Network


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

Hotspot


Dive into the research topics where Cécile Rochette-Egly is active.

Publication


Featured researches published by Cécile Rochette-Egly.


Molecular Aspects of Medicine | 2015

Retinoic acid receptors: From molecular mechanisms to cancer therapy

Alessandra di Masi; Loris Leboffe; Elisabetta De Marinis; Francesca Pagano; Laura Cicconi; Cécile Rochette-Egly; Francesco Lo-Coco; Paolo Ascenzi; Clara Nervi

Retinoic acid (RA), the major bioactive metabolite of retinol or vitamin A, induces a spectrum of pleiotropic effects in cell growth and differentiation that are relevant for embryonic development and adult physiology. The RA activity is mediated primarily by members of the retinoic acid receptor (RAR) subfamily, namely RARα, RARβ and RARγ, which belong to the nuclear receptor (NR) superfamily of transcription factors. RARs form heterodimers with members of the retinoid X receptor (RXR) subfamily and act as ligand-regulated transcription factors through binding specific RA response elements (RAREs) located in target genes promoters. RARs also have non-genomic effects and activate kinase signaling pathways, which fine-tune the transcription of the RA target genes. The disruption of RA signaling pathways is thought to underlie the etiology of a number of hematological and non-hematological malignancies, including leukemias, skin cancer, head/neck cancer, lung cancer, breast cancer, ovarian cancer, prostate cancer, renal cell carcinoma, pancreatic cancer, liver cancer, glioblastoma and neuroblastoma. Of note, RA and its derivatives (retinoids) are employed as potential chemotherapeutic or chemopreventive agents because of their differentiation, anti-proliferative, pro-apoptotic, and anti-oxidant effects. In humans, retinoids reverse premalignant epithelial lesions, induce the differentiation of myeloid normal and leukemic cells, and prevent lung, liver, and breast cancer. Here, we provide an overview of the biochemical and molecular mechanisms that regulate the RA and retinoid signaling pathways. Moreover, mechanisms through which deregulation of RA signaling pathways ultimately impact on cancer are examined. Finally, the therapeutic effects of retinoids are reported.


Cancer Research | 2009

Inhibition of the Peptidyl-Prolyl-Isomerase Pin1 Enhances the Responses of Acute Myeloid Leukemia Cells to Retinoic Acid via Stabilization of RARα and PML-RARα

Maurizio Gianni; Andrea Boldetti; Valeria Guarnaccia; Alessandro Rambaldi; Edoardo Parrella; Ivan Raska; Cécile Rochette-Egly; Giannino Del Sal; Alessandra Rustighi; Mineko Terao; Enrico Garattini

The peptidyl-prolyl-isomerase Pin1 interacts with phosphorylated proteins, altering their conformation. The retinoic acid receptor RARalpha and the acute-promyelocytic-leukemia-specific counterpart PML-RARalpha directly interact with Pin1. Overexpression of Pin1 inhibits ligand-dependent activation of RARalpha and PML-RARalpha. Inhibition is relieved by Pin1-targeted short interfering RNAs and by pharmacologic inhibition of the catalytic activity of the protein. Mutants of Pin1 catalytically inactive or defective for client-protein-binding activity are incapable of inhibiting ligand-dependent RARalpha transcriptional activity. Functional inhibition of RARalpha and PML-RARalpha by Pin1 correlates with degradation of the nuclear receptors via the proteasome-dependent pathway. In the acute myelogenous leukemia cell lines HL-60 and NB4, Pin1 interacts with RARalpha in a constitutive fashion. Suppression of Pin1 by a specific short hairpin RNA in HL-60 or NB4 cells stabilizes RARalpha and PML-RARalpha, resulting in increased sensitivity to the cytodifferentiating and antiproliferative activities of all-trans retinoic acid. Treatment of the two cell lines and freshly isolated acute myelogenous leukemia blasts (M1 to M4) with ATRA and a pharmacologic inhibitor of Pin1 causes similar effects. Our results add a further layer of complexity to the regulation of nuclear retinoic acid receptors and suggest that Pin1 represents an important target for strategies aimed at increasing the therapeutic index of retinoids.


Cellular Signalling | 2012

Regulation of the transcriptional activity of nuclear receptors by the MEK/ERK1/2 pathway

Fabien Zassadowski; Cécile Rochette-Egly; Christine Chomienne; Bruno Cassinat

Cells undergo continuous and simultaneous external influences regulating their behavior. As an example, during differentiation, they go through different stages of maturation and gene expression is regulated by several simultaneous signaling pathways. We often tend at separating the nuclear pathways from the signaling ones initiated at membrane receptors. However, it is essential to keep in mind that all these pathways are interconnected to achieve a fine regulation of cell functions. The regulation of transcription by nuclear receptors has been thoroughly studied, but it now appears that a critical level of this regulation involves the action of several kinases that target the nuclear receptors themselves as well as their partners. The purpose of this review is to highlight the importance of one family of the mitogen-activated protein kinase (MAPK) superfamily, the MEK/ERK1/2 pathway, in the transcriptional activity of nuclear receptors.


Molecular Pharmacology | 2006

Antitumor activity of the retinoid-related molecules (E)-3-(4'-hydroxy-3'-adamantylbiphenyl-4-yl)acrylic acid (ST1926) and 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid (CD437) in F9 teratocarcinoma : Role of retinoic acid receptor gamma and retinoid-independent pathways

Edoardo Parrella; Maurizio Gianni; Maddalena Fratelli; Maria Monica Barzago; Ivan Raska; Luisa Diomede; Mami Kurosaki; Claudio Pisano; Paolo Carminati; Lucio Merlini; Sabrina Dallavalle; Michele Tavecchio; Cécile Rochette-Egly; Mineko Terao; Enrico Garattini

The retinoid-related molecules (RRMs) ST1926 [(E)-3-(4′-hydroxy-3′-adamantylbiphenyl-4-yl)acrylic acid] and CD437 (6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid) are promising anticancer agents. We compared the retinoic acid receptor (RAR) trans-activating properties of the two RRMs and all-trans-retinoic acid (ATRA). ST1926 and CD437 are better RARγ agonists than ATRA. We used three teratocarcinoma cell lines to evaluate the significance of RARγ in the activity of RRMs: F9-wild type (WT); F9γ-/-, lacking the RARγ gene; F9γ51, aF9γ-/-derivative, complemented for the RARγ deficit. Similar to ATRA, ST1926 and CD437 activate cytodifferentiation only in F9-WT cells. Unlike ATRA, ST1926 and CD437 arrest cells in the G2/M phase of the cell cycle and induce apoptosis in all F9 cell lines. Our data indicate that RARγ and the classic retinoid pathway are not relevant for the antiproliferative and apoptotic activities of RRMs in vitro. Increases in cytosolic calcium are fundamental for apoptosis, in that intracellular calcium chelators abrogate the process. Comparison of the gene expression profiles associated with ST1926 and ATRA in F9-WT and F9γ-/-indicates that the RRM activates a conspicuous nonretinoid response in addition to the classic and RAR-dependent pathway. The pattern of genes regulated by ST1926 selectively, in a RARγ-independent manner, provides novel insights into the possible molecular determinants underlying the activity of RRMs in vitro. Furthermore, it suggests that RARγ-dependent responses are relevant to the activity of RRMs in vivo. Indeed, the receptor hinders the antitumor activity in vivo, in that both syngeneic and immunosuppressed SCID mice bearing F9γ-/- tumors have increased life spans after treatment with ST1926 and CD437 relative to their F9-WT counterparts.


Journal of Biological Chemistry | 2009

SUG-1 Plays Proteolytic and Non-proteolytic Roles in the Control of Retinoic Acid Target Genes via Its Interaction with SRC-3

Christine Ferry; Maurizio Gianni; Sébastien Lalevée; Nathalie Bruck; Jean Luc Plassat; Ivan Raska; Enrico Garattini; Cécile Rochette-Egly

Nuclear retinoic acid receptor α (RARα) activates gene expression through dynamic interactions with coregulatory protein complexes, the assembly of which is directed by the ligand and the AF-2 domain of RARα. Then RARα and its coactivator SRC-3 are degraded by the proteasome. Recently it has emerged that the proteasome also plays a key role in RARα-mediated transcription. Here we show that SUG-1, one of the six ATPases of the 19 S regulatory complex of the 26 S proteasome, interacts with SRC-3, is recruited at the promoters of retinoic acid (RA) target genes, and thereby participates to their transcription. In addition, SUG-1 also mediates the proteasomal degradation of SRC-3. However, when present in excess amounts, SUG-1 blocks the activation of RARα target genes and the degradation of RARα that occurs in response to RA, via its ability to interfere with the recruitment of SRC-3 and other coregulators at the AF-2 domain of RARα. We propose a model in which the ratio between SUG-1 and SRC-3 is crucial for the control of RARα functioning. This study provides new insights into how SUG-1 has a unique role in linking the transcription and degradation processes via its ability to interact with SRC-3.


Journal of Cell Science | 2014

Genes involved in cell adhesion and signaling: a new repertoire of retinoic acid receptor target genes in mouse embryonic fibroblasts

Ziad Al Tanoury; Aleksandr Piskunov; Dina Andriamoratsiresy; Samia Gaouar; Régis Lutzing; Tao Ye; Bernard Jost; Céline Keime; Cécile Rochette-Egly

ABSTRACT Nuclear retinoic acid (RA) receptors (RAR&agr;, &bgr; and &ggr;) are ligand-dependent transcription factors that regulate the expression of a battery of genes involved in cell differentiation and proliferation. They are also phosphoproteins and we previously showed the importance of their phosphorylation in their transcriptional activity. In the study reported here, we conducted a genome-wide analysis of the genes that are regulated by RARs in mouse embryonic fibroblasts (MEFs) by comparing wild-type MEFs to MEFs lacking the three RARs. We found that in the absence of RA, RARs control the expression of several gene transcripts associated with cell adhesion. Consequently the knockout MEFs are unable to adhere and to spread on substrates and they display a disrupted network of actin filaments, compared with the WT cells. In contrast, in the presence of the ligand, RARs control the expression of other genes involved in signaling and in RA metabolism. Taking advantage of rescue cell lines expressing the RAR&agr; or RAR&ggr; subtypes (either wild-type or mutated at the N-terminal phosphorylation sites) in the null background, we found that the expression of RA-target genes can be controlled either by a specific single RAR or by a combination of RAR isotypes, depending on the gene. We also selected genes that require the phosphorylation of the receptors for their regulation by RA. Our results increase the repertoire of genes that are regulated by RARs and highlight the complexity and diversity of the transcriptional programs regulated by RARs, depending on the gene.


Leukemia | 2012

p38αMAPK interacts with and inhibits RARα: suppression of the kinase enhances the therapeutic activity of retinoids in acute myeloid leukemia cells.

M Gianni; M Peviani; N Bruck; Alessandro Rambaldi; G Borleri; M Terao; M Kurosaki; G Paroni; Cécile Rochette-Egly; E Garattini

All-trans retinoic acid (ATRA) is the only clinically useful differentiating agent, being used in the treatment of acute promyelocytic leukemia (APL). The use of ATRA in other types of acute myelogenous leukemia (AML) calls for the identification of novel strategies aimed at increasing its therapeutic activity. Here, we provide evidence that pharmacological inhibition of the mitogen-activated protein kinase, p38α, or silencing of the corresponding gene sensitizes APL and AML cell lines, as well as primary cultures of AML blasts to the anti-proliferative and cyto-differentiating activity of ATRA and synthetic retinoids. P38α inhibits ligand-dependent transactivation of the nuclear retinoic acid receptor, RARα, and the derived chimeric protein expressed in the majority of APL cases, PML-RARα. Inhibition is the consequence of ligand-independent binding of p38α, which results in stabilization of RARα and PML-RARα via blockade of their constitutive degradation by the proteasome. The inhibitory effect requires a catalytically active p38α and direct physical interaction with RARα and PML-RARα. Ser-369 in the E-region of RARα is essential for the binding of p38α and the ensuing functional effects on the activity of the receptor.


Sub-cellular biochemistry | 2014

Nuclear and Extra-Nuclear Effects of Retinoid Acid Receptors: How They Are Interconnected

Aleksandr Piskunov; Ziad Al Tanoury; Cécile Rochette-Egly

The nuclear retinoic acid receptors (RAR α, β and γ) and their isoforms are ligand-dependent regulators of transcription Transcription , which mediate the effects of all-trans retinoic acid (RA), the active endogenous metabolite of Vitamin A. They heterodimerize with Retinoid X Receptors (RXRs α, β and γ), and regulate the expression of a battery of target genes Target genes involved in cell growth and differentiation Differentiation . During the two last decades, the description of the crystallographic structures of RARs, the characterization of the polymorphic response elements of their target genes Target genes , and the identification of the multiprotein complexes involved in their transcriptional activity have provided a wealth of information on their pleiotropic effects. However, the regulatory scenario became even more complicated once it was discovered that RARs are phosphoproteins and that RA can activate kinase signaling cascades via a pool of RARs present in membrane lipid rafts. Now it is known that these RA-activated kinases Kinases translocate to the nucleus where they phosphorylate RARs and other retinoid signaling factors. The phosphorylation Phosphorylation state of the RARs dictates whether the transcriptional programs which are known to be induced by RA are facilitated and/or switched on. Thus, kinase signaling pathways appear to be crucial for fine-tuning the appropriate physiological activity of RARs.


PLOS ONE | 2016

Phosphoproteome and Transcriptome of RA-Responsive and RA-Resistant Breast Cancer Cell Lines.

Marilyn Carrier; Mathilde Joint; Régis Lutzing; Adeline Page; Cécile Rochette-Egly

Retinoic acid (RA), the main active vitamin A metabolite, controls multiple biological processes such as cell proliferation and differentiation through genomic programs and kinase cascades activation. Due to these properties, RA has proven anti-cancer capacity. Several breast cancer cells respond to the antiproliferative effects of RA, while others are RA-resistant. However, the overall signaling and transcriptional pathways that are altered in such cells have not been elucidated. Here, in a large-scale analysis of the phosphoproteins and in a genome-wide analysis of the RA-regulated genes, we compared two human breast cancer cell lines, a RA-responsive one, the MCF7 cell line, and a RA-resistant one, the BT474 cell line, which depicts several alterations of the “kinome”. Using high-resolution nano-LC-LTQ-Orbitrap mass spectrometry associated to phosphopeptide enrichment, we found that several proteins involved in signaling and in transcription, are differentially phosphorylated before and after RA addition. The paradigm of these proteins is the RA receptor α (RARα), which was phosphorylated in MCF7 cells but not in BT474 cells after RA addition. The panel of the RA-regulated genes was also different. Overall our results indicate that RA resistance might correlate with the deregulation of the phosphoproteome with consequences on gene expression.


Virchows Archiv | 2013

Retinoid acid receptor expression is helpful to distinguish between adenoma and well-differentiated carcinoma in the thyroid

Guillaume Gauchotte; Stéphanie Lacomme; Lydia Brochin; Benjamin Tournier; Virginie Cahn; Nathalie Monhoven; Françoise Piard; M. Klein; Nadine Martinet; Cécile Rochette-Egly; Jean-Michel Vignaud

Retinoid receptors (RRs) play a key role in cell proliferation and differentiation. We characterized the expression of RA receptors and retinoid X receptors (RARs and RXRs) in a series of 111 thyroid tumors and investigated the mechanisms responsible for their deregulation: hypermethylation of the RARB2 promoter, loss of heterozygosity (LOH) in the regions of RARB and RXRA, and altered expression of CRBP1 and enzymes involved in RA biosynthesis (RDH10 and RALDH2). Expression of RALDH2 and RDH10 was conserved in 100xa0% of adenomas and in 90 and 98xa0%, respectively, of carcinomas, whereas staining for CRBP1 was decreased in 9xa0% of FAs and 28xa0% of carcinomas, mainly anaplastic carcinomas (55xa0%). We found an abnormal expression of RARA, RARB, RXRA, and RXRB in 67, 69, 66, and 73xa0%, respectively, of thyroid carcinomas (nu2009=u200978) and in 9, 9, 9, and 33xa0% of follicular adenomas (nu2009=u200933) (pu2009<u20090.001). An abnormal staining pattern of at least two of these markers had 90xa0% sensitivity and 91xa0% specificity for a diagnosis of malignancy. Promoter hypermethylation of RARB2 was observed in some anaplastic carcinomas (14xa0%). LOH was found to be common at the RARB locus (3p24–3p25) and the RXRA locus (9q34), respectively, in 44 and 55xa0% of carcinomas and in 27 and 43xa0% of adenomas. In conclusion, immunohistochemical staining for RARs and RXRs may help in the differential diagnosis between well-differentiated carcinoma and follicular adenoma. Further investigation should be carried out to determine whether the characterization of RR expression might identify patients who could benefit from therapy with RA derivatives.

Collaboration


Dive into the Cécile Rochette-Egly's collaboration.

Top Co-Authors

Avatar

Régis Lutzing

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

Enrico Garattini

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Maurizio Gianni

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mineko Terao

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pierre Chambon

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge