output arms and a 32MHz polyvinylidene difluoride (PVDF) detector with a 34 MHz bandwidth (-6 dB level), 12.7 mm focal length, and a 0.25 numerical aperture. A single optical pulse delivered through the fiber bundle to the biotissue being investigated, in combination with a metalized surface on the PVDF detector allows us to obtain both PA and US A-scans. To demonstrate the in vivo capabilities of the proposed method we present the results of bimodal imaging of the brain of a newborn rat, a mouse tail and a mouse tumor.


Journal of Biomedical Optics | 2013

Phototoxic effects of lysosome-associated genetically encoded photosensitizer KillerRed

Ekaterina O. Serebrovskaya; Alina P. Ryumina; Maria E. Boulina; Marina V. Shirmanova; Elena V. Zagaynova; Ekaterina A. Bogdanova; Sergey Lukyanov; Konstantin A. Lukyanov

Abstract. KillerRed is a unique phototoxic red fluorescent protein that can be used to induce local oxidative stress by green-orange light illumination. Here we studied phototoxicity of KillerRed targeted to cytoplasmic surface of lysosomes via fusion with Rab7, a small GTPase that is known to be attached to membranes of late endosomes and lysosomes. It was found that lysosome-associated KillerRed ensures efficient light-induced cell death similar to previously reported mitochondria- and plasma membrane–localized KillerRed. Inhibitory analysis demonstrated that lysosomal cathepsins play an important role in the manifestation of KillerRed-Rab7 phototoxicity. Time-lapse monitoring of cell morphology, membrane integrity, and nuclei shape allowed us to conclude that KillerRed-Rab7-mediated cell death occurs via necrosis at high light intensity or via apoptosis at lower light intensity. Potentially, KillerRed-Rab7 can be used as an optogenetic tool to direct target cell populations to either apoptosis or necrosis.


Scientific Reports | 2016

Probing metabolic states of differentiating stem cells using two-photon FLIM

Aleksandra V. Meleshina; Varvara V. Dudenkova; Marina V. Shirmanova; Vladislav I. Shcheslavskiy; Wolfgang Becker; Alena S. Bystrova; Elena I. Cherkasova; Elena V. Zagaynova

The ability of stem cells to differentiate into specialized cell types presents a number of opportunities for regenerative medicine, stem cell therapy and developmental biology. Because traditional assessments of stem cells are destructive, time consuming, and logistically intensive, the use of a non-invasive, label-free approach to study of cell differentiation provides a powerful tool for rapid, high-content characterization of cell and tissue cultures. Here, we elucidate the metabolic changes in MSCs during adipogenic differentiation, based on the fluorescence of the metabolic co-factors NADH, NADPH, and FAD using the methods of two-photon fluorescence microscopy combined with FLIM. To estimate the contribution of energy metabolism and lipogenesis in the observed changes of the metabolic profile, a separate analysis of NADH and NADPH is required. In our study we demonstrated, for the first time, an increased contribution of protein-bound NADPH in adipocytes that is associated with lipogenesis. The optical redox ratio FAD/NAD(P)H decreased during adipogenic differentiation, and that this was likely to be explained by the intensive biosynthesis of lipids and the enhanced NADPH production associated with this. Based on the data on the fluorescence lifetime contribution of protein-bound NAD(P)H, we registered a metabolic switch from glycolysis to oxidative phosphorylation in adipocytes.

Collaboration


Dive into the Marina V. Shirmanova's collaboration.

Top Co-Authors

Avatar

Elena V. Zagaynova

Nizhny Novgorod State Medical Academy

View shared research outputs
Top Co-Authors

Avatar

Varvara V. Dudenkova

Nizhny Novgorod State Medical Academy

View shared research outputs
Top Co-Authors

Avatar

Maria M. Lukina

Nizhny Novgorod State Medical Academy

View shared research outputs
Top Co-Authors

Avatar

Ilya V. Turchin

Russian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marina A. Sirotkina

Nizhny Novgorod State Medical Academy

View shared research outputs
Top Co-Authors

Avatar

Irina N. Druzhkova

Nizhny Novgorod State Medical Academy

View shared research outputs
Top Co-Authors

Avatar

Ludmila B. Snopova

Nizhny Novgorod State Medical Academy

View shared research outputs
Top Co-Authors

Avatar

Sergey Lukyanov

Russian National Research Medical University

View shared research outputs
Top Co-Authors

Avatar

Anna G. Orlova

Nizhny Novgorod State Medical Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge