Network


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

Hotspot


Dive into the research topics where Evgenia V. Gerasimovskaya is active.

Publication


Featured researches published by Evgenia V. Gerasimovskaya.


Annual Review of Physiology | 2013

The Adventitia: Essential Regulator of Vascular Wall Structure and Function

Kurt R. Stenmark; Michael E. Yeager; Karim C. El Kasmi; Eva Nozik-Grayck; Evgenia V. Gerasimovskaya; Min Li; Suzette R. Riddle; Maria G. Frid

The vascular adventitia acts as a biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. It is the most complex compartment of the vessel wall and is composed of a variety of cells, including fibroblasts, immunomodulatory cells (dendritic cells and macrophages), progenitor cells, vasa vasorum endothelial cells and pericytes, and adrenergic nerves. In response to vascular stress or injury, resident adventitial cells are often the first to be activated and reprogrammed to influence the tone and structure of the vessel wall; to initiate and perpetuate chronic vascular inflammation; and to stimulate expansion of the vasa vasorum, which can act as a conduit for continued inflammatory and progenitor cell delivery to the vessel wall. This review presents the current evidence demonstrating that the adventitia acts as a key regulator of vascular wall function and structure from the outside in.


American Journal of Physiology-cell Physiology | 2011

P2Y1 and P2Y13 purinergic receptors mediate Ca2+ signaling and proliferative responses in pulmonary artery vasa vasorum endothelial cells

Taras Lyubchenko; Heather Woodward; Kristopher D. Veo; Nana Burns; Hala Nijmeh; Ganna A. Liubchenko; Kurt R. Stenmark; Evgenia V. Gerasimovskaya

Extracellular ATP and ADP have been shown to exhibit potent angiogenic effects on pulmonary artery adventitial vasa vasorum endothelial cells (VVEC). However, the molecular signaling mechanisms of extracellular nucleotide-mediated angiogenesis remain not fully elucidated. Since elevation of intracellular Ca(2+) concentration ([Ca(2+)](i)) is required for cell proliferation and occurs in response to extracellular nucleotides, this study was undertaken to delineate the purinergic receptor subtypes involved in Ca(2+) signaling and extracellular nucleotide-mediated mitogenic responses in VVEC. Our data indicate that stimulation of VVEC with extracellular ATP resulted in the elevation of [Ca(2+)](i) via Ca(2+) influx through plasma membrane channels as well as Ca(2+) mobilization from intracellular stores. Moreover, extracellular ATP induced simultaneous Ca(2+) responses in both cytosolic and nuclear compartments. An increase in [Ca(2+)](i) was observed in response to a wide range of purinergic receptor agonists, including ATP, ADP, ATPγS, ADPβS, UTP, UDP, 2-methylthio-ATP (MeSATP), 2-methylthio-ADP (MeSADP), and BzATP, but not adenosine, AMP, diadenosine tetraphosphate, αβMeATP, and βγMeATP. Using RT-PCR, we identified mRNA for the P2Y1, P2Y2, P2Y4, P2Y13, P2Y14, P2X2, P2X5, P2X7, A1, A2b, and A3 purinergic receptors in VVEC. Preincubation of VVEC with the P2Y1 selective antagonist MRS2179 and the P2Y13 selective antagonist MRS2211, as well as with pertussis toxin, attenuated at varying degrees agonist-induced intracellular Ca(2+) responses and activation of ERK1/2, Akt, and S6 ribosomal protein, indicating that P2Y1 and P2Y13 receptors play a major role in VVEC growth responses. Considering the broad physiological implications of purinergic signaling in the regulation of angiogenesis and vascular homeostasis, our findings suggest that P2Y1 and P2Y13 receptors may represent novel and specific targets for treatment of pathological vascular remodeling involving vasa vasorum expansion.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2009

PI3K, Rho, and ROCK play a key role in hypoxia-induced ATP release and ATP-stimulated angiogenic responses in pulmonary artery vasa vasorum endothelial cells

