Network


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

Hotspot


Dive into the research topics where Tatiana N. Sidorova is active.

Publication


Featured researches published by Tatiana N. Sidorova.


Journal of Molecular and Cellular Cardiology | 2015

Reactive γ-Ketoaldehydes Promote Protein Misfolding and Preamyloid Oligomer Formation in Rapidly-Activated Atrial Cells

Tatiana N. Sidorova; Liudmila V. Yermalitskaya; Lisa C. Mace; K. Sam Wells; Olivier Boutaud; Joseph K. Prinsen; Sean S. Davies; L. Jackson Roberts; Sergey Dikalov; Charles G. Glabe; Venkataraman Amarnath; Joey V. Barnett; Katherine T. Murray

Rapid activation causes remodeling of atrial myocytes resembling that which occurs in experimental and human atrial fibrillation (AF). Using this cellular model, we previously observed transcriptional upregulation of proteins implicated in protein misfolding and amyloidosis. For organ-specific amyloidoses such as Alzheimers disease, preamyloid oligomers (PAOs) are now recognized to be the primary cytotoxic species. In the setting of oxidative stress, highly-reactive lipid-derived mediators known as γ-ketoaldehydes (γ-KAs) have been identified that rapidly adduct proteins and cause PAO formation for amyloid β1-42 implicated in Alzheimers. We hypothesized that rapid activation of atrial cells triggers oxidative stress with lipid peroxidation and formation of γ-KAs, which then rapidly crosslink proteins to generate PAOs. To investigate this hypothesis, rapidly-paced and control, spontaneously-beating atrial HL-1 cells were probed with a conformation-specific antibody recognizing PAOs. Rapid stimulation of atrial cells caused the generation of cytosolic PAOs along with a myocyte stress response (e.g., transcriptional upregulation of Nppa and Hspa1a), both of which were absent in control, unpaced cells. Rapid activation also caused the formation of superoxide and γ-KA adducts in atriomyocytes, while direct exposure of cells to γ-KAs resulted in PAO production. Increased cytosolic atrial natriuretic peptide (ANP), and the generation of ANP oligomers with exposure to γ-KAs and rapid atrial HL-1 cell stimulation, strongly suggest a role for ANP in PAO formation. Salicylamine (SA) is a small molecule scavenger of γ-KAs that can protect proteins from modification by these reactive compounds. PAO formation and transcriptional remodeling were inhibited when cells were stimulated in the presence of SA, but not with the antioxidant curcumin, which is incapable of scavenging γ-KAs. These results demonstrate that γ-KAs promote protein misfolding and PAO formation as a component of the atrial cell stress response to rapid activation, and they provide a potential mechanistic link between oxidative stress and atrial cell injury.


Journal of Histochemistry and Cytochemistry | 2014

Quantitative Imaging of Preamyloid Oligomers, a Novel Structural Abnormality, in Human Atrial Samples

Tatiana N. Sidorova; Lisa C. Mace; K. Sam Wells; Liudmila V. Yermalitskaya; Pei Fang Su; Yu Shyr; John G. Byrne; Michael R. Petracek; James P. Greelish; Steven J. Hoff; Stephen K. Ball; Charles G. Glabe; Nancy J. Brown; Joey V. Barnett; Katherine T. Murray

Abnormalities in atrial myocardium increase the likelihood of arrhythmias, including atrial fibrillation (AF). The deposition of misfolded protein, or amyloidosis, plays an important role in the pathophysiology of many diseases, including human cardiomyopathies. We have shown that genes implicated in amyloidosis are activated in a cellular model of AF, with the development of preamyloid oligomers (PAOs). PAOs are intermediates in the formation of amyloid fibrils, and they are now recognized to be the cytotoxic species during amyloidosis. To investigate the presence of PAOs in human atrium, we developed a microscopic imaging-based protocol to enable robust and reproducible quantitative analysis of PAO burden in atrial samples harvested at the time of elective cardiac surgery. Using PAO- and myocardial-specific antibodies, we found that PAO distribution was typically heterogeneous within a myocardial sample. Rigorous imaging and analysis protocols were developed to quantify the relative area of myocardium containing PAOs, termed the Green/Red ratio (G/R), for a given sample. Using these methods, reproducible G/R values were obtained when different sections of a sample were independently processed, imaged, and analyzed by different investigators. This robust technique will enable studies to investigate the role of this novel structural abnormality in the pathophysiology of and arrhythmia generation in human atrial tissue.


Journal of the American Heart Association | 2014

Hypertension Is Associated With Preamyloid Oligomers in Human Atrium: A Missing Link in Atrial Pathophysiology?

Tatiana N. Sidorova; Lisa C. Mace; K. Sam Wells; Liudmila V. Yermalitskaya; Pei Fang Su; Yu Shyr; James B. Atkinson; Agnes B. Fogo; Joseph K. Prinsen; John G. Byrne; Michael R. Petracek; James P. Greelish; Steven J. Hoff; Stephen K. Ball; Charles G. Glabe; Nancy J. Brown; Joey V. Barnett; Katherine T. Murray

Background Increasing evidence indicates that proteotoxicity plays a pathophysiologic role in experimental and human cardiomyopathy. In organ‐specific amyloidoses, soluble protein oligomers are the primary cytotoxic species in the process of protein aggregation. While isolated atrial amyloidosis can develop with aging, the presence of preamyloid oligomers (PAOs) in atrial tissue has not been previously investigated. Methods and Results Atrial samples were collected during elective cardiac surgery in patients without a history of atrial arrhythmias, congestive heart failure, cardiomyopathy, or amyloidosis. Immunohistochemistry was performed for PAOs using a conformation‐specific antibody, as well as for candidate proteins identified previously in isolated atrial amyloidosis. Using a myocardium‐specific marker, the fraction of myocardium colocalizing with PAOs (PAO burden) was quantified (green/red ratio). Atrial samples were obtained from 92 patients, with a mean age of 61.7±13.8 years. Most patients (62%) were male, 23% had diabetes, 72% had hypertension, and 42% had coronary artery disease. A majority (n=62) underwent aortic valve replacement, with fewer undergoing coronary artery bypass grafting (n=34) or mitral valve replacement/repair (n=24). Immunostaining detected intracellular PAOs in a majority of atrial samples, with a heterogeneous distribution throughout the myocardium. Mean green/red ratio value for the samples was 0.11±0.1 (range 0.03 to 0.77), with a value ≥0.05 in 74 patients. Atrial natriuretic peptide colocalized with PAOs in myocardium, whereas transthyretin was located in the interstitium. Adjusting for multiple covariates, PAO burden was independently associated with the presence of hypertension. Conclusion PAOs are frequently detected in human atrium, where their presence is associated with clinical hypertension.


Circulation-arrhythmia and Electrophysiology | 2017

Azithromycin Causes a Novel Proarrhythmic Syndrome

Zhenjiang Yang; Joseph K. Prinsen; Kevin Bersell; Wangzhen Shen; Liudmila V. Yermalitskaya; Tatiana N. Sidorova; Paula B. Luis; Lynn Hall; Wei Zhang; Liping Du; Ginger L. Milne; Patrick Tucker; Alfred L. George; Courtney M. Campbell; Robert A. Pickett; Christian M. Shaffer; Nagesh Chopra; Tao Yang; Björn C. Knollmann; Dan M. Roden; Katherine T. Murray

Background— The widely used macrolide antibiotic azithromycin increases risk of cardiovascular and sudden cardiac death, although the underlying mechanisms are unclear. Case reports, including the one we document here, demonstrate that azithromycin can cause rapid, polymorphic ventricular tachycardia in the absence of QT prolongation, indicating a novel proarrhythmic syndrome. We investigated the electrophysiological effects of azithromycin in vivo and in vitro using mice, cardiomyocytes, and human ion channels heterologously expressed in human embryonic kidney (HEK 293) and Chinese hamster ovary (CHO) cells. Methods and Results— In conscious telemetered mice, acute intraperitoneal and oral administration of azithromycin caused effects consistent with multi-ion channel block, with significant sinus slowing and increased PR, QRS, QT, and QTc intervals, as seen with azithromycin overdose. Similarly, in HL-1 cardiomyocytes, the drug slowed sinus automaticity, reduced phase 0 upstroke slope, and prolonged action potential duration. Acute exposure to azithromycin reduced peak SCN5A currents in HEK cells (IC50=110±3 &mgr;mol/L) and Na+ current in mouse ventricular myocytes. However, with chronic (24 hour) exposure, azithromycin caused a ≈2-fold increase in both peak and late SCN5A currents, with findings confirmed for INa in cardiomyocytes. Mild block occurred for K+ currents representing IKr (CHO cells expressing hERG; IC50=219±21 &mgr;mol/L) and IKs (CHO cells expressing KCNQ1+KCNE1; IC50=184±12 &mgr;mol/L), whereas azithromycin suppressed L-type Ca++ currents (rabbit ventricular myocytes, IC50=66.5±4 &mgr;mol/L) and IK1 (HEK cells expressing Kir2.1, IC50=44±3 &mgr;mol/L). Conclusions— Chronic exposure to azithromycin increases cardiac Na+ current to promote intracellular Na+ loading, providing a potential mechanistic basis for the novel form of proarrhythmia seen with this macrolide antibiotic.


Acta Biomaterialia | 2017

I-Wire Heart-on-a-Chip I: Three-dimensional cardiac tissue constructs for physiology and pharmacology

Veniamin Y. Sidorov; Philip C. Samson; Tatiana N. Sidorova; Jeffrey M. Davidson; Chee Chew Lim; John P. Wikswo


American Journal of Pathology | 2016

Cardiomyocyte-Specific Human Bcl2-Associated Anthanogene 3 P209L Expression Induces Mitochondrial Fragmentation, Bcl2-Associated Anthanogene 3 Haploinsufficiency, and Activates p38 Signaling.

Megan T. Quintana; Traci L. Parry; Jun He; Cecelia C. Yates; Tatiana N. Sidorova; Katherine T. Murray; James R. Bain; Christopher B. Newgard; Michael J. Muehlbauer; Samuel C. Eaton; Akinori Hishiya; Shin Takayama; Monte S. Willis


Cardiovascular Pathology | 2016

BRG1 and BRM SWI/SNF ATPases redundantly maintain cardiomyocyte homeostasis by regulating cardiomyocyte mitophagy and mitochondrial dynamics in vivo

Scott J. Bultman; Darcy Holley; Gustaaf G. de Ridder; Salvatore V. Pizzo; Tatiana N. Sidorova; Katherine T. Murray; Brian C. Jensen; Zhongjing Wang; Ariana Bevilacqua; Xin Chen; Megan T. Quintana; Manasi Tannu; Gary B. Rosson; Kumar Pandya; Monte S. Willis


The FASEB Journal | 2016

Pharmacological Inhibition of p38/MAPK Improves Cardiac Function in Cardiac-specific Bag3-P209L Transgenic Mice

Samuel C. Eaton; Shinuchi Takayama; Tatiana N. Sidorova; Katherine T. Murray; Monte S. Willis


Circulation | 2013

Abstract 13925: Reactive ?-Ketoaldehyde Products of Lipid Peroxidation Promote Protein Misfolding and Preamyloid Oligomer Formation in Experimental Atrial Fibrillation

Tatiana N. Sidorova; Lisa C. Mace; Liudmila V. Yermalitskaya; Sean S. Davies; Jackson Roberts; Charles G. Glabe; Joey V. Barnett; Katherine T. Murray


Investigative Ophthalmology & Visual Science | 2012

The TRPV1 Response to Stress of Retinal Ganglion Cells

Nicholas J. Ward; Karen W. Ho; Tatiana N. Sidorova; David J. Calkins

Collaboration


Dive into the Tatiana N. Sidorova's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David J. Calkins

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Monte S. Willis

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge