Network


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

Hotspot


Dive into the research topics where Olga Rafikova is active.

Publication


Featured researches published by Olga Rafikova.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Catalysis of S-nitrosothiols formation by serum albumin: the mechanism and implication in vascular control.

Olga Rafikova; Ruslan Rafikov; Evgeny Nudler

Nitric oxide (NO⋅) is a short-lived physiological messenger. Its various biological activities can be preserved in a more stable form of S-nitrosothiols (RS-NO). Here we demonstrate that at physiological NO⋅ concentrations, plasma albumin becomes saturated with NO⋅ and accelerates formation of low-molecular-weight (LMW) RS-NO in vitro and in vivo. The mechanism involves micellar catalysis of NO⋅ oxidation in the albumin hydrophobic core and specific transfer of NO+ to LMW thiols. Albumin-mediated S-nitrosylation and its vasodilatory effect directly depend on the concentration of circulating LMW thiols. Results suggest that the hydrophobic phase formed by albumin serves as a major reservoir of NO⋅ and its reactive oxides and controls the dynamics of NO⋅-dependant processes in the vasculature.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

NADPH Oxidase 4 Is Expressed in Pulmonary Artery Adventitia and Contributes to Hypertensive Vascular Remodeling

Scott A. Barman; Feng Chen; Yunchao Su; Christiana Dimitropoulou; Yusi Wang; John D. Catravas; Weihong Han; Laszlo Orfi; Csaba Szántai-Kis; György Kéri; István Szabadkai; Nektarios Barabutis; Olga Rafikova; Ruslan Rafikov; Stephen M. Black; Danny Jonigk; Athanassios Giannis; Reto Asmis; David W. Stepp; Ganesan Ramesh; David J.R. Fulton

Objective— Pulmonary hypertension (PH) is a progressive disease arising from remodeling and narrowing of pulmonary arteries (PAs) resulting in high pulmonary blood pressure and ultimately right ventricular failure. Elevated production of reactive oxygen species by NADPH oxidase 4 (Nox4) is associated with increased pressure in PH. However, the cellular location of Nox4 and its contribution to aberrant vascular remodeling in PH remains poorly understood. Therefore, we sought to identify the vascular cells expressing Nox4 in PAs and determine the functional relevance of Nox4 in PH. Approach and Results— Elevated expression of Nox4 was detected in hypertensive PAs from 3 rat PH models and human PH using qualititative real-time reverse transcription polymerase chain reaction, Western blot, and immunofluorescence. In the vascular wall, Nox4 was detected in both endothelium and adventitia, and perivascular staining was prominently increased in hypertensive lung sections, colocalizing with cells expressing fibroblast and monocyte markers and matching the adventitial location of reactive oxygen species production. Small-molecule inhibitors of Nox4 reduced adventitial reactive oxygen species generation and vascular remodeling as well as ameliorating right ventricular hypertrophy and noninvasive indices of PA stiffness in monocrotaline-treated rats as determined by morphometric analysis and high-resolution digital ultrasound. Nox4 inhibitors improved PH in both prevention and reversal protocols and reduced the expression of fibroblast markers in isolated PAs. In fibroblasts, Nox4 overexpression stimulated migration and proliferation and was necessary for matrix gene expression. Conclusion— These findings indicate that Nox4 is prominently expressed in the adventitia and contributes to altered fibroblast behavior, hypertensive vascular remodeling, and development of PH.


Free Radical Biology and Medicine | 2013

Bosentan inhibits oxidative and nitrosative stress and rescues occlusive pulmonaryhypertension

Olga Rafikova; Ruslan Rafikov; Sanjiv Kumar; Shruti Sharma; Saurabh Aggarwal; Frank Schneider; Danny Jonigk; Stephen M. Black; Stevan P. Tofovic

Pulmonary arterial hypertension (PH) is a fatal disease marked by excessive pulmonary vascular cell proliferation. Patients with idiopathic PH express endothelin-1 (ET-1) at high levels in their lungs. As the activation of both types of ET-1 receptor (ETA and ETB) leads to increased generation of superoxide and hydrogen peroxide, this may contribute to the severe oxidative stress found in PH patients. As a number of pathways may induce oxidative stress, the particular role of ET-1 remains unclear. The aim of this study was to determine whether inhibition of ET-1 signaling could reduce pulmonary oxidative stress and attenuate the progression of disease in rats with occlusive-angioproliferative PH induced by a single dose of SU5416 (200 mg/kg) and subsequent exposure to hypoxia for 21 days. Using this regimen, animals developed severe PH as evidenced by a progressive increase in right-ventricle (RV) peak systolic pressure (RVPSP), severe RV hypertrophy, and pulmonary endothelial and smooth muscle cell proliferation, resulting in plexiform vasculopathy. PH rats also had increased oxidative stress, correlating with endothelial nitric oxide synthase uncoupling and NADPH oxidase activation, leading to enhanced protein nitration and increases in markers of vascular remodeling. Treatment with the combined ET receptor antagonist bosentan (250 mg/kg/day; day 10 to 21) prevented further increase in RVPSP and RV hypertrophy, decreased ETA/ETB protein levels, reduced oxidative stress and protein nitration, and resulted in marked attenuation of pulmonary vascular cell proliferation. We conclude that inhibition of ET-1 signaling significantly attenuates the oxidative and nitrosative stress associated with PH and prevents its progression.


Journal of Biological Chemistry | 2013

Asymmetric Dimethylarginine Induces Endothelial Nitric Oxide Synthase Mitochondrial Redistribution through the nitration-mediated activation of Akt1

Ruslan Rafikov; Olga Rafikova; Saurabh Aggarwal; Christine Gross; Xutong Sun; Julin Desai; David Fulton; Stephen M. Black

Background: Asymmetric dimethylarginine (ADMA) can induce endothelial nitric-oxide synthase (eNOS) redistribution from the plasma membrane to the mitochondria. Results: AMDA induces nitration of Akt1 at Tyr350 within the client-binding domain, increasing its activation and enhancing eNOS phosphorylation. Conclusion: Under physiologic conditions, Akt1-mediated redistribution of eNOS to the mitochondria enhances mitochondrial coupling. Significance: Reducing Akt1 nitration may reduce the deleterious effects of Akt1 signaling in various pathologies. We have recently demonstrated that asymmetric dimethylarginine (ADMA) induces the translocation of endothelial nitric-oxide synthase (eNOS) to the mitochondrion via a mechanism that requires protein nitration. Thus, the goal of this study was elucidate how eNOS redistributes to mitochondria and to identify the nitrated protein responsible for this event. Our data indicate that exposure of pulmonary arterial endothelial cells to ADMA enhanced eNOS phosphorylation at the Akt1-dependent phosphorylation sites Ser617 and Ser1179. Mutation of these serine residues to alanine (S617A and S1179A) inhibited nitration-mediated eNOS translocation to the mitochondria, whereas the phosphormimic mutations (S617D and S1179D) exhibited increased mitochondrial redistribution in the absence of ADMA. The overexpression of a dominant-negative Akt1 also attenuated ADMA-mediated eNOS mitochondrial translocation. Furthermore, ADMA enhanced Akt1 nitration and increased its activity. Mass spectrometry identified a single nitration site in Akt1 located at the tyrosine residue (Tyr350) located within the client-binding domain. Replacement of Tyr350 with phenylalanine abolished peroxynitrite-mediated eNOS translocation to mitochondria. We also found that in the absence of ADMA, eNOS translocation decreased mitochondrial oxygen consumption and superoxide production without altering cellular ATP level. This suggests that under physiologic conditions, eNOS translocation enhances mitochondria coupling. In conclusion, we have identified a new mechanism by which eNOS translocation to mitochondria is regulated by the phosphorylation of eNOS at Ser617 and Ser1179 by Akt1 and that this is enhanced when Akt1 becomes nitrated at Tyr350.


American Journal of Respiratory Cell and Molecular Biology | 2014

Endothelin-1 Induces a Glycolytic Switch in Pulmonary Arterial Endothelial Cells via the Mitochondrial Translocation of Endothelial Nitric Oxide Synthase

Xutong Sun; Sanjiv Kumar; Shruti Sharma; Saurabh Aggarwal; Qing Shi Lu; Christine Gross; Olga Rafikova; Sung Gon Lee; Sridevi Dasarathy; Yali Hou; Mary L. Meadows; Weihong Han; Yunchao Su; Jeffrey R. Fineman; Stephen M. Black

Recent studies have indicated that, during the development of pulmonary hypertension (PH), there is a switch from oxidative phosphorylation to glycolysis in the pulmonary endothelium. However, the mechanisms underlying this phenomenon have not been elucidated. Endothelin (ET)-1, an endothelial-derived vasoconstrictor peptide, is increased in PH, and has been shown to play an important role in the oxidative stress associated with PH. Thus, in this study, we investigated whether there was a potential link between increases in ET-1 and mitochondrial remodeling. Our data indicate that ET-1 induces the redistribution of endothelial nitric oxide synthase (eNOS) from the plasma membrane to the mitochondria in pulmonary arterial endothelial cells, and that this was dependent on eNOS uncoupling. We also found that ET-1 disturbed carnitine metabolism, resulting in the attenuation of mitochondrial bioenergetics. However, ATP levels were unchanged due to a compensatory increase in glycolysis. Further mechanistic investigations demonstrated that ET-1 mediated the redistribution of eNOS via the phosphorylation of eNOS at Thr495 by protein kinase C δ. In addition, the glycolytic switch appeared to be dependent on mitochondrial-derived reactive oxygen species that led to the activation of hypoxia-inducible factor signaling. Finally, the cell culture data were confirmed in vivo using the monocrotaline rat model of PH. Thus, we conclude that ET-1 induces a glycolytic switch in pulmonary arterial endothelial cells via the redistribution of uncoupled eNOS to the mitochondria, and that preventing this event may be an approach for the treatment of PH.


Redox biology | 2015

Complex I dysfunction underlies the glycolytic switch in pulmonary hypertensive smooth muscle cells.

Ruslan Rafikov; Xutong Sun; Olga Rafikova; Mary L. Meadows; Ankit A. Desai; Zain Khalpey; Jason X.-J. Yuan; Jeffrey R. Fineman; Stephen M. Black

ATP is essential for cellular function and is usually produced through oxidative phosphorylation. However, mitochondrial dysfunction is now being recognized as an important contributing factor in the development cardiovascular diseases, such as pulmonary hypertension (PH). In PH there is a metabolic change from oxidative phosphorylation to mainly glycolysis for energy production. However, the mechanisms underlying this glycolytic switch are only poorly understood. In particular the role of the respiratory Complexes in the mitochondrial dysfunction associated with PH is unresolved and was the focus of our investigations. We report that smooth muscle cells isolated from the pulmonary vessels of rats with PH (PH-PASMC), induced by a single injection of monocrotaline, have attenuated mitochondrial function and enhanced glycolysis. Further, utilizing a novel live cell assay, we were able to demonstrate that the mitochondrial dysfunction in PH-PASMC correlates with deficiencies in the activities of Complexes I–III. Further, we observed that there was an increase in mitochondrial reactive oxygen species generation and mitochondrial membrane potential in the PASMC isolated from rats with PH. We further found that the defect in Complex I activity was due to a loss of Complex I assembly, although the assembly of Complexes II and III were both maintained. Thus, we conclude that loss of Complex I assembly may be involved in the switch of energy metabolism in smooth muscle cells to glycolysis and that maintaining Complex I activity may be a potential therapeutic target for the treatment of PH.


PLOS ONE | 2016

Metabolic Changes Precede the Development of Pulmonary Hypertension in the Monocrotaline Exposed Rat Lung

Olga Rafikova; Mary L. Meadows; Jason M. Kinchen; Robert P. Mohney; Emin Maltepe; Ankit A. Desai; Jason X.-J. Yuan; Joe G. N. Garcia; Jeffrey R. Fineman; Ruslan Rafikov; Stephen M. Black

There is increasing interest in the potential for metabolic profiling to evaluate the progression of pulmonary hypertension (PH). However, a detailed analysis of the metabolic changes in lungs at the early stage of PH, characterized by increased pulmonary artery pressure but prior to the development of right ventricle hypertrophy and failure, is lacking in a preclinical animal model of PH. Thus, we undertook a study using rats 14 days after exposure to monocrotaline (MCT), to determine whether we could identify early stage metabolic changes prior to the manifestation of developed PH. We observed changes in multiple pathways associated with the development of PH, including activated glycolysis, increased markers of proliferation, disruptions in carnitine homeostasis, increased inflammatory and fibrosis biomarkers, and a reduction in glutathione biosynthesis. Further, our global metabolic profile data compare favorably with prior work carried out in humans with PH. We conclude that despite the MCT-model not recapitulating all the structural changes associated with humans with advanced PH, including endothelial cell proliferation and the formation of plexiform lesions, it is very similar at a metabolic level. Thus, we suggest that despite its limitations it can still serve as a useful preclinical model for the study of PH.


Pulmonary circulation | 2015

The sexual dimorphism associated with pulmonary hypertension corresponds to a fibrotic phenotype

Olga Rafikova; Ruslan Rafikov; Mary L. Meadows; Archana Kangath; Danny Jonigk; Stephen M. Black

Although female predominance in the development of all types of pulmonary hypertension (PH) is well established, many clinical studies have confirmed that females have better prognosis and higher survival rate than males. There is no clear explanation of why sex influences the pathogenesis and progression of PH. Using a rat angioproliferative model of PH, which closely resembles the primary pathological changes observed in humans, we evaluated the role of sex in the development and progression of PH. Female rats had a more pronounced increase in medial thickness in the small pulmonary arteries. However, the infiltration of small pulmonary arteries by inflammatory cells was found only in male rats, and this corresponded to increased myeloperoxidase activity and abundant adventitial and medial fibrosis that were not present in female rats. Although the level of right ventricle (RV) peak systolic pressure was similar in both groups, the survival rate in male rats was significantly lower. Moreover, male rats presented with a more pronounced increase in RV thickness that correlated with diffuse RV fibrosis and significantly impaired right cardiac function. The reduction in fibrosis in female rats correlated with increased expression of caveolin-1 and reduced endothelial nitric oxide synthase–derived superoxide. We conclude that, in the pathogenesis of PH, female sex is associated with greater remodeling of the pulmonary arteries but greater survival. Conversely, in males, the development of pulmonary and cardiac fibrosis leads to early and severe RV failure, and this may be an important reason for the lower survival rate among males.


Free Radical Biology and Medicine | 2016

Redox regulation of epidermal growth factor receptor signaling during the development of pulmonary hypertension

Olga Rafikova; Ruslan Rafikov; Archana Kangath; Ning Qu; Saurabh Aggarwal; Shruti Sharma; Julin Desai; Taylor Fields; Britta Ludewig; Jason X.-Y. Yuan; Danny Jonigk; Stephen M. Black

The development of pulmonary hypertension (PH) involves the uncontrolled proliferation of pulmonary smooth muscle cells via increased growth factor receptor signaling. However, the role of epidermal growth factor receptor (EGFR) signaling is controversial, as humans with advanced PH exhibit no changes in EGFR protein levels and purpose of the present study was to determine whether there are post-translational mechanisms that enhance EGFR signaling in PH. The EGFR inhibitor, gefinitib, significantly attenuated EGFR signaling and prevented the development of PH in monocrotaline (MCT)-exposed rats, confirming the contribution of EGFR activation in MCT induced PH. There was an early MCT-mediated increase in hydrogen peroxide, which correlated with the binding of the active metabolite of MCT, monocrotaline pyrrole, to catalase Cys377, disrupting its multimeric structure. This early oxidative stress was responsible for the oxidation of EGFR and the formation of sodium dodecyl sulfate (SDS) stable EGFR dimers through dityrosine cross-linking. These cross-linked dimers exhibited increased EGFR autophosphorylation and signaling. The activation of EGFR signaling did not correlate with pp60(src) dependent Y845 phosphorylation or EGFR ligand expression. Importantly, the analysis of patients with advanced PH revealed the same enhancement of EGFR autophosphorylation and covalent dimer formation in pulmonary arteries, while total EGFR protein levels were unchanged. As in the MCT exposed rat model, the activation of EGFR in human samples was independent of pp60(src) phosphorylation site and ligand expression. This study provides a novel molecular mechanism of oxidative stress stimulated covalent EGFR dimerization via tyrosine dimerization that contributes into development of PH.


Biochemical Pharmacology | 2017

RhoA S-nitrosylation as a regulatory mechanism influencing endothelial barrier function in response to G+-bacterial toxins

Feng Chen; Y. Wang; Ruslan Rafikov; S. Haigh; W.B. Zhi; Sanjiv Kumar; Paschalis-Thomas Doulias; Olga Rafikova; H. Pillich; Trinad Chakraborty; Rudolf Lucas; Alexander D. Verin; John D. Catravas; J.X. She; Stephen M. Black; D.J.R. Fulton

Graphical abstract Figure. No Caption available. ABSTRACT Disruption of the endothelial barrier in response to Gram positive (G+) bacterial toxins is a major complication of acute lung injury (ALI) and can be further aggravated by antibiotics which stimulate toxin release. The integrity of the pulmonary endothelial barrier is mediated by the balance of disruptive forces such as the small GTPase RhoA, and protective forces including endothelium‐derived nitric oxide (NO). How NO protects against the barrier dysfunction is incompletely understood and our goal was to determine whether NO and S‐nitrosylation can modulate RhoA activity and whether this mechanism is important for G+ toxin‐induced microvascular permeability. We found that the G+ toxin listeriolysin‐O (LLO) increased RhoA activity and that NO and S‐NO donors inhibit RhoA activity. RhoA was robustly S‐nitrosylated as determined by biotin‐switch and mercury column analysis. MS revealed that three primary cysteine residues are S‐nitrosylated including cys16, cys20 and cys159. Mutation of these residues to serine diminished S‐nitrosylation to endogenous NO and mutant RhoA was less sensitive to inhibition by S‐NO. G+‐toxins stimulated the denitrosylation of RhoA which was not mediated by S‐nitrosoglutathione reductase (GSNOR), thioredoxin (TRX) or thiol‐dependent enzyme activity but was instead stimulated directly by elevated calcium levels. Calcium‐promoted the direct denitrosylation of WT but not mutant RhoA and mutant RhoA adenovirus was more effective than WT in disrupting the barrier integrity of human lung microvascular endothelial cells. In conclusion, we reveal a novel mechanism by which NO and S‐nitrosylation reduces RhoA activity which may be of significance in the management of pulmonary endothelial permeability induced by G+‐toxins.

Collaboration


Dive into the Olga Rafikova's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Saurabh Aggarwal

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Danny Jonigk

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary L. Meadows

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Sanjiv Kumar

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Shruti Sharma

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Christine Gross

Georgia Regents University

View shared research outputs
Researchain Logo
Decentralizing Knowledge