Network


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

Hotspot


Dive into the research topics where Margarita M. Ivanova is active.

Publication


Featured researches published by Margarita M. Ivanova.


Environmental Toxicology | 2014

Effect of nonpersistent pesticides on estrogen receptor, androgen receptor, and aryl hydrocarbon receptor.

Svjetlana Medjakovic; Alfred Zoechling; Petra Gerster; Margarita M. Ivanova; Yun Teng; Carolyn M. Klinge; Barbara Schildberger; Michael Gartner; Alois Jungbauer

Nonpersistent pesticides are considered less harmful for the environment, but their impact as endocrine disruptors has not been fully explored. The pesticide Switch was applied to grape vines, and the maximum residue concentration of its active ingredients was quantified. The transactivation potential of the pesticides Acorit, Frupica, Steward, Reldan, Switch, Cantus, Teldor, and Scala and their active compounds (hexythiazox, mepanipyrim, indoxacarb, chlorpyrifos‐methyl, cyprodinil, fludioxonil, boscalid, fenhexamid, and pyrimethanil) were tested on human estrogen receptor α (ERα), androgen receptor (AR) and arylhydrocarbon receptor (AhR) in vitro. Relative binding affinities of the pure pesticide constituents for AR and their effect on human breast cancer and prostate cancer cell lines were evaluated. Residue concentrations of Switchs ingredients were below maximum residue limits. Fludioxonil and fenhexamid were ERα agonists (EC50‐values of 3.7 and 9.0 μM, respectively) and had time‐dependent effects on endogenous ERα‐target gene expression (cyclin D1, progesterone receptor, and nuclear respiratory factor 1) in MCF‐7 human breast cancer cells. Fludioxonil, mepanipyrim, cyprodinil, pyrimethanil, and chlorpyrifos‐methyl were AhR‐agonists (EC50s of 0.42, 0.77, 1.4, 4.6, and 5.1 μM, respectively). Weak AR binding was shown for chlorpyrifos‐methyl, cyprodinil, fenhexamid, and fludioxonil. Assuming a total uptake which does not take metabolism and clearance rates into account, our in vitro evidence suggests that pesticides could activate pathways affecting hormonal balance, even within permitted limits, thus potentially acting as endocrine disruptors.


The FASEB Journal | 2008

Resveratrol stimulates nitric oxide production by increasing estrogen receptor α-Src-caveolin-1 interaction and phosphorylation in human umbilical vein endothelial cells

Carolyn M. Klinge; Nalinie S. Wickramasinghe; Margarita M. Ivanova; Susan M. Dougherty

Epidemiological studies correlate moderate red wine consumption to reduced incidence of cardiovascular disease. Resveratrol is a polyphenolic compound in red wine that has cardioprotective effects in rodents. Although endothelial cell (EC) studies indicate that micromolar resveratrol has diverse biological activities, these concentrations are not physiologically relevant because human oral ingestion provides only brief exposure to nanomolar plasma levels. Previously, we reported that nanomolar resveratrol activated ERK1/2 signaling in bovine aortic ECs (BAECs). The goal of this study was to determine the mechanisms by which nanomolar resveratrol rapidly activates endothelial nitric oxide synthase (eNOS) in human umbilical vein ECs (HUVECs). We report for the first time that resveratrol increased interaction between estrogen receptor α ( ERα ), caveolin‐1 (Cav‐1) and c‐Src, and increased phosphorylation of Cav‐1, c‐Src, and eNOS. Pretreatment with the lipid raft disruptor beta‐methyl cyclodextrin or Gα inhibitor pertussis toxin blocked resveratrol‐ and E2‐induced eNOS activation and NO production. Depletion of endogenous ERα, not ERβ, by siRNA attenuated resveratrol‐ and E2‐induced ERK1/2, Src, and eNOS phosphorylation. Our data demonstrate that nanomolar resveratrol induces ERα‐Cav‐1‐c‐SRC interaction, resulting in NO production through a Gα‐protein‐coupled mechanism. This study provides important new insights into mechanisms for the beneficial effects of resveratrol in ECs.—Klinge, C. M., Wickramasinghe, N. S., Ivanova, M. M., Dougherty, S. M. Resveratrol stimulates nitric oxide produc tion by increasing estrogen receptor α‐Src‐caveolin‐1 interaction and phosphorylation in human umbilical vein endothelial cells. FASEB J. 22, 2185–2197 (2008)


Molecular and Cellular Endocrinology | 2009

Activity and intracellular location of estrogen receptors α and β in human bronchial epithelial cells

Margarita M. Ivanova; Williard Mazhawidza; Susan M. Dougherty; John D. Minna; Carolyn M. Klinge

Gender differences in lung disease and cancer are well-established. We reported estrogenic transcriptional responses in lung adenocarcinoma cells from females but not males despite similar estrogen receptor (ER) expression. Here we tested the hypothesis that normal human bronchial epithelial cells (HBECs) show gender-independent estrogenic responses. We report that a small sample of HBECs express approximately twice as much ERbeta as ERalpha. ERalpha and ERbeta were located in the cytoplasm, nucleus, and mitochondria. In contrast to lung adenocarcinoma cells, estradiol (E2) induced estrogen response element (ERE)-mediated luciferase reporter activity in transiently transfected HBECs regardless of donor gender. Overexpression of ERalpha-VP16 increased ERE-mediated transcriptional activity in all HBECs. E2 increased and 4-hydroxytamoxifen and ICI 182,780 inhibited HBEC proliferation and cyclin D1 expression in a cell line-specific manner. In conclusion, the response of HBECs to ER ligands is gender-independent suggesting that estrogenic sensitivity may be acquired during lung carcinogenesis.


American Journal of Respiratory Cell and Molecular Biology | 2010

Sex Differences in Estrogen Receptor Subcellular Location and Activity in Lung Adenocarcinoma Cells

Margarita M. Ivanova; Williard Mazhawidza; Susan M. Dougherty; Carolyn M. Klinge

The role of estrogens in the increased risk of lung adenocarcinoma in women remains uncertain. We reported that lung adenocarcinoma cell lines from female, but not male, patients with non-small cell lung cancer respond proliferatively and transcriptionally to estradiol (E(2)), despite equal protein expression of estrogen receptors (ER) alpha and beta. To test the hypothesis that nuclear localization of ER alpha corresponds to genomic E(2) activity in lung adenocarcinoma cells from females, cell fractionation, immunoblot, and confocal immunohistochemical microscopy were performed. We report for the first time that E(2) increases phospho-serine-118-ER alpha (P-ser118-ER alpha) and cyclin D1 (CCND1) nuclear colocalization in H1793, but not A549 lung adenocarcinoma cells, derived from a female and male patient, respectively. ER beta was primarily in the cytoplasm and mitochondria, independent of E(2) treatment, and showed no difference between H1793 and A549 cells. E(2) induced higher transcription of endogenous ER alpha-regulated CCND1 in H1793 than in A549 cells. Likewise, higher rapid, non-genomic E(2)-induced extracellular signal-regulated kinase 1/2 activation was detected in H1793 compared with A549 cells, linking extracellular signal-regulated kinase activation to increased P-ser118-ER alpha. Furthermore, E(2) increased cyclin D1 and P-ser118-ER alpha nuclear localization in H1793, but not A549 cells. Together, our results indicate that nuclear localization of P-ser118-ER alpha provides one explanation for sex-dependent differences in E(2)-genomic responses in lung adenocarcinoma cell lines.


Steroids | 2013

DHEA metabolites activate estrogen receptors alpha and beta.

Kristy K. Michael Miller; Numan Al-Rayyan; Margarita M. Ivanova; Kathleen A. Mattingly; Sharon L. Ripp; Carolyn M. Klinge; Russell A. Prough

Dehydroepiandrosterone (DHEA) levels were reported to associate with increased breast cancer risk in postmenopausal women, but some carcinogen-induced rat mammary tumor studies question this claim. The purpose of this study was to determine how DHEA and its metabolites affect estrogen receptors α or β (ERα or ERβ)-regulated gene transcription and cell proliferation. In transiently transfected HEK-293 cells, androstenediol, DHEA, and DHEA-S activated ERα. In ERβ transfected HepG2 cells, androstenedione, DHEA, androstenediol, and 7-oxo DHEA stimulated reporter activity. ER antagonists ICI 182,780 (fulvestrant) and 4-hydroxytamoxifen, general P450 inhibitor miconazole, and aromatase inhibitor exemestane inhibited activation by DHEA or metabolites in transfected cells. ERβ-selective antagonist R,R-THC (R,R-cis-diethyl tetrahydrochrysene) inhibited DHEA and DHEA metabolite transcriptional activity in ERβ-transfected cells. Expression of endogenous estrogen-regulated genes: pS2, progesterone receptor, cathepsin D1, and nuclear respiratory factor-1 was increased by DHEA and its metabolites in an ER-subtype, gene, and cell-specific manner. DHEA metabolites, but not DHEA, competed with 17β-estradiol for ERα and ERβ binding and stimulated MCF-7 cell proliferation, demonstrating that DHEA metabolites interact directly with ERα and ERβin vitro, modulating estrogen target genes in vivo.


Molecular and Cellular Biology | 2005

Scaffold Attachment Factor B1 Functions in Development, Growth, and Reproduction

Margarita M. Ivanova; Klaudia M. Dobrzycka; Shiming Jiang; Kai Michaelis; Rene Meyer; Kaiyan Kang; Brian Adkins; Oleg A. Barski; Simeen Zubairy; Jana Divisova; Adrian V. Lee; Steffi Oesterreich

ABSTRACT Scaffold attachment factor B1 (SAFB1) is a multifunctional protein that can bind both DNA and RNA and is involved in RNA processing and stress response. In addition, SAFB1 contains a transcriptional repression domain and can bind certain hormone receptors and repress their activity. To assess the role of SAFB1 in vivo, we generated SAFB1 mutant mice through targeted deletion in embryonic stem cells. While viable homozygous mutant (SAFB1−/−) mice were obtained, genotypic distribution indicated that homozygous deficiency resulted in both prenatal and neonatal lethality. Mice lacking SAFB1 exhibited dwarfism, as a result of in utero growth retardation, and had low serum insulin-like growth factor 1 (IGF1) levels. In agreement with the previous characterization of SAFB1 as a corepressor for hormone receptors, we found that SAFB1−/− mice displayed dramatic defects in the development and function of the reproductive system. Male SAFB1 null mice were infertile, apparently because of low circulating levels of testosterone. SAFB1−/− testes were small and showed progressive degeneration of the germinal epithelium, increased apoptosis of germ cells, and Leydig cell hyperplasia. SAFB−/− female mice were subfertile and showed progressive infertility, in part because of defects in oviductal transport and reduced numbers of follicles. Immortalized SAFB1−/− mouse embryonic fibroblasts showed cell-intrinsic defects including increased transcriptional estrogen receptor α activity and enhanced responsiveness to IGF1. Together, these in vivo findings establish a critical role for SAFB1 in development, growth regulation, and reproduction.


Molecular Cancer Therapeutics | 2010

Anacardic acid inhibits estrogen receptor alpha-DNA binding and reduces target gene transcription and breast cancer cell proliferation

David J. Schultz; Nalinie S. Wickramasinghe; Margarita M. Ivanova; Susan M. Isaacs; Susan M. Dougherty; Yoannis Imbert-Fernandez; Albert R. Cunningham; Chunyuan Chen; Carolyn M. Klinge

Anacardic acid (AnAc; 2-hydroxy-6-alkylbenzoic acid) is a dietary and medicinal phytochemical with established anticancer activity in cell and animal models. The mechanisms by which AnAc inhibits cancer cell proliferation remain undefined. AnAc 24:1ω5 was purified from geranium (Pelargonium × hortorum) and shown to inhibit the proliferation of estrogen receptor α (ERα)–positive MCF-7 and endocrine-resistant LCC9 and LY2 breast cancer cells with greater efficacy than ERα-negative primary human breast epithelial cells, MCF-10A normal breast epithelial cells, and MDA-MB-231 basal-like breast cancer cells. AnAc 24:1ω5 inhibited cell cycle progression and induced apoptosis in a cell-specific manner. AnAc 24:1ω5 inhibited estradiol (E2)–induced estrogen response element (ERE) reporter activity and transcription of the endogenous E2 target genes pS2, cyclin D1, and cathepsin D in MCF-7 cells. AnAc 24:1ω5 did not compete with E2 for ERα or ERβ binding, nor did AnAc 24:1ω5 reduce ERα or ERβ steady-state protein levels in MCF-7 cells; rather, AnAc 24:1ω5 inhibited ER-ERE binding in vitro. Virtual screening with the molecular docking software Surflex evaluated AnAc 24:1ω5 interaction with ERα ligand binding (LBD) and DNA binding (DBD) domains in conjunction with experimental validation. Molecular modeling revealed AnAc 24:1ω5 interaction with the ERα DBD but not the LBD. Chromatin immunoprecipitation experiments revealed that AnAc 24:1ω5 inhibited E2-ERα interaction with the endogenous pS2 gene promoter region containing an ERE. These data indicate that AnAc 24:1ω5 inhibits cell proliferation, cell cycle progression, and apoptosis in an ER-dependent manner by reducing ER-DNA interaction and inhibiting ER-mediated transcriptional responses. Mol Cancer Ther; 9(3); 594–605


PLOS ONE | 2012

Identification and characterization of nucleolin as a COUP-TFII coactivator of retinoic acid receptor β transcription in breast cancer cells.

Lacey M. Litchfield; Krista A. Riggs; Alyson M. Hockenberry; Laura Don Oliver; Katelyn G. Barnhart; Jian Cai; William M. Pierce; Margarita M. Ivanova; Paula J. Bates; Savitri Appana; Susmita Datta; Piotr Kulesza; Jean McBryan; Leonie Young; Carolyn M. Klinge

Introduction The orphan nuclear receptor COUP-TFII plays an undefined role in breast cancer. Previously we reported lower COUP-TFII expression in tamoxifen/endocrine- resistant versus sensitive breast cancer cell lines. The identification of COUP-TFII-interacting proteins will help to elucidate its mechanism of action as a transcriptional regulator in breast cancer. Results FLAG-affinity purification and multidimensional protein identification technology (MudPIT) identified nucleolin among the proteins interacting with COUP-TFII in MCF-7 tamoxifen-sensitive breast cancer cells. Interaction of COUP-TFII and nucleolin was confirmed by coimmunoprecipitation of endogenous proteins in MCF-7 and T47D breast cancer cells. In vitro studies revealed that COUP-TFII interacts with the C-terminal arginine-glycine repeat (RGG) domain of nucleolin. Functional interaction between COUP-TFII and nucleolin was indicated by studies showing that siRNA knockdown of nucleolin and an oligonucleotide aptamer that targets nucleolin, AS1411, inhibited endogenous COUP-TFII-stimulated RARB2 expression in MCF-7 and T47D cells. Chromatin immunoprecipitation revealed COUP-TFII occupancy of the RARB2 promoter was increased by all-trans retinoic acid (atRA). RARβ2 regulated gene RRIG1 was increased by atRA and COUP-TFII transfection and inhibited by siCOUP-TFII. Immunohistochemical staining of breast tumor microarrays showed nuclear COUP-TFII and nucleolin staining was correlated in invasive ductal carcinomas. COUP-TFII staining correlated with ERα, SRC-1, AIB1, Pea3, MMP2, and phospho-Src and was reduced with increased tumor grade. Conclusions Our data indicate that nucleolin plays a coregulatory role in transcriptional regulation of the tumor suppressor RARB2 by COUP-TFII.


The FASEB Journal | 2011

Tamoxifen increases nuclear respiratory factor 1 transcription by activating estrogen receptor β and AP-1 recruitment to adjacent promoter binding sites

Margarita M. Ivanova; Kristen H. Luken; Amber S. Zimmer; Felicia L. Lenzo; Ryan J. Smith; Maia W. Arteel; Tara J. Kollenberg; Kathleen A. Mattingly; Carolyn M. Klinge

Little is known about endogenous estrogen receptor β (ERβ) gene targets in human breast cancer. We reported that estradiol (E2) induces nuclear respiratory factor‐1 (NRF‐1) transcription through ERα in MCF‐7 breast cancer cells. Here we report that 4‐hydroxytamoxifen (4‐OHT), with an EC50 of ˜1.7 nM, increases NRF‐1 expression by recruiting ERβ, cJun, cFos, CBP, and RNA polymerase II to and dismissing NCoR from the NRF1 promoter. Promoter deletion and transient transfection studies showed that the estrogen response element (ERE) is essential and that an adjacent AP‐1 site contributes to maximal 4‐OHT‐induced NRF‐1 transcription. siRNA knockdown of ERβ revealed that ERβ inhibits basal NRF‐1 expression and is required for 4‐OHT‐induced NRF‐1 transcription. An AP‐1 inhibitor blocked 4‐OHT‐in‐duced NRF‐1 expression. The 4‐OHT‐induced increase in NRF‐1 resulted in increased transcription of NRF‐1 target CAPNS1 but not CYC1, CYC2,or TFAM despite increased NRF‐1 coactivator PGC‐1α protein. The absence of TFAM induction corresponds to a lack of Akt‐dependent phosphorylation of NRF‐1 with 4‐OHT treatment. Overexpression of NRF‐1 inhibited 4‐OHT‐induced apoptosis and siRNA knockdown of NRF‐1 increased apoptosis, indicating an antiapoptotic role for NRF‐1. Overall, NRF‐1 expression and activity is regulated by 4‐OHT via endogenous ERβ in MCF‐7 cells.—Ivanova, M. M., Luken, K. H., Zimmer, A. S., Lenzo, F. L., Smith, R. J., Arteel, M. W., Kollenberg, T. J., Mattingly, K. A., Klinge, C. M. Tamoxifen increases nuclear respiratory factor 1 transcription by activating estrogen receptor β and AP‐1 recruitment to adjacent promoter binding sites. FASEB J. 25, 1402–1416 (2011). www.fasebj.org


Journal of Biological Chemistry | 2015

Dehydroepiandrosterone Activation of G-protein-coupled Estrogen Receptor Rapidly Stimulates MicroRNA-21 Transcription in Human Hepatocellular Carcinoma Cells

Yun Teng; Brandie N. Radde; Lacey M. Litchfield; Margarita M. Ivanova; Russell A. Prough; Barbara J. Clark; Mark A. Doll; David W. Hein; Carolyn M. Klinge

Background: MicroRNA-21 (miR-21) is an oncomiR in human hepatocellular carcinoma and is highly expressed in liver, but its regulation is uncharacterized. Results: Dehydroepiandrosterone (DHEA) rapidly increases miR-21 transcription in HepG2 cells by activating G-protein-coupled estrogen receptor (GPER). Conclusion: miR-21 transcription is regulated by DHEA through GPER. Significance: GPER may be among the activators of miR-21 expression in human hepatocellular carcinoma. Little is known about the regulation of the oncomiR miR-21 in liver. Dehydroepiandrosterone (DHEA) regulates gene expression as a ligand for a G-protein-coupled receptor and as a precursor for steroids that activate nuclear receptor signaling. We report that 10 nm DHEA increases primary miR-21 (pri-miR-21) transcription and mature miR-21 expression in HepG2 cells in a biphasic manner with an initial peak at 1 h followed by a second, sustained response from 3–12 h. DHEA also increased miR-21 in primary human hepatocytes and Hep3B cells. siRNA, antibody, and inhibitor studies suggest that the rapid DHEA-mediated increase in miR-21 involves a G-protein-coupled estrogen receptor (GPER/GPR30), estrogen receptor α-36 (ERα36), epidermal growth factor receptor-dependent, pertussis toxin-sensitive pathway requiring activation of c-Src, ERK1/2, and PI3K. GPER antagonist G-15 attenuated DHEA- and BSA-conjugated DHEA-stimulated pri-miR-21 transcription. Like DHEA, GPER agonists G-1 and fulvestrant increased pri-miR-21 in a GPER- and ERα36-dependent manner. DHEA, like G-1, increased GPER and ERα36 mRNA and protein levels. DHEA increased ERK1/2 and c-Src phosphorylation in a GPER-responsive manner. DHEA increased c-Jun, but not c-Fos, protein expression after 2 h. DHEA increased androgen receptor, c-Fos, and c-Jun recruitment to the miR-21 promoter. These results suggest that physiological concentrations of DHEA activate a GPER intracellular signaling cascade that increases pri-miR-21 transcription mediated at least in part by AP-1 and androgen receptor miR-21 promoter interaction.

Collaboration


Dive into the Margarita M. Ivanova's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yun Teng

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge