Network


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

Hotspot


Dive into the research topics where Igor Mikaelian is active.

Publication


Featured researches published by Igor Mikaelian.


Genome Biology | 2007

Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors

Jason I. Herschkowitz; Karl Simin; Victor J. Weigman; Igor Mikaelian; Jerry Usary; Zhiyuan Hu; Karen Rasmussen; Laundette P Jones; Shahin Assefnia; Subhashini Chandrasekharan; Michael G. Backlund; Yuzhi Yin; Andrey Khramtsov; Roy Bastein; John Quackenbush; Robert I. Glazer; Powel H. Brown; Jeffrey Green; Levy Kopelovich; Priscilla A. Furth; Juan P. Palazzo; Olufunmilayo I. Olopade; Philip S. Bernard; Gary A. Churchill; Terry Van Dyke; Charles M. Perou

BackgroundAlthough numerous mouse models of breast carcinomas have been developed, we do not know the extent to which any faithfully represent clinically significant human phenotypes. To address this need, we characterized mammary tumor gene expression profiles from 13 different murine models using DNA microarrays and compared the resulting data to those from human breast tumors.ResultsUnsupervised hierarchical clustering analysis showed that six models (TgWAP-Myc, TgMMTV-Neu, TgMMTV-PyMT, TgWAP-Int3, TgWAP-Tag, and TgC3(1)-Tag) yielded tumors with distinctive and homogeneous expression patterns within each strain. However, in each of four other models (TgWAP-T121, TgMMTV-Wnt1, Brca1Co/Co;TgMMTV-Cre;p53+/- and DMBA-induced), tumors with a variety of histologies and expression profiles developed. In many models, similarities to human breast tumors were recognized, including proliferation and human breast tumor subtype signatures. Significantly, tumors of several models displayed characteristics of human basal-like breast tumors, including two models with induced Brca1 deficiencies. Tumors of other murine models shared features and trended towards significance of gene enrichment with human luminal tumors; however, these murine tumors lacked expression of estrogen receptor (ER) and ER-regulated genes. TgMMTV-Neu tumors did not have a significant gene overlap with the human HER2+/ER- subtype and were more similar to human luminal tumors.ConclusionMany of the defining characteristics of human subtypes were conserved among the mouse models. Although no single mouse model recapitulated all the expression features of a given human subtype, these shared expression features provide a common framework for an improved integration of murine mammary tumor models with human breast tumors.


Toxicologic Pathology | 2010

Time Course Characterization of Serum Cardiac Troponins, Heart Fatty Acid-binding Protein, and Morphologic Findings With Isoproterenol-induced Myocardial Injury in the Rat

Peter J. Clements; Sally Brady; Malcolm York; Brian R. Berridge; Igor Mikaelian; Rosemary Nicklaus; Mitul Gandhi; Ian Roman; Clare Stamp; Dai Davies; Paul McGill; Tom C. Williams; Syril Pettit; Dana Walker; John Turton

We investigated the kinetics of circulating biomarker elevation, specifically correlated with morphology in acute myocardial injury. Male Hanover Wistar rats underwent biomarker and morphologic cardiac evaluation at 0.5 to seventy-two hours after a single subcutaneous isoproterenol administration (100 or 4000 µg/kg). Dose-dependent elevations of serum cardiac troponins I and T (cTnI, cTnT), and heart fatty acid–binding protein (H-FABP) occurred from 0.5 hour, peaked at two to three hours, and declined to baseline by twelve hours (H-FABP) or forty-eight to seventy-two hours (Serum cTns). They were more sensitive in detecting cardiomyocyte damage than other serum biomarkers. The Access 2 platform, an automated chemiluminescence analyzer (Beckman Coulter), showed the greatest cTnI fold-changes and low range sensitivity. Myocardial injury was detected morphologically from 0.5 hour, correlating well with loss of cTnI immunoreactivity and serum biomarker elevation at early time points. Ultrastructurally, there was no evidence of cardiomyocyte death at 0.5 hour. After three hours, a clear temporal disconnect occurred: lesion scores increased with declining cTnI, cTnT, and H-FABP values. Serum cTns are sensitive and specific markers for detecting acute/active cardiomyocyte injury in this rat model. Heart fatty acid–binding protein is a good early marker but is less sensitive and nonspecific. Release of these biomarkers begins early in myocardial injury, prior to necrosis. Assessment of cTn merits increased consideration for routine screening of acute/ongoing cardiomyocyte injury in rat toxicity studies.


Toxicologic Pathology | 2008

Temporal Gene Expression Profiling Indicates Early Up-regulation of Interleukin-6 in Isoproterenol-induced Myocardial Necrosis in Rat

Igor Mikaelian; Denise Coluccio; Kevin Morgan; Teona Johnson; Amber L. Ryan; Erik Roy Rasmussen; Rosemary Nicklaus; Charu Kanwal; Holly Hilton; Karl Frank; Luke Fritzky; Eric Wheeldon

Gene expression was evaluated in the myocardium of male Wistar rats after a single subcutaneous administration of 0.5 mg of isoproterenol, a β-adrenergic agonist that causes acute tachycardia with subsequent myocardial necrosis. Histology of the heart, clinical chemistry, and hematology were evaluated at 9 time points (0.5 hours to 14 days postinjection). Myocardial gene expression was evaluated at 4 time points (1 hour to 3 days). Contraction bands and loss of cross-striation were identified on phosphotungstic acid-hematoxylin-stained sections 0.5 hours postdosing. Plasma troponin I elevation was detected at 0.5 hours, peaked at 3 hours, and returned to baseline values at 3 days postdosing. Interleukin 6 (Il6) expression spiked at 1 to 3 hours and was followed by a short-lived, time-dependent dysregulation of its downstream targets. Concurrently and consistent with the kinetics of the histologic findings, many pathways indicative of necrosis/apoptosis (p38 mitogen-activated protein kinase [MAPK] signaling, NF-κB signaling) and adaptation to hypertension (PPAR signaling) were overrepresented at 3 hours. The 1-day and 3-day time points indicated an adaptive response, with down-regulation of the fatty acid metabolism pathway, up-regulation of the fetal gene program, and superimposed inflammation and repair at 3 days. These results suggest early involvement of Il6 in isoproterenol-induced myocardial necrosis and emphasize the value of early time points in transcriptomic studies.


Toxicologic Pathology | 2009

Assessment of the Toxicity of Hydralazine in the Rat Using an Ultrasensitive Flow-based Cardiac Troponin I Immunoassay

Igor Mikaelian; Denise Coluccio; Gerard Hirkaler; John C. Downing; Erik Roy Rasmussen; John A. Todd; Joel Estis; Quynh Anh Lu; Rosemary Nicklaus

The purpose of this study was to correlate the histologic changes in the heart to serum cardiac troponin I (cTnI) concentrations assayed with the Erenna Immunoassay System in Wistar rats (Crl:Wi[Han]) using the hydralazine model of cardiotoxicity. A single dose of hydralazine caused an increase of cTnI concentrations at six hours post-dose, followed by a sharp decrease at twenty-four hours and a return to baseline at forty-eight hours. The second dose of hydralazine caused a smaller magnitude increase in cTnI concentrations at six hours as compared to the first dose. Also, cTnI concentrations returned to baseline at twenty-four hours after the second dose. The increased cTnI concentrations coincided with acute myocardial necrosis at histology. However, increased cTnI concentrations in the absence of microscopic lesions were identified in several rats. As cTnI concentrations decreased, microscopic changes in the heart matured to cardiomyophagy. In conclusion, the increases in cTnI concentrations six hours after the administration of hydralazine were indicative of a myocardial damage that did not consistently have a microscopic correlate. However, the window of increased cTnI concentrations was short, and only microscopic evaluation of the heart detected the damage at twenty-four to forty-eight hours after the episode of acute myocardial necrosis.


Experimental Lung Research | 2005

The Mouse Tumor Biology Database: integrated access to mouse cancer biology data.

Debra M. Krupke; Dieter Näf; Matthew J. Vincent; Theresa Allio; Igor Mikaelian; John P. Sundberg; Janan T. Eppig

Mice have long been used as models for the study of human cancer. The National Cancer Institute has included among its research areas of extraordinary opportunity the development of new mouse genetic models of human cancer and the exploration of cancer imaging as a research tool. Because of the volume and interconnectedness of relevant data, the creation and maintenance of bioinformatics resources for mouse tumor biology is necessary to facilitate current and future cancer research. The Mouse Tumor Biology (MTB) Database provides electronic access to data generated through the study of spontaneous and induced tumors in genetically defined mice (inbred, hybrid, spontaneous and induced mutant, and genetically engineered strains of mice).


Cancer Research | 2017

Abstract 653: Inhibition of MMP9 improves anti-tumor immunity by changing the tumor microenvironment to promote T cell trafficking and activation

Vladi Juric; Amanda Mikels-Vigdal; Chris O'Sullivan; Andrew Greenstein; Erin Stefanutti; Vivian Barry-Hamilton; Igor Mikaelian; Ted Sullivan; Erik G. Huntzicker; Jeremiah Degenhardt; Peng Yue; Victoria C. Smith

