10 research outputs found

    Role of E4F1 in the regulation of cancer cell homeostasis

    No full text
    E4F1 est un facteur de transcription liant l'ADN, exprimé de façon ubiquitaire par tous les tissus, et qui possède une activité E3 ubiquitine ligase atypique dirigée contre le suppresseur de tumeur p53. La protéine E4F1 interagit directement avec plusieurs suppresseurs de tumeurs cellulaires et des oncogènes viraux (p53, pRb, DRAL, RASSF1A, p19ARF, BMI1, HBX et GAM1...), suggérant qu’elle est elle-même impliquée dans la tumorigenèse. La perte d’E4F1 dans des fibroblastes embryonnaires (Mefs) transformés ou dans des cellules de sarcomes histiocytaires déficientes pour la voie p53, entraine la mort de ces cellules. La même inactivation d'E4F1 dans les cellules normales n'affecte pas leur survie mais entraine un arrêt de prolifération. Des analyses transcriptomiques et de liaison à l'ADN à l'échelle du génome entier (ChIP-seq et analyses différentielles des transcriptomes de cellules E4F1 WT et KO) nous ont permis d’identifier une centaine de gènes liés et régulés directement par E4F1. Ces gènes codent notamment pour des protéines mitochondriales impliquées dans le métabolisme et l'homéostasie de cette organelle, dont plusieurs composants et régulateurs de l'enzyme multimérique, pyruvate déshydrogénase. Un second groupe de gênes cibles d’E4F1 est impliqué dans la réponse aux dommages à l'ADN, dont le gène codant pour la kinase CHK1 qui joue un rôle essentiel dans le contrôle de la stabilité du génome. En accord avec la fonction de ces gènes cibles, la perte d’E4F1 entraine des perturbations du métabolisme cellulaire et des checkpoints de réponse aux stress génotoxiques. Dans les cellules déficientes pour la voie p53, ces perturbations conduisent à des stress oxydatifs (surproduction de ROS mitochondriaux) et énergétiques, suivis de dommages aux protéines et à l'ADN, et in fine, à une mort cellulaire massive. Dans les cellules compétentes pour la voie p53 ces altérations sont fortement atténuées et conduisent à un arrêt de la prolifération. Une partie des effets protecteurs de p53 est due à sa capacité à stimuler l'expression du gène ALDH4a1 qui code pour une aldehyde dehydrogenase impliquée dans le catabolisme de la proline et dont l'activité possède des propriétés anti-oxydantes.Mes travaux mettent également en évidence que le niveau d'expression de la protéine E4F1 et son activité augmentent lors de la transformation cellulaire ainsi qu'en réponse à des stress énergétiques, génotoxiques ou oxydatifs. Dans ces trois dernières conditions, la phosphorylation d'E4F1 est également augmentée au niveau de plusieurs sérines qui ont été identifiées par spectrométrie de masse. En résumé, tous ces éléments indiquent qu'E4F1 est un acteur important du contrôle de l'homéostasie métabolique et de la réponse aux stress, particulièrement essentiel pour la survie des cellules cancéreuses déficientes pour la voie p53. Mes observations suggèrent également qu'E4F1 est activé en réponse à différents stress et qu'il pourrait jouer un rôle essentiel dans la capacité des cellules cancéreuses à s'adapter aux multiples stress environnementaux auxquels elles sont exposées au cours de la tumorigenèse.The ubiquitously expressed E4F1 protein acts as a transcription factor that binds a consensus DNA sequence at promoters, and as an atypical E3-ligase for the tumor suppressor p53. E4F1 physically interacts with several bona fide cellular tumor suppressors and viral oncoproteins (including p53, pRb, DRAL, RASSF1A, p19ARF, BMI1, HBX and GAM1...), suggesting that it might itself be involved in tumorigenesis. E4F1 genetic inactivation in transformed mouse embryo fibroblasts (Mefs) and in hematopoietic tumors deficient for the p53 pathway, results in massive cell death. Importantly, inactivation of E4F1 in normal cells does not affect cell survival. Genome wide approaches (ChIP-seq profiling and comparative transcriptomics performed on E4F1 WT and KO cells) identified a limited list (100) of genes that are bound and directly regulated by E4F1. Several E4F1 target genes code for mitochondrial proteins involved in mitochondria metabolism and homeostasis, including several components of the pyruvate dehydrogenase complex. Another set of E4F1 target genes codes for factors involved in DNA repair and damage checkpoints, including the checkpoint kinase CHK1. Accordingly, both mitochondrial and checkpoint functions are altered in E4F1 KO cells. In proliferating cells deficient for the p53 pathway, these defects lead to energetic and oxidative stresses, protein and DNA damages, and in fine, massive cell death. In p53-proficient cells, these alterations are attenuated and lead to growth arrest. Part of this protective effect of p53 is mediated by ALDH4a1, a p53 target gene encoding an aldehyde dehydrogenase involved in proline catabolism and that exhibits antioxidant properties.In this thesis, I also demonstrate that E4F1 protein level and activity is increased during cell transformation and upon exposure to genotoxic, energetic or oxidative stresses. These stresses also lead to E4F1 phosphorylation at specific serine residues that were identified by mass spectrometry. All together this work shows that E4F1 controls cellular functions that are important for mammalian cells metabolic homeostasis and stress responses, and that are essential for the survival of p53-deficient cancer cells. This work also suggests that E4F1 is activated in response to various stresses and therefore, that it could play an essential role in allowing cancer cells to adapt to environmental stresses

    Description of an optimized ChIP-seq analysis pipeline dedicated to genome wide identification of E4F1 binding sites in primary and transformed MEFs

    No full text
    International audienceThis Data in Brief report describes the experimental and bioinformatic procedures that we used to analyze and interpret E4F1 ChIP-seq experiments published in Rodier et al. (2015) [10]. Raw and processed data are available at the GEO DataSet repository under the subseries # GSE57228. E4F1 is a ubiquitously expressed zinc-finger protein of the GLI-Kruppel family that was first identified in the late eighties as a cellular transcription factor targeted by the adenoviral oncoprotein E1A13S (Ad type V) and required for the transcription of adenoviral genes (Raychaudhuri et al., 1987) [8]. It is a multifunctional factor that also acts as an atypical E3 ubiquitin ligase for p53 (Le Cam et al., 2006) [2]. Using KO mouse models we then demonstrated that E4F1 is essential for early embryonic development (Le Cam et al., 2004), for proliferation of mouse embryonic cell (Rodier et al., 2015), for the maintenance of epidermal stem cells (Lacroix et al., 2010) [6], and strikingly, for the survival of cancer cells (Hatchi et al., 2007) [4]; (Rodier et al., 2015) [10]. The latter survival phenotype was p53-independent and suggested that E4F1 was controlling a transcriptional program driving essential functions in cancer cells. To identify this program, we performed E4F1 ChIP-seq analyses in primary Mouse Embryonic Fibroblasts (MEF) and in p53(-/-), H-Ras(V12)-transformed MEFs. The program directly controlled by E4F1 was obtained by intersecting the lists of E4F1 genomic targets with the lists of genes differentially expressed in E4F1 KO and E4F1 WT cells (Rodier et al., 2015). We describe hereby how we improved our ChIP-seq analyses workflow by applying prefilters on raw data and by using a combination of two publicly available programs, Cisgenome and QESEQ

    Functional and Structural Insights into ASB2α, a Novel Regulator of Integrin-dependent Adhesion of Hematopoietic Cells

    No full text
    International audienceBy providing contacts between hematopoietic cells and the bone marrow microenvironment, integrins are implicated in cell adhesion and thereby in control of cell fate of normal and leukemia cells. The ASB2 gene, initially identified as a retinoic acid responsive gene and a target of the promyelocytic leukemia retinoic acid receptor α oncoprotein in acute promyelocytic leukemia cells, encodes two isoforms, a hematopoietic-type (ASB2α) and a muscle-type (ASB2β) that are involved in hematopoietic and myogenic differentiation, respectively. ASB2α is the specificity subunit of an E3 ubiquitin ligase complex that targets filamins to proteasomal degradation. To examine the relationship of the ASB2α structure to E3 ubiquitin ligase function, functional assays and molecular modeling were performed. We show that ASB2α, through filamin A degradation, enhances adhesion of hematopoietic cells to fibronectin, the main ligand of β1 integrins. Furthermore, we demonstrate that a short N-terminal region specific to ASB2α, together with ankyrin repeats 1 to 10, is necessary for association of ASB2α with filamin A. Importantly, the ASB2α N-terminal region comprises a 9-residue segment with predicted structural homology to the filamin-binding motifs of migfilin and β integrins. Together, these data provide new insights into the molecular mechanisms of ASB2α binding to filamin

    A Chemoproteomic Approach Reveals Massive Reprogramming of the Epithelial Cell Surface during Oncogenic KRAS-mediated Transformation.

    No full text
    Background KRAS is frequently mutated in human cancers, including ~45% of colorectal adenocarcinoma (CRC). The most promising therapeutic strategy for advanced CRC is the use of monoclonal antibodies (cetuximab, panitumumab) that block activation of the epidermal growth factor receptor (EGFR). However, activating mutations in KRAS were shown to be common drivers of acquired resistance, and recent retrospective studies have shown that they negatively predict responsiveness to anti-EGFR therapy. Despite continuous efforts, oncogenic KRAS is still deemed “undruggable”, warranting the need for alternative therapeutic approaches. While oncogenic KRAS is described to regulate many intracellular signaling events that are currently being evaluated as potential therapeutic targets, much less is known about its potential impact on the cell surface. Elucidating how oncogenic KRAS modifies the cell surface proteome (surfaceome) could help understand its complex mechanism of action, and possibly identify new “druggable” targets and/or tumor-specific biomarkers. Material and methods Herein, we have optimized a cutting-edge chemoproteomic approach based on the labeling of cell surface proteins with biotin reagents, their subsequent purification with avidin chromatography, and quantification using label-free quantitative proteomics with liquid chromatography-tandem mass spectrometry (LC-MS/MS). Results Using an intestinal crypt epithelial cell model that reflects KRAS-induced malignant transformation, our LC-MS/MS analyses allowed the identification of over 350 cell surface molecules from which 13% and 22% were significantly upregulated and downregulated in KRAS-transformed cells, respectively. Thus, we found that oncogenic KRAS modulates the surface expression of a large network of proteins, including cell adhesion molecules, receptor tyrosine kinases, G protein-coupled receptors, ion channels, transporters, and peptidases/proteinases. Interestingly, while many of these changes are associated with a KRAS-dependent gene expression signature, we also identified numerous surface proteins that appear to be regulated in a transcription-independent manner. Conclusion Taken together, these results indicate that oncogenic KRAS leads to a massive reprogramming of the epithelial cell surface, and suggest multiple cell surface proteins as molecular targets or diagnostic markers for KRAS-dependent cancers

    Multi-Level Control of the ATM/ATR-CHK1 Axis by the Transcription Factor E4F1 in Triple-Negative Breast Cancer

    No full text
    International audienceE4F1 is essential for early embryonic mouse development and for controlling the balance between proliferation and survival of actively dividing cells. We previously reported that E4F1 is essential for the survival of murine p53-deficient cancer cells by controlling the expression of genes involved in mitochondria functions and metabolism, and in cell-cycle checkpoints, including CHEK1, a major component of the DNA damage and replication stress responses. Here, combining ChIP-Seq and RNA-Seq approaches, we identified the transcriptional program directly controlled by E4F1 in Human Triple-Negative Breast Cancer cells (TNBC). E4F1 binds and regulates a limited list of direct target genes (57 genes) in these cells, including the human CHEK1 gene and, surprisingly, also two other genes encoding post-transcriptional regulators of the ATM/ATR-CHK1 axis, namely, the TTT complex component TTI2 and the phosphatase PPP5C, that are essential for the folding and stability, and the signaling of ATM/ATR kinases, respectively. Importantly, E4F1 also binds the promoter of these genes in vivo in Primary Derived Xenograft (PDX) of human TNBC. Consequently, the protein levels and signaling of CHK1 but also of ATM/ATR kinases are strongly downregulated in E4F1-depleted TNBC cells resulting in a deficiency of the DNA damage and replicative stress response in these cells. The E4F1-depleted cells fail to arrest into S-phase upon treatment with the replication-stalling agent Gemcitabine, and are highly sensitized to this drug, as well as to other DNA-damaging agents, such as Cisplatin. Altogether, our data indicate that in breast cancer cells the ATM/ATR-CHK1 signaling pathway and DNA damage-stress response are tightly controlled at the transcriptional and post-transcriptional level by E4F1

    E4F1 controls a transcriptional program essential for pyruvate dehydrogenase activity

    No full text
    The mitochondrial pyruvate dehydrogenase (PDH) complex (PDC) acts as a central metabolic node that mediates pyruvate oxidation and fuels the tricarboxylic acid cycle to meet energy demand. Here, we reveal another level of regulation of the pyruvate oxidation pathway in mammals implicating the E4 transcription factor 1 (E4F1). E4F1 controls a set of four genes [dihydrolipoamide acetlytransferase (Dlat), dihydrolipoyl dehydrogenase (Dld), mitochondrial pyruvate carrier 1 (Mpc1), and solute carrier family 25 member 19 (Slc25a19)] involved in pyruvate oxidation and reported to be individually mutated in human metabolic syndromes. E4F1 dysfunction results in 80% decrease of PDH activity and alterations of pyruvate metabolism. Genetic inactivation of murine E4f1 in striated muscles results in viable animals that show low muscle PDH activity, severe endurance defects, and chronic lactic acidemia, recapitulating some clinical symptoms described in PDC-deficient patients. These phenotypes were attenuated by pharmacological stimulation of PDH or by a ketogenic diet, two treatments used for PDH deficiencies. Taken together, these data identify E4F1 as a master regulator of the PDC
    corecore