Heather Woodward; Adil Anwar; Suzette R. Riddle; Laimute Taraseviciene-Stewart; Miguel Fragoso; Kurt R. Stenmark; Evgenia V. Gerasimovskaya

We recently reported that vasa vasorum expansion occurs in the pulmonary artery (PA) adventitia of chronically hypoxic animals and that extracellular ATP is a pro-angiogenic factor for isolated vasa vasorum endothelial cells (VVEC). However, the sources of extracellular ATP in the PA vascular wall, as well as the molecular mechanisms underlying its release, remain elusive. Studies were undertaken to explore whether VVEC release ATP in response to hypoxia and to determine signaling pathways involved in this process. We found that hypoxia (1-3% O2) resulted in time- and O2-dependent ATP release from VVEC. Preincubation with the inhibitors of vesicular transport (monensin, brefeldin A, and N-ethylmaleimide) significantly decreased ATP accumulation in the VVEC conditioned media, suggesting that hypoxia-induced ATP release occurs through vesicular exocytosis. Additionally, both hypoxia and exogenously added ATP resulted in the activation of PI3K and accumulation of GTP-bound RhoA in a time-dependent manner. Pharmacological inhibition of PI3K and ROCK or knockout of RhoA by small interfering RNA significantly abolished hypoxia-induced ATP release from VVEC. Moreover, RhoA and ROCK play a critical role in ATP-induced increases in VVEC DNA synthesis, migration, and tube formation, indicating a functional contribution of PI3K, Rho, and ROCK to both the autocrine mechanism of ATP release and ATP-mediated angiogenic activation of VVEC. Taken together, our findings provide novel evidence for the signaling mechanisms that link hypoxia-induced increases in extracellular ATP and vasa vasorum expansion.


PLOS ONE | 2013

Adenosine A1 Receptors Promote Vasa Vasorum Endothelial Cell Barrier Integrity via Gi and Akt-Dependent Actin Cytoskeleton Remodeling

Nagavedi S. Umapathy; Elzbieta Kaczmarek; Nooreen Fatteh; Nana Burns; Rudolf Lucas; Kurt R. Stenmark; Alexander D. Verin; Evgenia V. Gerasimovskaya

Background In a neonatal model of hypoxic pulmonary hypertension, a dramatic pulmonary artery adventitial thickening, accumulation of inflammatory cells in the adventitial compartment, and angiogenic expansion of the vasa vasorum microcirculatory network are observed. These pathophysiological responses suggest that rapidly proliferating vasa vasorum endothelial cells (VVEC) may exhibit increased permeability for circulating blood cells and macromolecules. However, the molecular mechanisms underlying these observations remain unexplored. Some reports implicated extracellular adenosine in the regulation of vascular permeability under hypoxic and inflammatory conditions. Thus, we aimed to determine the role of adenosine in barrier regulation of VVEC isolated from the pulmonary arteries of normoxic (VVEC-Co) or chronically hypoxic (VVEC-Hyp) neonatal calves. Principal Findings We demonstrate via a transendothelial electrical resistance measurement that exogenous adenosine significantly enhanced the barrier function in VVEC-Co and, to a lesser extent, in VVEC-Hyp. Our data from a quantitative reverse transcription polymerase chain reaction show that both VVEC-Co and VVEC-Hyp express all four adenosine receptors (A1, A2A, A2B, and A3), with the highest expression level of A1 receptors (A1Rs). However, A1R expression was significantly lower in VVEC-Hyp compared to VVEC-Co. By using an A1R-specific agonist/antagonist and siRNA, we demonstrate that A1Rs are mostly responsible for adenosine-induced enhancement in barrier function. Adenosine-induced barrier integrity enhancement was attenuated by pretreatment of VVEC with pertussis toxin and GSK690693 or LY294002, suggesting the involvement of Gi proteins and the PI3K-Akt pathway. Moreover, we reveal a critical role of actin cytoskeleton in VVEC barrier regulation by using specific inhibitors of actin and microtubule polymerization. Further, we show that adenosine pretreatment blocked the tumor necrosis factor alpha (TNF-α)-induced permeability in VVEC-Co, validating its anti-inflammatory effects. Conclusions We demonstrate for the first time that stimulation of A1Rs enhances the barrier function in VVEC by activation of the Gi/PI3K/Akt pathway and remodeling of actin microfilament.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2014

High proliferative potential endothelial colony-forming cells contribute to hypoxia-induced pulmonary artery vasa vasorum neovascularization

Hala Nijmeh; Vivek Balasubramaniam; Nana Burns; Aftab Ahmad; Kurt R. Stenmark; Evgenia V. Gerasimovskaya

Angiogenic expansion of the vasa vasorum (VV) is an important contributor to pulmonary vascular remodeling in the pathogenesis of pulmonary hypertension (PH). High proliferative potential endothelial progenitor-like cells have been described in vascular remodeling and angiogenesis in both systemic and pulmonary circulations. However, their role in hypoxia-induced pulmonary artery (PA) VV expansion in PH is not known. We hypothesized that profound PA VV neovascularization observed in a neonatal calf model of hypoxia-induced PH is due to increased numbers of subsets of high proliferative cells within the PA adventitial VV endothelial cells (VVEC). Using a single cell clonogenic assay, we found that high proliferative potential colony-forming cells (HPP-CFC) comprise a markedly higher percentage in VVEC populations isolated from the PA of hypoxic (VVEC-Hx) compared with control (VVEC-Co) calves. VVEC-Hx populations that comprised higher numbers of HPP-CFC also demonstrated markedly higher expression levels of CD31, CD105, and c-kit than VVEC-Co. In addition, significantly higher expression of CD31, CD105, and c-kit was observed in HPP-CFC vs. the VVEC of the control but not of hypoxic animals. HPP-CFC exhibited migratory and tube formation capabilities, two important attributes of angiogenic phenotype. Furthermore, HPP-CFC-Co and some HPP-CFC-Hx exhibited elevated telomerase activity, consistent with their high replicative potential, whereas a number of HPP-CFC-Hx exhibited impaired telomerase activity, suggestive of their senescence state. In conclusion, our data suggest that hypoxia-induced VV expansion involves an emergence of HPP-CFC populations of a distinct phenotype with increased angiogenic capabilities. These cells may serve as a potential target for regulating VVEC neovascularization.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2012

Interplay of macrophages and T cells in the lung vasculature

Evgenia V. Gerasimovskaya; Adelheid Kratzer; Asya Sidiakova; Jonas Salys; Martin R. Zamora; Laimute Taraseviciene-Stewart

In severe pulmonary arterial hypertension (PAH), vascular lesions are composed of phenotypically altered vascular and inflammatory cells that form clusters or tumorlets. Because macrophages are found in increased numbers in intravascular and perivascular space in human PAH, here we address the question whether macrophages play a role in pulmonary vascular remodeling and whether accumulation of macrophages in the lung vasculature could be compromised by the immune system. We used the mouse macrophage cell line RAW 264.7 because these cells are resistant to apoptosis, have high proliferative capacity, and resemble cells in the plexiform lesions that tend to pile up instead of maintaining a monolayer. Cells were characterized by immunocytochemistry with cell surface markers (Lycopersicon Esculentum Lectin, CD117, CD133, FVIII, CD31, VEGFR-2, and S100). Activated, but not quiescent, T cells were able to suppress RAW 264.7 cell proliferative and migration activity in vitro. The carboxyfluorescein diacetate-labeled RAW 264.7 cells were injected into the naïve Sprague Dawley (SD) rat and athymic nude rat. Twelve days later, cells were found in the lung vasculature of athymic nude rats that lack functional T cells, contributing to vascular remodeling. No labeled RAW 264.7 cells were detected in the lungs of immune-competent SD rats. Our data demonstrate that T cells can inhibit in vitro migration and in vivo accumulation of macrophage-like cells.


Vascular Cell | 2011

Complementary effects of extracellular nucleotides and platelet-derived extracts on angiogenesis of vasa vasorum endothelial cells in vitro and subcutaneous Matrigel plugs in vivo.

Hala Nijmeh; Nana Burns; Asya Sidiakova; Kurt R. Stenmark; Evgenia V. Gerasimovskaya

BackgroundPlatelets contribute to vascular homeostasis and angiogenesis through the release of multiple growth factors, cytokines and nucleotides, such as ATP and ADP. Recent reports have demonstrated a marked growth-promoting effect of total platelet extracts and selected platelet growth factors on therapeutic angiogenesis. However, since endogenous adenine nucleotides are rapidly degraded during the platelet isolation and storage, we examined whether supplementing a platelet-derived extract with exogenous adenine nucleotides would augment their pro-angiogenic effects.MethodsPulmonary artery vasa vasorum endothelial cells (VVEC) were used to examine the effects of dialyzed platelet-derived soluble extracts and extracellular adenine nucleotides on proliferation, migration and tube formation. In addition, an in vivo Matrigel plug assay was used to examine the effects of platelet extracts and adenine nucleotides on neovascularization of plugs subcutaneously placed in 50 ICR mice. The number of vascular structures in Matrigel plugs were evaluated by histological and statistical methods.ResultsPlatelet extracts (6.4-64 μg/ml) significantly induced DNA synthesis and at a concentration of 64 μg/ml had a biphasic effect on VVEC proliferation (an increase at 48 hrs followed by a decrease at 60 hrs). Stimulation of VVEC with platelet extracts also significantly (up to several-fold) increased cell migration and tube formation on Matrigel. Stimulation of VVEC with extracellular ATP (100 μM) dramatically (up to ten-fold) increased migration and tube formation on Matrigel; however, no significant effects on cell proliferation were observed. We also found that ATP moderately diminished platelet extract-induced VVEC proliferation (48 hrs) and migration, but potentiated tube formation. Neither ATP, or a mixture of non-hydrolyzable nucleotides (ATPγS, ADPβS, MeSATP, MeSADP) induced vascularization of Matrigel plugs subcutaneously injected in mice, however, the combination of these nucleotides with platelet extracts dramatically increased the number of functional capillaries in the Matrigel plugs.ConclusionData from this study suggest that platelet-derived growth factors and extracellular nucleotides represent important regulatory signals for angiogenesis. Supplementation of platelet extracts with exogenous adenine nucleotides may reveal new possibilities for therapeutic angiogenesis and tissue regeneration approaches.


Archive | 2010

Extracellular ATP and Adenosine as Regulators of Endothelial Cell Function

Evgenia V. Gerasimovskaya; Elzbieta Kaczmarek

Extracellular ATP and adenosine as regulators of endothelial cell function : , Extracellular ATP and adenosine as regulators of endothelial cell function : , کتابخانه دیجیتال جندی شاپور اهواز


American Journal of Physiology-cell Physiology | 2017

Glycolysis and oxidative phosphorylation are essential for purinergic receptor-mediated angiogenic responses in vasa vasorum endothelial cells

Martin Lapel; Philip Weston; Derek Strassheim; Vijaya Karoor; Nana Burns; Taras Lyubchenko; Petr Paucek; Kurt R. Stenmark; Evgenia V. Gerasimovskaya

Angiogenesis is an energy-demanding process; however, the role of cellular energy pathways and their regulation by extracellular stimuli, especially extracellular nucleotides, remain largely unexplored. Using metabolic inhibitors of glycolysis (2-deoxyglucose) and oxidative phosphorylation (OXPHOS) (oligomycin, rotenone, and FCCP), we demonstrate that glycolysis and OXPHOS are both essential for angiogenic responses of vasa vasorum endothelial cell (VVEC). Treatment with P2R agonists, ATP, and 2-methylthioadenosine diphosphate trisodium salt (MeSADP), but not P1 receptor agonist, adenosine, increased glycolytic activity in VVEC (measured by extracellular acidification rate and lactate production). Stimulation of glycolysis was accompanied by increased levels of phospho-phosphofructokinase B3, hexokinase (HK), and GLUT-1, but not lactate dehydrogenase. Moreover, extracellular ATP and MeSADP, and to a lesser extent adenosine, increased basal and maximal oxygen consumption rates in VVEC. These effects were potentiated when the cells were cultured in 20 mM galactose and 5 mM glucose compared with 25 mM glucose. Treatment with P2R agonists decreased phosphorylation of pyruvate dehydrogenase (PDH)-E1α and increased succinate dehydrogenase (SDH), cytochrome oxidase IV, and β-subunit of F1F0 ATP synthase expression. In addition, P2R stimulation transiently elevated mitochondrial Ca2+ concentration, implying involvement of mitochondria in VVEC angiogenic activation. We also demonstrated a critical role of phosphatidylinositol 3-kinase and Akt pathways in lactate production, PDH-E1α phosphorylation, and the expression of HK, SDH, and GLUT-1 in ATP-stimulated VVEC. Together, our findings suggest that purinergic and metabolic regulation of VVEC energy pathways is essential for VV angiogenesis and may contribute to pathologic vascular remodeling in pulmonary hypertension.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2018

Sustained Activation of Rho GTPases Promotes a Synthetic Pulmonary Artery Smooth Muscle Cell Phenotype in Neprilysin Null Mice

Vijaya Karoor; Mehdi A. Fini; Zoe Loomis; Timothy M. Sullivan; Louis B. Hersh; Evgenia V. Gerasimovskaya; David Irwin; Edward C. Dempsey

Objective— Pulmonary artery smooth muscle cells (PASMCs) from neprilysin (NEP) null mice exhibit a synthetic phenotype and increased activation of Rho GTPases compared with their wild-type counterparts. Although Rho GTPases are known to promote a contractile SMC phenotype, we hypothesize that their sustained activity decreases SM-protein expression in these cells. Approach and Results— PASMCs isolated from wild-type and NEP−/− mice were used to assess levels of SM-proteins (SM-actin, SM-myosin, SM22, and calponin) by Western blotting, and were lower in NEP−/− PASMCs compared with wild-type. Rac and Rho (ras homology family member) levels and activity were higher in NEP−/− PASMCs, and ShRNA to Rac and Rho restored SM-protein, and attenuated the enhanced migration and proliferation of NEP−/− PASMCs. SM-gene repressors, p-Elk-1, and Klf4 (Kruppel lung factor 4), were higher in NEP−/− PASMCs and decreased by shRNA to Rac and Rho. Costimulation of wild-type PASMCs with PDGF (platelet-derived growth factor) and the NEP substrate, ET-1 (endothelin-1), increased Rac and Rho activity, and decreased SM-protein levels mimicking the NEP knock-out phenotype. Activation of Rac and Rho and downstream effectors was observed in lung tissue from NEP−/− mice and humans with chronic obstructive pulmonary disease. Conclusions— Sustained Rho activation in NEP−/− PASMCs is associated with a decrease in SM-protein levels and increased migration and proliferation. Inactivation of RhoGDI (Rho guanine dissociation inhibitor) and RhoGAP (Rho GTPase activating protein) by phosphorylation may contribute to prolonged activation of Rho in NEP−/− PASMCs. Rho GTPases may thus have a role in integration of signals between vasopeptides and growth factor receptors and could influence pathways that suppress SM-proteins to promote a synthetic phenotype.

Collaboration


Dive into the Evgenia V. Gerasimovskaya's collaboration.

Top Co-Authors

Avatar

Kurt R. Stenmark

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Maria G. Frid

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nana Burns

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Suzette R. Riddle

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Adil Anwar

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Heather Woodward

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Elzbieta Kaczmarek

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hala Nijmeh

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge