16 research outputs found

    Modalities of ERα36 regulation and their consequences on the pathophysiology of the mammary gland

    No full text
    Les récepteurs nucléaires aux œstrogènes, ERα66 et ERβ1, sont les principaux médiateurs des effets des œstrogènes. Ces hormones régulent le développement physiologique de la glande mammaire mais participent aussi à la progression du cancer sein. L’expression d’ERα66 est d’ailleurs utilisée dans la classification moléculaire des tumeurs mammaires afin d’orienter la stratégie thérapeutique. Depuis son clonage, le variant des récepteurs alpha aux œstrogènes, ERα36, a été principalement décrit dans la littérature pour son rôle dans la progression des tumeurs mammaires et dans l’acquisition de résistances aux anti-œstrogènes comme le Tamoxifène. Si une forte expression d’ERα36 dans les cellules cancéreuses mammaires apparaît nettement comme un facteur de mauvais pronostic, peu de données sont disponibles concernant son rôle dans le développement de la glande mammaire saine. C’est pourquoi le premier objectif de ce travail était de déterminer le rôle d’ERα36 dans le développement physiologique de cette glande. Grâce à une approche pluridisciplinaire, incluant des études in vivo sur un modèle de souris transgéniques MMTV-ERα36 et des études in vitro et in silico sur des cellules épithéliales mammaires immortalisées, nous avons montré que l’expression d’ERα36 perturbe le phénotype des cellules épithéliales mammaires et conduit à l’apparition d’altérations structurales des canaux mammaires à l’âge adulte. De plus, nous avons mis en évidence que les alkylphénols, qui sont des perturbateurs endocriniens œstrogèno-mimétiques, stimulent l’expression endogène de ce variant dans les cellules MCF-10A et augmentent leurs capacités migratoires sans pour autant amplifier les effets d’ERα36 sur l’histologie des canaux mammaires. En parallèle, afin de mieux comprendre l’implication d’ERα36 au moment de l’initiation et de la progression tumorale, nous avons étudié les modalités de régulation de l’expression de ce variant dans les cellules cancéreuses mammaires. Les résultats obtenus indiquent que l’expression d’ERα36 est positivement corrélée au statut de méthylation de sa région promotrice et que l’ARNm codant ce variant est la cible d’hsa-miR136-5p. Enfin, le dernier objectif de ce travail était de développer une approche visant à identifier in silico de nouveaux partenaires d’ERα36. L’ensemble de ce travail s’inscrit dans une démarche de raffinement de la classification moléculaire actuelle des tumeurs mammaires en y ajoutant une composante associée à l’expression d’ERα36.The estrogen nuclear receptors, represented by the canonical forms ERα66 and ERβ1, are the main mediators of the estrogenic effects in mammals. These hormones, which regulate the physiological development of the mammary gland, participate in the initiation and progression of breast cancer. In fact, ERα66 expression is a key molecular classifier of breast tumors used in order to guide the therapeutic strategies toward hormonotherapy. However, in 30% of cases, therapeutic failures are observed, which highlights the importance of identifying new biomarkers. The estrogen receptor variant, ERα36, has been cloned in 2005 and mainly described in the literature to be involved in the progression of mammary tumors and in the acquired resistance to anti-estrogen drugs, such as Tamoxifen. Even if a high expression of ERα36 in breast cancer cells appears to be associated with a poor prognosis, few data are available concerning its role in the normal development of the mammary gland. Therefore, the aim of this work was to determine the role of ERα36 in the physiological development of the mammary gland. Thanks to a multidisciplinary approach, that combines in vivo studies on MMTV-ERα36 transgenic mice, and in vitro and in silico studies on immortalized normal epithelial mammary cells (MCF-10A), we showed that ERα36 expression is sufficient to disturb the mammary epithelial cells phenotype, leading to the emergence of structural alterations of mammary ducts at adulthood. Moreover, we showed that exposure to the estrogen mimicking compounds alkylphenols stimulates the endogenous expression of this variant in MCF-10A cells, and increases their migratory ability. Then, in order to get a better understanding of ERα36 contribution to tumor initiation and/or progression, we studied classical and epigenetic regulation of this variant expression in breast cancer cells. Our results show that ERα36 expression is positively correlated with the methylation status of its promoter region, and that the ERα36 mRNA is the target of the microRNA, has-miR-136-5p. Finally, the last aim of this work was to develop a bioinformatic approach in order to study the ERα36 partners. To summarize, all of this work falls within a need of the current breast tumor molecular classification refinement by adding a component related with ERα36 expression

    Modalités de régulation d’ERα36 et leurs conséquences sur la physiopathologie de la glande mammaire

    No full text
    The estrogen nuclear receptors, represented by the canonical forms ERα66 and ERβ1, are the main mediators of the estrogenic effects in mammals. These hormones, which regulate the physiological development of the mammary gland, participate in the initiation and progression of breast cancer. In fact, ERα66 expression is a key molecular classifier of breast tumors used in order to guide the therapeutic strategies toward hormonotherapy. However, in 30% of cases, therapeutic failures are observed, which highlights the importance of identifying new biomarkers. The estrogen receptor variant, ERα36, has been cloned in 2005 and mainly described in the literature to be involved in the progression of mammary tumors and in the acquired resistance to anti-estrogen drugs, such as Tamoxifen. Even if a high expression of ERα36 in breast cancer cells appears to be associated with a poor prognosis, few data are available concerning its role in the normal development of the mammary gland. Therefore, the aim of this work was to determine the role of ERα36 in the physiological development of the mammary gland. Thanks to a multidisciplinary approach, that combines in vivo studies on MMTV-ERα36 transgenic mice, and in vitro and in silico studies on immortalized normal epithelial mammary cells (MCF-10A), we showed that ERα36 expression is sufficient to disturb the mammary epithelial cells phenotype, leading to the emergence of structural alterations of mammary ducts at adulthood. Moreover, we showed that exposure to the estrogen mimicking compounds alkylphenols stimulates the endogenous expression of this variant in MCF-10A cells, and increases their migratory ability. Then, in order to get a better understanding of ERα36 contribution to tumor initiation and/or progression, we studied classical and epigenetic regulation of this variant expression in breast cancer cells. Our results show that ERα36 expression is positively correlated with the methylation status of its promoter region, and that the ERα36 mRNA is the target of the microRNA, has-miR-136-5p. Finally, the last aim of this work was to develop a bioinformatic approach in order to study the ERα36 partners. To summarize, all of this work falls within a need of the current breast tumor molecular classification refinement by adding a component related with ERα36 expression.Les récepteurs nucléaires aux œstrogènes, ERα66 et ERβ1, sont les principaux médiateurs des effets des œstrogènes. Ces hormones régulent le développement physiologique de la glande mammaire mais participent aussi à la progression du cancer sein. L’expression d’ERα66 est d’ailleurs utilisée dans la classification moléculaire des tumeurs mammaires afin d’orienter la stratégie thérapeutique. Depuis son clonage, le variant des récepteurs alpha aux œstrogènes, ERα36, a été principalement décrit dans la littérature pour son rôle dans la progression des tumeurs mammaires et dans l’acquisition de résistances aux anti-œstrogènes comme le Tamoxifène. Si une forte expression d’ERα36 dans les cellules cancéreuses mammaires apparaît nettement comme un facteur de mauvais pronostic, peu de données sont disponibles concernant son rôle dans le développement de la glande mammaire saine. C’est pourquoi le premier objectif de ce travail était de déterminer le rôle d’ERα36 dans le développement physiologique de cette glande. Grâce à une approche pluridisciplinaire, incluant des études in vivo sur un modèle de souris transgéniques MMTV-ERα36 et des études in vitro et in silico sur des cellules épithéliales mammaires immortalisées, nous avons montré que l’expression d’ERα36 perturbe le phénotype des cellules épithéliales mammaires et conduit à l’apparition d’altérations structurales des canaux mammaires à l’âge adulte. De plus, nous avons mis en évidence que les alkylphénols, qui sont des perturbateurs endocriniens œstrogèno-mimétiques, stimulent l’expression endogène de ce variant dans les cellules MCF-10A et augmentent leurs capacités migratoires sans pour autant amplifier les effets d’ERα36 sur l’histologie des canaux mammaires. En parallèle, afin de mieux comprendre l’implication d’ERα36 au moment de l’initiation et de la progression tumorale, nous avons étudié les modalités de régulation de l’expression de ce variant dans les cellules cancéreuses mammaires. Les résultats obtenus indiquent que l’expression d’ERα36 est positivement corrélée au statut de méthylation de sa région promotrice et que l’ARNm codant ce variant est la cible d’hsa-miR136-5p. Enfin, le dernier objectif de ce travail était de développer une approche visant à identifier in silico de nouveaux partenaires d’ERα36. L’ensemble de ce travail s’inscrit dans une démarche de raffinement de la classification moléculaire actuelle des tumeurs mammaires en y ajoutant une composante associée à l’expression d’ERα36

    Dual Epigenetic Regulation of ERα36 Expression in Breast Cancer Cells

    No full text
    Breast cancer remains the major cause of cancer-induced morbidity and mortality in women. Among the different molecular subtypes, luminal tumors yet considered of good prognosis often develop acquired resistance to endocrine therapy. Recently, misregulation of ERα36 was reported to play a crucial role in this process. High expression of this ERα isoform was associated to preneoplastic phenotype in mammary epithelial cells, disease progression, and enhanced resistance to therapeutic agents in breast tumors. In this study, we identified two mechanisms that could together contribute to ERα36 expression regulation. We first focused on hsa-miR-136-5p, an ERα36 3’UTR-targeting microRNA, the expression of which inversely correlated to the ERα36 one in breast cancer cells. Transfection of hsa-miR136-5p mimic in MCF-7 cells resulted in downregulation of ERα36. Moreover, the demethylating agent decitabine was able to stimulate hsa-miR-136-5p endogenous expression, thus indirectly decreasing ERα36 expression and counteracting tamoxifen-dependent stimulation. The methylation status of ERα36 promoter also directly modulated its expression level, as demonstrated after decitabine treatment of breast cancer cell and confirmed in a set of tumor samples. Taken together, these results open the way to a direct and an indirect ERα36 epigenetic modulation by decitabine as a promising clinical strategy to counteract acquired resistance to treatment and prevent relapse

    Structure, Activity, and Function of PRMT1

    No full text
    International audiencePRMT1, the major protein arginine methyltransferase in mammals, catalyzes monomethylation and asymmetric dimethylation of arginine side chains in proteins. Initially described as a regulator of chromatin dynamics through the methylation of histone H4 at arginine 3 (H4R3), numerous non-histone substrates have since been identified. The variety of these substrates underlines the essential role played by PRMT1 in a large number of biological processes such as transcriptional regulation, signal transduction or DNA repair. This review will provide an overview of the structural, biochemical and cellular features of PRMT1. After a description of the genomic organization and protein structure of PRMT1, special consideration was given to the regulation of PRMT1 enzymatic activity. Finally, we discuss the involvement of PRMT1 in embryonic development, DNA damage repair, as well as its participation in the initiation and progression of several types of cancers

    Non-genomic signaling of steroid receptors in cancer

    No full text
    International audienceSteroid receptors (SRs) are members of the nuclear receptor family, which are ligand-activated transcription factors. SRs regulate many physiological functions including development and reproduction, though they can also be involved in several pathologies, especially cancer. Highly controlled cellular responses to steroids involve transcriptional regulation (genomic activity) combined with direct activation of signaling cascades (non-genomic activity). Non-genomic signaling has been extensively studied in cancer, mainly in breast cancer for ER and PR, and prostate cancer for AR. Even though most of the studies have been conducted in cells, some of them have been confirmed in vivo, highlighting the relevance of this pathway in cancer. This review provides an overview of the current and emerging knowledge on non-genomic signaling with a focus on breast and prostate cancers and its clinical relevance. A thorough understanding of ER, PR, AR and GR non-genomic pathways may open new perspectives for the development of therapeutic strategies

    Mammary epithelial cell phenotype disruption in vitro and in vivo through ERalpha36 overexpression.

    No full text
    Estrogen receptor alpha 36 (ERα36) is a variant of the canonical estrogen receptor alpha (ERα66), widely expressed in hormone sensitive cancer cells and whose high expression level correlates with a poor survival prognosis for breast cancer patients. While ERα36 activity have been related to breast cancer progression or acquired resistance to treatment, expression level and location of ERα36 are poorly documented in the normal mammary gland. Therefore, we explored the consequences of a ERα36 overexpression in vitro in MCF-10A normal mammary epithelial cells and in vivo in a unique model of MMTV-ERα36 transgenic mouse strain wherein ERα36 mRNA was specifically expressed in the mammary gland. By a combination of bioinformatics and computational analyses of microarray data, we identified hierarchical gene networks, downstream of ERα36 and modulated by the JAK2/STAT3 signaling pathway. Concomitantly, ERα36 overexpression lowered proliferation rate but enhanced migration potential and resistance to staurosporin-induced apoptosis of the MCF-10A cell line. In vivo, ERα36 expression led to duct epithelium thinning and disruption in adult but not in prepubescent mouse mammary gland. These phenotypes correlated with a loss of E-cadherin expression. Here, we show that an enhanced expression of ERα36 is sufficient, by itself, to disrupt normal breast epithelial phenotype in vivo and in vitro through a dominant-positive effect on nongenomic estrogen signaling pathways. These results also suggest that, in the presence of adult endogenous steroid levels, ERα36 overexpression in vivo contributes to alter mammary gland architecture which may support pre-neoplastic lesion and augment breast cancer risk

    ERα36 overexpression enhances migratory potential.

    No full text
    <p>A. A wound was performed on a confluent monolayer culture of MCF-10A/Zeo and MCF-10A/ERα36 cells. Histogram depicts the wound healing when measured after a 6-hour culture (left panel). A representative picture of the migrating MCF-10A/ERα36 cells is presented in the right panel. Each bar represents mean ± S.E.M. N = 5. *: <i>P</i> <0.05. B-C. MCF-10A/Zeo and MCF-10A/ERα36 were cultured for 24 hours. B. CDH1 and CDH2 gene expression was measured by RT-PCR analysis. The housekeeping gene RPLPO was used as the reference gene. Each bar represents mean ± S.E.M. N = 3. *: <i>P</i> <0.05. **: <i>P</i> <0.01. C. Expression of cell-cell adhesion proteins was studied by immunofluorescence with specific antibodies: anti-β-catenin, anti-E-cadherin and anti-N-cadherin (AlexaFluor 555). Average fluorescent signal intensities are quantified from 5 cells from 5 fields per condition. Each bar represents mean ± S.E.M. N = 3. *: <i>P</i> <0.05, ** <i>P</i> <0.01. </p

    ERα36 overexpression stimulates apoptosis resistance.

    No full text
    <p>MCF-10A/Zeo and MCF-10A/ERα36 cells were exposed to 0.25μM staurosporin (STS) or vehicle (Veh) for 6 hours. A. Cleavage of PARP1 (cPARP1), Caspase 7 (cCasp7) and Caspase 3 (cCasp3) were evaluated with specific antibodies (left panel). GAPDH was used as a loading control. Results depicted in the corresponding histogram are represented as STS versus Vehicle ratio (right panel). ERα36 overexpression triggered a significant 34%, 60% and 30% decrease of PARP1, Caspase 7 and Caspase 3 cleavage, respectively. Each bar represents mean ± S.E.M. N = 4. *: <i>P</i> <0.05. B. Cytochrome c (Cyt. C) release (red, AlexaFluor 555) and DNA fragmentation were respectively assessed by immunofluorescence and TUNEL assay after STS exposure in MCF-10A/Zeo and MCF-10A/ERα36 cells (left panel), then quantified as shown in the corresponding histogram (right panel). No cytochrome c release or DNA fragmentation can be detected in untreated cells (not shown). Each bar represents mean ± S.E.M. N = 3. *: <i>P</i> <0.05, ***: <i>P</i> <0.001.</p
    corecore