Background: Matrix metalloproteinase 9 (MMP9) acts via diverse mechanisms to promote tumor growth, invasion, and metastasis. It activates growth factors and signaling pathways, promotes angiogenesis, and impedes anti-tumor immune responses. We developed a monoclonal antibody that inhibits mouse MMP9 (AB0046) and assessed its mechanism of action in immunocompetent mouse tumor models and in vitro assays. Methods: We examined MMP9 expression in a variety of human tumor tissues via immunohistochemistry. Human monocytes were differentiated in vitro and protein expression was assessed via enzyme-linked immunosorbent assay. Primary tumor growth in orthotopic, syngeneic tumor models was examined following anti-MMP9 antibody treatment. RNA sequencing, immunohistochemical and flow cytometry analyses were performed on tumor tissues to assess gene expression, stromal remodeling and macrophage polarization in response to AB0046 treatment. Results: MMP9 levels are elevated in human tumors compared to healthy tissues. The protein is expressed predominantly in stromal cells, including macrophages and neutrophils, with more occasional heterogeneous expression in tumor epithelia. Differentiation of human monocyte-derived macrophages in vitro revealed that M2 polarization is associated with increased expression of MMP9 and Th2 markers CCL18 and TGFβ. Anti-MMP9 treatment in three independent mouse tumor models (HC11-NeuT, CT26, Lewis lung carcinoma (LLC)) resulted in decreased primary tumor growth (p=0.001 and p=0.018 for HC11-NeuT and CT26 respectively) and increased animal survival (p=0.024 for LLC). Gene expression profiling of tumors from the various models demonstrated that inhibition of MMP9 resulted in elevated expression of genes associated with immune cell activation pathways (Hallmark Interferon Gamma Response, p Conclusions: These analyses show that MMP9 is expressed in a variety of human tumors. Our data suggest that inhibition of MMP9 promotes anti-tumor immunity and enhances a Th1 immune response. GS-5745, a humanized anti-MMP9 inhibitory antibody, is being evaluated in gastric cancer in phase 3 and 2 studies with chemotherapy and nivolumab, respectively (NCT02545504, NCT02864381). Citation Format: Vladi Juric, Amanda Mikels-Vigdal, Chris O9Sullivan, Andrew Greenstein, Erin Stefanutti, Vivian Barry-Hamilton, Igor Mikaelian, Ted Sullivan, Erik Huntzicker, Jeremiah Degenhardt, Peng Yue, Victoria Smith. Inhibition of MMP9 improves anti-tumor immunity by changing the tumor microenvironment to promote T cell trafficking and activation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 653. doi:10.1158/1538-7445.AM2017-653


Toxicologic Pathology | 2016

Syk Inhibition Induces Platelet Dependent Peri-islet Hemorrhage in the Rat Pancreas

Andrew J. Long; Erik Sampson; Richard McCarthy; Christopher M. Harris; Marc Barnard; Dan Shi; Donna Conlon; Robert Caldwell; David Honor; Neil Wishart; Michael Hoemann; Lori Duggan; Douglas Fritz; Christopher Stedman; Elizabeth O’Connor; Igor Mikaelian; Annette Schwartz

Spleen tyrosine kinase (Syk) is a nonreceptor tyrosine kinase that is an important signaling enzyme downstream of immunoreceptors containing an intracellular immunoreceptor tyrosine activating motif (ITAM). These receptors encompass a wide variety of biological functions involved in autoimmune disease pathogenesis. There has been considerable interest in the development of inhibitors of the Syk pathway for the treatment of rheumatoid arthritis and systemic lupus erythematosus. We report that Syk inhibition mechanistically caused peri-islet hemorrhages and fibrin deposition in the rat pancreas and that this finding is due to a homeostatic functional defect in platelets. In more limited studies, similar lesions could not be induced in mice, dogs, and cynomolgus monkeys at similar or higher plasma drug concentrations. Irradiation-induced thrombocytopenia caused a phenotypically similar peri-islet pancreas lesion and the formation of this lesion could be prevented by platelet transfusion. In addition, Syk inhibitor-induced lesions were prevented by the coadministration of prednisone. A relatively greater sensitivity of rat platelets to Syk inhibition was supported by functional analyses demonstrating rat-specific differences in response to convulxin, a glycoprotein VI agonist that signals through Syk. These data demonstrate that the Syk pathway is critical in platelet–endothelial cell homeostasis in the peri-islet pancreatic microvasculature in rats.


Toxicologic Pathology | 2004

Antibodies that label paraffin-embedded mouse tissues: a collaborative endeavor.

Igor Mikaelian; Lillian B. Nanney; Kelly S. Parman; Donna F. Kusewitt; Jerrold M. Ward; Dieter Näf; Debra M. Krupke; Janan T. Eppig; Rose Seymour; Tsutomu Ichiki; John P. Sundberg


Breast Cancer Research | 2004

Proteotypic classification of spontaneous and transgenic mammary neoplasms

Igor Mikaelian; Natalie J. Blades; Gary A. Churchill; Karen S. Fancher; Barbara B. Knowles; Janan T. Eppig; John P. Sundberg


The Laboratory Mouse | 2004

CHAPTER 30 – Necropsy Methods

Rosemarie Seymour; Tsutomu Ichiki; Igor Mikaelian; Dawnalyn Boggess; Kathleen A. Silva; John P. Sundberg

Collaboration


Dive into the Igor Mikaelian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles M. Perou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Janan T. Eppig

European Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juan P. Palazzo

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Karen Rasmussen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Karl Simin

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Levy Kopelovich

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge