Network


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

Hotspot


Dive into the research topics where Mercedes Tkach is active.

Publication


Featured researches published by Mercedes Tkach.


Molecular and Cellular Biology | 2010

Progesterone Receptor Induces ErbB-2 Nuclear Translocation To Promote Breast Cancer Growth via a Novel Transcriptional Effect: ErbB-2 Function as a Coactivator of Stat3

Wendy Béguelin; María C. Díaz Flaqué; Cecilia J. Proietti; Florencia Cayrol; Martín A. Rivas; Mercedes Tkach; Cinthia Rosemblit; Johanna M. Tocci; Eduardo H. Charreau; Roxana Schillaci; Patricia V. Elizalde

ABSTRACT Progesterone receptor (PR) and ErbB-2 bidirectional cross talk participates in breast cancer development. Here, we identified a new mechanism of the PR and ErbB-2 interaction involving the PR induction of ErbB-2 nuclear translocation and the assembly of a transcriptional complex in which ErbB-2 acts as a coactivator of Stat3. We also highlighted that the function of ErbB-2 as a Stat3 coactivator drives progestin-induced cyclin D1 promoter activation. Notably, PR is also recruited together with Stat3 and ErbB-2 to the cyclin D1 promoter, unraveling a new and unexpected nonclassical PR genomic mechanism. The assembly of the nuclear Stat3/ErbB-2 transcriptional complex plays a key role in the proliferation of breast tumors with functional PR and ErbB-2. Our findings reveal a novel therapeutic intervention for PR- and ErbB-2-positive breast tumors via the specific blockage of ErbB-2 nuclear translocation.


PLOS ONE | 2012

Small Interfering RNA Targeted to IGF-IR Delays Tumor Growth and Induces Proinflammatory Cytokines in a Mouse Breast Cancer Model

Tiphanie Durfort; Mercedes Tkach; Mariya I. Meschaninova; Martín A. Rivas; Patricia V. Elizalde; Alya G. Venyaminova; Roxana Schillaci; Jean François

Insulin-like growth factor I (IGF-I) and its type I receptor (IGF-IR) play significant roles in tumorigenesis and in immune response. Here, we wanted to know whether an RNA interference approach targeted to IGF-IR could be used for specific antitumor immunostimulation in a breast cancer model. For that, we evaluated short interfering RNA (siRNAs) for inhibition of in vivo tumor growth and immunological stimulation in immunocompetent mice. We designed 2′-O-methyl-modified siRNAs to inhibit expression of IGF-IR in two murine breast cancer cell lines (EMT6, C4HD). Cell transfection of IGF-IR siRNAs decreased proliferation, diminished phosphorylation of downstream signaling pathway proteins, AKT and ERK, and caused a G0/G1 cell cycle block. The IGF-IR silencing also induced secretion of two proinflammatory cytokines, TNF- α and IFN-γ. When we transfected C4HD cells with siRNAs targeting IGF-IR, mammary tumor growth was strongly delayed in syngenic mice. Histology of developing tumors in mice grafted with IGF-IR siRNA treated C4HD cells revealed a low mitotic index, and infiltration of lymphocytes and polymorphonuclear neutrophils, suggesting activation of an antitumor immune response. When we used C4HD cells treated with siRNA as an immunogen, we observed an increase in delayed-type hypersensitivity and the presence of cytotoxic splenocytes against wild-type C4HD cells, indicative of evolving immune response. Our findings show that silencing IGF-IR using synthetic siRNA bearing 2′-O-methyl nucleotides may offer a new clinical approach for treatment of mammary tumors expressing IGF-IR. Interestingly, our work also suggests that crosstalk between IGF-I axis and antitumor immune response can mobilize proinflammatory cytokines.


Breast Cancer Research and Treatment | 2010

Transactivation of ErbB-2 induced by tumor necrosis factor α promotes NF-κB activation and breast cancer cell proliferation

Martín A. Rivas; Mercedes Tkach; Wendy Béguelin; Cecilia J. Proietti; Cinthia Rosemblit; Eduardo H. Charreau; Patricia V. Elizalde; Roxana Schillaci

Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine which, acting locally, induces tumor growth. Accumulating evidence, including our findings, showed that TNFα is mitogenic in breast cancer cells in vitro and in vivo. In the present study, we explored TNFα involvement on highly aggressive ErbB-2-overexpressing breast cancer cells. We found that TNFα induces ErbB-2 phosphorylation in mouse breast cancer C4HD cells and in the human breast cancer cell lines SK-BR-3 and BT-474. ErbB-2 phosphorylation at Tyr877 residue was mediated by TNFα-induced c-Src activation. Moreover, TNFα promoted ErbB-2/ErbB-3 heterocomplex formation, Akt activation and NF-κB transcriptional activation. Inhibition of ErbB-2 by addition of AG825, an epidermal growth factor receptor/ErbB-2-tyrosine kinase inhibitor, or knockdown of ErbB-2 by RNA interference strategy, blocked TNFα-induced NF-κB activation and proliferation. However, the humanized monoclonal antibody anti-ErbB-2 Herceptin could not inhibit TNFα ability to promote breast cancer growth. Interestingly, our work disclosed that TNFα is able to transactivate ErbB-2 and use it as an obligatory downstream signaling molecule in the generation of mitogenic signals. As TNFα has been shown to be present in the tumor microenvironment of a significant proportion of human infiltrating breast cancers, our findings would have clinical implication in ErbB-2-positive breast cancer treatment.


Journal of Immunology | 2012

Targeting Stat3 Induces Senescence in Tumor Cells and Elicits Prophylactic and Therapeutic Immune Responses against Breast Cancer Growth Mediated by NK Cells and CD4+ T Cells

Mercedes Tkach; Lorena M. Coria; Cinthia Rosemblit; A. Rivas; Cecilia J. Proietti; Wendy Béguelin; Isabel Frahm; Eduardo H. Charreau; Juliana Cassataro; Patricia V. Elizalde; Roxana Schillaci

Aberrant Stat3 activation and signaling contribute to malignant transformation by promoting cell cycle progression, inhibiting apoptosis, and mediating tumor immune evasion. Stat3 inhibition in tumor cells induces the expression of chemokines and proinflammatory cytokines, so we proposed to apply Stat3-inhibited breast cancer cells as a source of immunogens to induce an antitumor immune response. Studies were performed in two murine breast cancer models in which Stat3 is activated: progestin-dependent C4HD cells and 4T1 cells. We immunized BALB/c mice with irradiated cancer cells previously transfected with a dominant-negative Stat3 vector (Stat3Y705F) in either a prophylactic or a therapeutic manner. Prophylactic administration of breast cancer cells transfected with Stat3Y705F (Stat3Y705F-breast cancer cells) inhibited primary tumor growth compared with administration of empty vector-transfected cells in both models. In the 4T1 model, 50% of the challenged mice were tumor free, and the incidence of metastasis decreased by 90%. In vivo assays of C4HD tumors showed that the antitumor immune response involves the participation of CD4+ T cells and cytotoxic NK cells. Therapeutic immunization with Stat3Y705F-breast cancer cells inhibited tumor growth, promoted tumor cell differentiation, and decreased metastasis. Furthermore, inhibition of Stat3 activation in breast cancer cells induced cellular senescence, contributing to their immunogenic phenotype. In this work, we provide preclinical proof of concept that ablating Stat3 signaling in breast cancer cells results in an effective immunotherapy against breast cancer growth and metastasis. Moreover, our findings showing that Stat3 inactivation results in induction of a cellular senescence program disclose a potential mechanism for immunotherapy research.


Breast Cancer Research | 2013

Progesterone receptor assembly of a transcriptional complex along with activator protein 1, signal transducer and activator of transcription 3 and ErbB-2 governs breast cancer growth and predicts response to endocrine therapy

María Celeste Díaz Flaqué; Natalia M. Galigniana; Wendy Béguelin; Rocío Vicario; Cecilia J. Proietti; Rosalía Cordo Russo; Martín A. Rivas; Mercedes Tkach; Pablo Guzmán; Juan Carlos Roa; Esteban Maronna; Viviana Pineda; Sergio Muñoz; María Florencia Mercogliano; Eduardo H. Charreau; Patricio Yankilevich; Roxana Schillaci; Patricia V. Elizalde

IntroductionThe role of the progesterone receptor (PR) in breast cancer remains a major clinical challenge. Although PR induces mammary tumor growth, its presence in breast tumors is a marker of good prognosis. We investigated coordinated PR rapid and nonclassical transcriptional effects governing breast cancer growth and endocrine therapy resistance.MethodsWe used breast cancer cell lines expressing wild-type and mutant PRs, cells sensitive and resistant to endocrine therapy, a variety of molecular and cellular biology approaches, in vitro proliferation studies and preclinical models to explore PR regulation of cyclin D1 expression, tumor growth, and response to endocrine therapy. We investigated the clinical significance of activator protein 1 (AP-1) and PR interaction in a cohort of 99 PR-positive breast tumors by an immunofluorescence protocol we developed. The prognostic value of AP-1/PR nuclear colocalization in overall survival (OS) was evaluated using Kaplan-Meier method, and Cox model was used to explore said colocalization as an independent prognostic factor for OS.ResultsWe demonstrated that at the cyclin D1 promoter and through coordinated rapid and transcriptional effects, progestin induces the assembly of a transcriptional complex among AP-1, Stat3, PR, and ErbB-2 which functions as an enhanceosome to drive breast cancer growth. Our studies in a cohort of human breast tumors identified PR and AP-1 nuclear interaction as a marker of good prognosis and better OS in patients treated with tamoxifen (Tam), an anti-estrogen receptor therapy. Rationale for this finding was provided by our demonstration that Tam inhibits rapid and genomic PR effects, rendering breast cancer cells sensitive to its antiproliferative effects.ConclusionsWe here provided novel insight into the paradox of PR action as well as new tools to identify the subgroup of ER+/PR + patients unlikely to respond to ER-targeted therapies.


Steroids | 2011

Novel role of signal transducer and activator of transcription 3 as a progesterone receptor coactivator in breast cancer

Cecilia J. Proietti; Wendy Béguelin; María Celeste Díaz Flaqué; Florencia Cayrol; Martín A. Rivas; Mercedes Tkach; Eduardo H. Charreau; Roxana Schillaci; Patricia V. Elizalde

Interactions between progesterone receptor (PR) and signal transducer and activator of transcription 3 (Stat3)-mediated signaling pathways have already been described. In the present study, we explored the capacity of Stat3 to functionally interact with progesterone receptor (PR) and modulate PR transcriptional activation in breast cancer cells. We found that the synthetic progestin medroxyprogesterone acetate (MPA) induced the association of a PR/Stat3 complex in which Stat3 acts as a coactivator of PR. We demonstrated that Stat3 activation is required for MPA modulation of the endogenous genes bcl-X and p21(CIP1) which are involved in MPA-induced cell cycle regulation. Stat3 activity as a coactivator of PR was observed in both the classical and nonclassical ligand activated-PR transcriptional mechanisms, since the effects described were identified in the bcl-X promoter which contains a progesterone responsive element and in the p21(CIP1) promoter which carries Sp1 binding sites where PR is recruited via the transcription factor Sp1. The data herein presented identifies a potential therapeutic intervention for PR-positive breast tumors consisting of targeting Stat3 function or PR/Stat3 interaction which will result in the inhibition of PR function.


Breast Cancer Research | 2014

Progesterone receptor activation downregulates GATA3 by transcriptional repression and increased protein turnover promoting breast tumor growth

Franco Izzo; Florencia Mercogliano; Leandro Venturutti; Mercedes Tkach; Gloria Inurrigarro; Roxana Schillaci; Leandro Cerchietti; Patricia V. Elizalde; Cecilia J. Proietti

IntroductionThe transcription factor GATA3 is involved in mammary gland development and is crucial for the maintenance of the differentiated status of luminal epithelial cells. The role of GATA3 in breast cancer as a tumor suppressor has been established, although insights into the mechanism of GATA3 expression loss are still required.MethodsChromatin immunoprecipitation assays were conducted to study progestin modulation of recruitment of transcription factors to GATA3 promoter. We performed western blot and reverse RT-qPCR experiments to explore progestin regulation of GATA3 protein and mRNA expression respectively. Confocal microscopy and in vitro phosphorylation studies were conducted to examine progestin capacity to induce GATA3 serine phosphorylation in its 308 residue. GATA3 participation in progestin-induced breast cancer growth was addressed in in vitro proliferation and in vivo tumor growth experiments.ResultsIn this study, we demonstrate that progestin-activated progesterone receptor (PR) reduces GATA3 expression through regulation at the transcriptional and post-translational levels in breast cancer cells. In the former mechanism, the histone methyltransferase enhancer of zeste homolog 2 is co-recruited with activated PR to a putative progesterone response element in the GATA3 proximal promoter, increasing H3K27me3 levels and inducing chromatin compaction, resulting in decreased GATA3 mRNA levels. This transcriptional regulation is coupled with increased GATA3 protein turnover through progestin-induced GATA3 phosphorylation at serine 308 followed by 26S proteasome-mediated degradation. Both molecular mechanisms converge to accomplish decreased GATA3 expression levels in breast cancer cells upon PR activation. In addition, we demonstrated that decreased GATA3 levels are required for progestin-induced upregulation of cyclin A2, which mediates the G1 to S phase transition of the cell cycle and was reported to be associated with poor prognosis in breast cancer. Finally, we showed that downregulation of GATA3 is required for progestin stimulation of both in vitro cell proliferation and in vivo tumor growth.ConclusionsIn the present study, we reveal that progestin-induced PR activation leads to loss of GATA3 expression in breast cancer cells through transcriptional and post-translational regulation. Importantly, we demonstrate that GATA3 downregulation is required for progestin-induced upregulation of cyclin A2 and for progestin-induced in vitro and in vivo breast cancer cell growth.


Oncogene | 2015

Targeting ErbB-2 nuclear localization and function inhibits breast cancer growth and overcomes trastuzumab resistance.

R I Cordo Russo; Wendy Béguelin; M C Díaz Flaqué; Cristina Proietti; Leandro Venturutti; Natalia M. Galigniana; Mercedes Tkach; Pablo Guzmán; Juan Carlos Roa; N A O'Brien; Eduardo H. Charreau; Roxana Schillaci; Patricia V. Elizalde

Membrane overexpression of ErbB-2/HER2 receptor tyrosine kinase (membrane ErbB-2 (MErbB-2)) has a critical role in breast cancer (BC). We and others have also shown the role of nuclear ErbB-2 (NErbB-2) in BC, whose presence we identified as a poor prognostic factor in MErbB-2-positive tumors. Current anti-ErbB-2 therapies, as with the antibody trastuzumab (Ttzm), target only MErbB-2. Here, we found that blockade of NErbB-2 action abrogates growth of BC cells, sensitive and resistant to Ttzm, in a scenario in which ErbB-2, ErbB-3 and Akt are phosphorylated, and ErbB-2/ErbB-3 dimers are formed. Also, inhibition of NErbB-2 presence suppresses growth of a preclinical BC model resistant to Ttzm. We showed that at the cyclin D1 promoter, ErbB-2 assembles a transcriptional complex with Stat3 (signal transducer and activator of transcription 3) and ErbB-3, another member of the ErbB family, which reveals the first nuclear function of ErbB-2/ErbB-3 dimer. We identified NErbB-2 as the major proliferation driver in Ttzm-resistant BC, and demonstrated that Ttzm inability to disrupt the Stat3/ErbB-2/ErbB-3 complex underlies its failure to inhibit growth. Furthermore, our results in the clinic revealed that nuclear interaction between ErbB-2 and Stat3 correlates with poor overall survival in primary breast tumors. Our findings challenge the paradigm of anti-ErbB-2 drug design and highlight NErbB-2 as a novel target to overcome Ttzm resistance.


Journal of Immunology | 2016

A Brucella spp. Protease Inhibitor Limits Antigen Lysosomal Proteolysis, Increases Cross-Presentation, and Enhances CD8+ T Cell Responses

Lorena M. Coria; Andrés E. Ibañez; Mercedes Tkach; Florencia Sabbione; Laura Bruno; Marianela Verónica Carabajal; Paula M. Berguer; Paula Barrionuevo; Roxana Schillaci; Analía S. Trevani; Guillermo H. Giambartolomei; Karina A. Pasquevich; Juliana Cassataro

In this study, we demonstrate that the unlipidated (U) outer membrane protein (Omp) 19 from Brucella spp. is a competitive inhibitor of human cathepsin L. U-Omp19 inhibits lysosome cathepsins and APC-derived microsome activity in vitro and partially inhibits lysosomal cathepsin L activity within live APCs. Codelivery of U-Omp19 with the Ag can reduce intracellular Ag digestion and increases Ag half-life in dendritic cells (DCs). U-Omp19 retains the Ag in Lamp-2+ compartments after its internalization and promotes a sustained expression of MHC class I/peptide complexes in the cell surface of DCs. Consequently, U-Omp19 enhances Ag cross-presentation by DCs to CD8+ T cells. U-Omp19 s.c. delivery induces the recruitment of CD11c+CD8α+ DCs and monocytes to lymph nodes whereas it partially limits in vivo Ag proteolysis inside DCs. Accordingly, this protein is able to induce CD8+ T cell responses in vivo against codelivered Ag. Antitumor responses were elicited after U-Omp19 coadministration, increasing survival of mice in a murine melanoma challenge model. Collectively, these results indicate that a cysteine protease inhibitor from bacterial origin could be a suitable component of vaccine formulations against tumors.


Cancer Research | 2012

Abstract 1549: Targeting Stat3 induces senescence in breast cancer cells and elicits an immune response inhibiting tumor growth and metastasis

Mercedes Tkach; Martín A. Rivas; Cecilia J. Proietti; María C. Díaz Flaqué; Isabel Frahm; Eduardo H. Charreau; Patricia V. Elizalde; Roxana Schillaci

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Having in mind that Stat3 inhibition in tumor cells induces the expression of chemokines and pro-inflammatory cytokines, we proposed the use of Stat3-inhibited breast cancer cells as a source of immunogens to induce an anti-tumor immune response. We have demonstrated that the administration of irradiated breast cancer cells that express a dominant negative (DN) form of Stat3 (Stat3Y705F-breast cancer cells) provides protection against the murine progestin-dependent C4HD tumor, through the activation of CD4+ T cells and cytotoxic natural killer (NK) cells. To extend our results to different breast cancer models, we worked with the hormone independent 4T1 mammary carcinoma cell line that displays constitutive activation of Stat3. Immunization with irradiated Stat3Y705F-4T1 cells prevented wild-type 4T1 tumor development in 50% of the challenged mice (P<0.001), and the tumor-bearing mice displayed tumors of smaller size (81% decrease in tumor volume, P<0.001) when compared to mice injected with pcDNA3.1-4T1 cells. Moreover, the number of metastasis per lung decreased by 90% in Stat3Y705F-4T1-immunized animals (P<0.05). When we analyzed the tumor milieu composition by flow cytometry, we observed that Stat3Y705F-4T1 immunized animals displayed an increase in the percentage of tumor infiltrating NK cells (CD3-DX5+) and a decrease in tumor infiltrating T regulatory lymphocytes (CD4+CD25+FoxP3+), compared to pcDNA3.1-4T1-immunized mice. In order to evaluate if this vaccination may be effective in a therapeutic setting, we immunized mice with irradiated Stat3Y705F-4T1 or pcDNA3.1-4T1 cells, 4, 11 and 18 days after challenging with 4T1 cells. On day 35, we observed a significant decrease on tumor volume and growth rate in Stat3Y705F-4T1 cell-immunized animals, when compared to mice immunized with pcDNA3.1-4T1 cells (37.5%, P<0.05) and a decrease in the number of metastasis per lung (P<0.05). On the other hand, cellular senescence is an important mechanism of tumor regression upon oncogene inactivation that leads to the secretion of pro-inflammatory cytokines that resemble the ones we found after blocking Stat3. Therefore, we wondered whether Stat3 inhibition could drive a senescence program. Inhibition of Stat3 in murine C4HD and 4T1 cells by transfection with Stat3Y705F, or Stat3 silencing by siRNA, resulted in increased senescence-associated-β-galactosidase (SA-α-gal) accumulation and increased expression of the senescence-associated markers p15INK4b and p16INK4a. As cellular senescence is associated to chromatin changes, we studied heterochromatin formation and observed an increase of trimethyl-K4 histone H3 upon Stat3 inhibition. As a whole, our findings indicate that Stat3 inhibition in breast cancer cells induce an increase in immunogenicity capable of eliciting an anti tumor immune response, presumably through the activation of a senescence program. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1549. doi:1538-7445.AM2012-1549

Collaboration


Dive into the Mercedes Tkach's collaboration.

Top Co-Authors

Avatar

Roxana Schillaci

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Patricia V. Elizalde

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Cecilia J. Proietti

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Eduardo H. Charreau

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Martín A. Rivas

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Wendy Béguelin

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

María Celeste Díaz Flaqué

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cinthia Rosemblit

Instituto de Biología y Medicina Experimental

View shared research outputs
Top Co-Authors

Avatar

Franco Izzo

Instituto de Biología y Medicina Experimental

View shared research outputs
Researchain Logo
Decentralizing Knowledge