Network


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

Hotspot


Dive into the research topics where Elena Karnaukhova is active.

Publication


Featured researches published by Elena Karnaukhova.


The Journal of Clinical Pharmacology | 2008

New Era in Drug Interaction Evaluation: US Food and Drug Administration Update on CYP Enzymes, Transporters, and the Guidance Process

Shiew Mei Huang; John M. Strong; Lei Zhang; Kellie S. Reynolds; Srikanth Nallani; Robert Temple; Sophia Abraham; Sayed Al Habet; Raman K. Baweja; Gilbert J. Burckart; Sang Chung; Philip Colangelo; David Frucht; Martin David Green; Paul Hepp; Elena Karnaukhova; Hon Sum Ko; Jang Ik Lee; Patrick J. Marroum; Janet Norden; Wei Qiu; Atiqur Rahman; Solomon Sobel; Toni Stifano; Kenneth E. Thummel; Xiao Xiong Wei; Sally Yasuda; Jenny H. Zheng; Hong Zhao; Lawrence J. Lesko

Predicting clinically significant drug interactions during drug development is a challenge for the pharmaceutical industry and regulatory agencies. Since the publication of the US Food and Drug Administrations (FDAs) first in vitro and in vivo drug interaction guidance documents in 1997 and 1999, researchers and clinicians have gained a better understanding of drug interactions. This knowledge has enabled the FDA and the industry to progress and begin to overcome these challenges. The FDA has continued its efforts to evaluate methodologies to study drug interactions and communicate recommendations regarding the conduct of drug interaction studies, particularly for CYP‐based and transporter‐based drug interactions, to the pharmaceutical industry. A drug interaction Web site was established to document the FDAs current understanding of drug interactions (http:www.fda.govcderdrugdrugInteractionsdefault.htm). This report provides an overview of the evolution of the drug interaction guidances, includes a synopsis of the steps taken by the FDA to revise the original drug interaction guidance documents, and summarizes and highlights updated sections in the current guidance document, Drug Interaction Studies—Study Design, Data Analysis, and Implications for Dosing and Labeling.


Amino Acids | 2006

Recombinant human alpha-1 proteinase inhibitor: towards therapeutic use

Elena Karnaukhova; Yakir Ophir; Basil Golding

Summary.Human alpha-1-proteinase inhibitor is a well-characterized protease inhibitor with a wide spectrum of anti-protease activity. Its major physiological role is inhibition of neutrophil elastase in the lungs, and its deficiency is associated with progressive ultimately fatal emphysema. Currently in the US, only plasma-derived human alpha-1-proteinase inhibitor is available for augmentation therapy, which appears to be insufficient to meet the anticipated clinical demand. Moreover, despite effective viral clearance steps in the manufacturing process, the potential risk of contamination with new and unknown pathogens still exists. In response, multiple efforts to develop recombinant versions of human alpha-1-proteinase inhibitor, as an alternative to the plasma-derived protein, have been reported. Over the last two decades, various systems have been used to express the human gene for alpha-1-proteinase inhibitor. This paper reviews the recombinant versions of human alpha-1-proteinase inhibitor produced in various hosts, considers current major safety and efficacy issues regarding recombinant glycoproteins as potential therapeutics, and the factors that are impeding progress in this area1.


Biomaterials | 2014

The effect of protein corona composition on the interaction of carbon nanotubes with human blood platelets

Silvia H. De Paoli; Lukas Diduch; Tseday Z. Tegegn; Martina Orecna; Michael Brad Strader; Elena Karnaukhova; John E. Bonevich; Karel Holada; Jan Simak

Carbon nanotubes (CNT) are one of the most promising nanomaterials for use in medicine. The blood biocompatibility of CNT is a critical safety issue. In the bloodstream, proteins bind to CNT through non-covalent interactions to form a protein corona, thereby largely defining the biological properties of the CNT. Here, we characterize the interactions of carboxylated-multiwalled carbon nanotubes (CNTCOOH) with common human proteins and investigate the effect of the different protein coronas on the interaction of CNTCOOH with human blood platelets (PLT). Molecular modeling and different photophysical techniques were employed to characterize the binding of albumin (HSA), fibrinogen (FBG), γ-globulins (IgG) and histone H1 (H1) on CNTCOOH. We found that the identity of protein forming the corona greatly affects the outcome of CNTCOOHs interaction with blood PLT. Bare CNTCOOH-induced PLT aggregation and the release of platelet membrane microparticles (PMP). HSA corona attenuated the PLT aggregating activity of CNTCOOH, while FBG caused the agglomeration of CNTCOOH nanomaterial, thereby diminishing the effect of CNTCOOH on PLT. In contrast, the IgG corona caused PLT fragmentation, and the H1 corona induced a strong PLT aggregation, thus potentiating the release of PMP.


Physical Chemistry Chemical Physics | 2013

Copper(II)–human amylin complex protects pancreatic cells from amylin toxicity

Elizabeth C. Lee; Emmeline Ha; Sanghamitra Singh; Linda Legesse; Sana Ahmad; Elena Karnaukhova; Robert P. Donaldson; Aleksandar Jeremic

Human amylin-derived oligomers and aggregates are believed to play an important role in the pathogenesis of type II diabetes mellitus (T2DM). In addition to amylin-evoked cell attrition, T2DM is often accompanied by elevated serum copper levels. Although previous studies have shown that human amylin, in the course of its aggregation, produces hydrogen peroxide (H2O2) in solution, and that this process is exacerbated in the presence of copper(ii) ions (Cu(2+)), very little is known about the mechanism of interaction between Cu(2+) and amylin in pancreatic β-cells, including its pathological significance. Hence, in this study we investigated the mechanism by which Cu(2+) and human amylin catalyze formation of reactive oxygen species (ROS) in cells and in vitro, and examined the modulatory effect of Cu(2+) on amylin aggregation and toxicity in pancreatic rat insulinoma (RIN-m5F) β-cells. Our results indicate that Cu(2+) interacts with human and rat amylin to form metalo-peptide complexes with low aggregative and oxidative properties. Human and non-amyloidogenic rat amylin produced minute (nM) amounts of H2O2, the accumulation of which was slightly enhanced in the presence of Cu(2+). In a marked contrast to human and rat amylin, and in the presence of the reducing agents glutathione and ascorbate, Cu(2+) produced μM concentrations of H2O2 surpassing the amylin effect by several fold. The current study shows that human and rat amylin not only produce but also quench H2O2, and that human but not rat amylin significantly decreases the amount of H2O2 in solution produced by Cu(2+) and glutathione. Similarly, human amylin was found to also decrease hydroxyl radical formation elicited by Cu(2+) and glutathione. Furthermore, Cu(2+) mitigated the toxic effect of human amylin by inhibiting activation of pro-apoptotic caspase-3 and stress-kinase signaling pathways in rat pancreatic insulinoma cells in part by stabilizing human amylin in its native conformational state. This sacrificial quenching of metal-catalyzed ROS by human amylin and coppers anti-aggregative and anti-apoptotic properties suggest a novel and protective role for the copper-amylin complex.


Biotechnology and Bioengineering | 2009

Production, purification, and characterization of human α1 proteinase inhibitor from Aspergillus niger

Liat Chill; Loc Trinh; Parastoo Azadi; Mayumi Ishihara; Roberto Sonon; Elena Karnaukhova; Yakir Ophir; Basil Golding; Joseph Shiloach

Human alpha one proteinase inhibitor (α1‐PI) was cloned and expressed in Aspergillus niger, filamentious fungus that can grow in defined media and can perform glycosylation. Submerged culture conditions were established using starch as carbon source, 30% dissolved oxygen concentration, pH 7.0 and 28°C. Eight milligrams per liter of active α1‐PI were secreted to the growth media in about 40 h. Controlling the protein proteolysis was found to be an important factor in the production. The effects of various carbon sources, pH and temperature on the production and stability of the protein were tested and the product was purified and characterized. Two molecular weights variants of the recombinant α1‐PI were produced by the fungus; the difference is attributed to the glycosylated part of the molecule. The two glycoproteins were treated with PNGAse F and the released glycans were analyzed by HPAEC, MALDI/TOF‐MS, NSI‐MSn, and GC‐MS. The MALDI and NSI‐ full MS spectra of permethylated N‐glycans revealed that the N‐glycans of both variants contain a series of high‐mannose type glycans with 5–20 hexose units. Monosaccharide analysis showed that these were composed of N‐acetylglucos‐amine, mannose, and galactose. Linkage analysis revealed that the galactosyl component was in the furanoic conformation, which was attaching in a terminal non‐reducing position. The Galactofuranose‐containing high‐mannnose type N‐glycans are typical structures, which recently have been found as part of several glycoproteins produced by Aspergillus niger. Biotechnol. Bioeng. 2009; 102: 828–844.


Biochimica et Biophysica Acta | 2008

Human cellular prion protein interacts directly with clusterin protein.

Fei Xu; Elena Karnaukhova; Jaroslav G. Vostal

Prion protein is a glycosyl-phosphatidyl-inositol anchored glycoprotein localized on the surface and within a variety of cells. Its conformation change is thought to be essential for the proliferation of prion neurodegenerative diseases. Using the yeast two-hybrid assay we identified an interaction between prion protein and clusterin, a chaperone glycoprotein. This interaction was confirmed in a mammalian system by in vivo co-immunoprecipitation and in vitro by circular dichroism analysis. Through deletion mapping analysis we demonstrated that the alpha subunit, but not the beta subunit, of clusterin binds to prion and that the C-terminal 62 amino acid segment of the putative alpha helix region of clusterin is essential for the binding interaction. The full prion protein as well as the N-terminal section (aa 23-95) and C-terminal (aa 96-231) were shown to interact with clusterin. These findings provide new insights into the molecular mechanisms of interaction between prion and clusterin protein and contribute to the understanding of prion proteins physiological function.


Frontiers in Physiology | 2014

Characterization of heme binding to recombinant α1-microglobulin

Elena Karnaukhova; Sigurbjörg Rutardottir; Mohsen Rajabi; Lena Wester Rosenlöf; Abdu I. Alayash; Bo Åkerström

Background: Alpha-1-microglobulin (A1M), a small lipocalin protein found in plasma and tissues, has been identified as a heme1 and radical scavenger that may participate in the mitigation of toxicities caused by degradation of hemoglobin. The objective of this work was to investigate heme interactions with A1M in vitro using various analytical techniques and to optimize analytical methodology suitable for rapid evaluation of the ligand binding properties of recombinant A1M versions. Methods: To examine heme binding properties of A1M we utilized UV/Vis absorption spectroscopy, visible circular dichroism (CD), catalase-like activity, migration shift electrophoresis, and surface plasmon resonance (SPR), which was specifically developed for the assessment of His-tagged A1M. Results: The results of this study confirm that A1M is a heme binding protein that can accommodate heme at more than one binding site and/or in coordination with different amino acid residues depending upon heme concentration and ligand-to-protein molar ratio. UV/Vis titration of A1M with heme revealed an unusually large bathochromic shift, up to 38 nm, observed for heme binding to a primary binding site. UV/Vis spectroscopy, visible CD and catalase-like activity suggested that heme is accommodated inside His-tagged (tgA1M) and tagless A1M (ntA1M) in a rather similar fashion although the His-tag is very likely involved into coordination with iron of the heme molecule. SPR data indicated kinetic rate constants and equilibrium binding constants with KD values in a μM range. Conclusions: This study provided experimental evidence of the A1M heme binding properties by aid of different techniques and suggested an analytical methodology for a rapid evaluation of ligand-binding properties of recombinant A1M versions, also suitable for other His-tagged proteins.


Journal of Thrombosis and Haemostasis | 2017

Expression and characterization of a codon-optimized blood coagulation factor VIII

Svetlana A. Shestopal; J.-J. Hao; Elena Karnaukhova; Yideng Liang; Mikhail V. Ovanesov; M. Lin; James H. Kurasawa; Timothy K. Lee; John H. McVey; Andrey G. Sarafanov

Essentials Recombinant factor VIII (FVIII) is known to be expressed at a low level in cell culture. To increase expression, we used codon‐optimization of a B‐domain deleted FVIII (BDD‐FVIII). This resulted in 7‐fold increase of the expression level in cell culture. The biochemical properties of codon‐optimized BDD‐FVIII were similar to the wild‐type protein.


PLOS ONE | 2012

Oversulfated Chondroitin Sulfate Inhibits the Complement Classical Pathway by Potentiating C1 Inhibitor

Zhao-Hua Zhou; Mohsen Rajabi; Trina Chen; Elena Karnaukhova; Steven Kozlowski

Oversulfated chondroitin sulfate (OSCS) has become the subject of multidisciplinary investigation as a non-traditional contaminant in the heparin therapeutic preparations that were linked to severe adverse events. In this study, it was found that OSCS inhibited complement fixation on bacteria and bacterial lysis mediated by the complement classical pathway. The inhibition of complement by OSCS is not due to interference with antibody/antigen interaction or due to consumption of C3 associated with FXII-dependent contact system activation. However, OSCS complement inhibition is dependent on C1 inhibitor (C1inh) since the depletion of C1inh from either normal or FXII-deficient complement plasma prevents OSCS inhibition of complement activity. Surface plasmon resonance measurements revealed that immobilized C1inhibitor bound greater than 5-fold more C1s in the presence of OSCS than in presence of heparin. Although heparin can also inhibit complement, OSCS and OSCS contaminated heparin are more potent inhibitors of complement. Furthermore, polysulfated glycosaminoglycan (PSGAG), an anti-inflammatory veterinary medicine with a similar structure to OSCS, also inhibited complement in the plasma of dogs and farm animals. This study provides a new insight that in addition to the FXII-dependent activation of contact system, oversulfated and polysulfated chondroitin-sulfate can inhibit complement activity by potentiating the classical complement pathway regulator C1inh. This effect on C1inh may play a role in inhibiting inflammation as well as impacting bacterial clearance.


Biochimica et Biophysica Acta | 2012

Potentiation of C1-esterase inhibitor by heparin and interactions with C1s protease as assessed by surface plasmon resonance.

Mohsen Rajabi; Evi Struble; Zhaohua Zhou; Elena Karnaukhova

BACKGROUND Human C1-esterase inhibitor (C1-INH) is a multifunctional plasma protein with a wide range of inhibitory and non-inhibitory properties, mainly recognized as a key down-regulator of the complement and contact cascades. The potentiation of C1-INH by heparin and other glycosaminoglycans (GAGs) regulates a broad spectrum of C1-INH activities in vivo both in normal and disease states. SCOPE OF RESEARCH: We have studied the potentiation of human C1-INH by heparin using Surface Plasmon Resonance (SPR), circular dichroism (CD) and a functional assay. To advance a SPR for multiple-unit interaction studies of C1-INH we have developed a novel (consecutive double capture) approach exploring different immobilization and layout. MAJOR CONCLUSIONS Our SPR experiments conducted in three different design versions showed marked acceleration in C1-INH interactions with complement protease C1s as a result of potentiation of C1-INH by heparin (from 5- to 11-fold increase of the association rate). Far-UV CD studies suggested that heparin binding did not alter C1-INH secondary structure. Functional assay using chromogenic substrate confirmed that heparin does not affect the amidolytic activity of C1s, but does accelerate its consumption due to C1-INH potentiation. GENERAL SIGNIFICANCE This is the first report that directly demonstrates a significant acceleration of the C1-INH interactions with C1s due to heparin by using a consecutive double capture SPR approach. The results of this study may be useful for further C-INH therapeutic development, ultimately for the enhancement of current C1-INH replacement therapies.

Collaboration


Dive into the Elena Karnaukhova's collaboration.

Top Co-Authors

Avatar

Basil Golding

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Mohsen Rajabi

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Yakir Ophir

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Andrey G. Sarafanov

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

James H. Kurasawa

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Svetlana A. Shestopal

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Timothy K. Lee

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Abdu I. Alayash

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Joseph Shiloach

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Loc Trinh

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge