10 research outputs found

    Fra-1 transcriptional mechanisms in Triple Negative Breast Cancer

    No full text
    Le complexe transcriptionnel AP-1 est une famille ubiquitaire de facteurs de transcription dimĂ©riques. Ses composants les mieux Ă©tudiĂ©s sont les membres des familles multigĂ©niques Fos et Jun. Les mĂ©canismes transcriptionnels gouvernĂ©s par ce complexe sont encore mal caractĂ©risĂ©s, en raison du grand nombre de dimĂšres AP-1 possibles, de l’abondance et de l’activitĂ© finement rĂ©gulĂ©es de ses constituants qui dĂ©pendent des contextes cellulaires et physiopathologiques. De plus, les limitations techniques ont longtemps donnĂ© l'impression que AP-1 rĂ©gule l’expression de ses gĂšnes cibles en se fixant principalement Ă  proximitĂ© de leurs promoteurs. Fra-1 est la protĂ©ine de la famille Fos la plus souvent impliquĂ©e dans les cancers Ă©pithĂ©liaux. En particulier, elle est surexprimĂ©e dans les cancers du sein triple nĂ©gatifs (TNBCs) oĂč elle contribue Ă  la tumorigenĂšse et Ă  l'agressivitĂ© tumorale par des effets plĂ©iotropes. Dans ce contexte, l’objectif de mes travaux de thĂšse Ă©tait d’aboutir Ă  une meilleure comprĂ©hension des actions transcriptionnelles de Fra-1 au niveau du gĂ©nome dans une lignĂ©e cellulaire TNBC de rĂ©fĂ©rence, la lignĂ©e MDA-MB-231. Pour ce faire, j'ai combinĂ© des donnĂ©es transcriptomiques avec des donnĂ©es de ChIP-seq et de NG-Capture C (technique Ă  haute rĂ©solution et Ă  haut dĂ©bit dĂ©rivĂ©e du 3C). J'ai Ă©galement inclus dans ces Ă©tudes le membre Fra-2, de la famille Fos, qui prĂ©sente la mĂȘme spĂ©cificitĂ© de fixation Ă  l’ADN et est Ă©galement exprimĂ© dans les TNBCs, bien qu'Ă  un niveau beaucoup plus bas, oĂč il contribue aussi au phĂ©notype tumoral. En accord avec leurs effets plĂ©iotropes, Fra-1 et Fra-2 activent ou rĂ©priment, soit individuellement soit de façon redondante ou complĂ©mentaire, l’expression de nombreux gĂšnes associĂ©s Ă  une large gamme de processus biologiques. Il est intĂ©ressant de noter que la rĂ©gulation des gĂšnes cibles est rarement due Ă  la liaison de Fra-1 et/ou Fra-2 au niveau des rĂ©gions promotrices de ces gĂšnes mais fait intervenir leur liaison sur des enhancers distaux. Mes rĂ©sultats de NG-Capture C suggĂšrent la prĂ©sence d’interactions chromatiniennes Ă  longue distance enhancer/promoteur, ainsi que des rĂ©seaux d’enhancers. Ces rĂ©seaux contiennent des enhancers liĂ©s par Fra-1 et d’autres indĂ©pendants de celui-ci. Aucune preuve d’un rĂŽle de Fra-1 dans le contrĂŽle des interactions chromatiniennes au niveau de ces rĂ©seaux n'a Ă©tĂ© trouvĂ©e en utilisant un panel de 35 gĂšnes rĂ©gulĂ©s par ce facteur. En parallĂšle, j'ai abordĂ© les mĂ©canismes de la rĂ©pression transcriptionnelle mĂ©diĂ©e par Fra-1, mĂ©canismes trĂšs rarement Ă©tudiĂ©s dans la littĂ©rature, en utilisant deux gĂšnes modĂšles, TGFB2 et SMAD6. Ces Ă©tudes ont mis en Ă©vidence des mĂ©canismes diffĂ©rents mis en jeu par Fra-1 pour la rĂ©pression de ces deux gĂšnes, ce qui montre la complexitĂ© des mĂ©canismes de la rĂ©gulation transcriptionnelle mĂ©diĂ©e par Fra-1.The AP-1 transcription complex is a ubiquitous family of dimeric transcription factors. Its best-studied components are the members of the Fos and Jun multigene families. The mechanisms whereby AP-1 exerts its transcriptional actions are still ill-understood due to the wide number of possible AP-1 dimers and the exquisitely regulated abundance and activity of its constituents, that all depend on the cell types and physiopathological contexts. Moreover, technical limitations have long given the impression that AP-1 mostly operates in the vicinity of gene promoters. Fra-1 is the Fos family protein that is most often implicated in epithelial cancers. In particular, it is overexpressed in triple negative breast cancers (TNBCs) where it contributes to tumorigenesis and tumor aggressiveness through pleiotropic effects. Based on this, the aim of my thesis was to gain a more comprehensive view of Fra-1 transcriptional actions at the genome-wide level in the MDA-MB-231 reference TNBC cell line, . To this aim, I have combined transcriptomic data with ChIP-seq and NG-Capture C (high resolution, high throughput 3C-derived technique) data. I have also included in my studies its Fos family kin Fra-2, as it displays the same DNA binding specificity and is also expressed in TNBCs, albeit at a much lower level, where it also contributes to the tumor phenotype. Consistently with their pleiotropic effects, Fra-1 and Fra-2 were found to up- or down-regulate either individually, together or redundantly many genes associated with a wide range of biological processes. Interestingly, the regulation of target genes is rarely due to Fra-1 and Fra-2 binding at gene promoters, but involves their binding to distal enhancers. My NG-Capture C results imply the presence of long-range chromatin interactions in Fra-1 modes of action, as well as enhancer hubs containing Fra-1- and non-Fra-1-binding enhancers. No evidence for a role for Fra-1 in the control of chromatin looping was however found using a panel of 35 Fra-1-regulated genes. Moreover, I have addressed the mechanisms of transcriptional repression mediated by Fra-1, as these have practically never been studied, using two model genes, TGFB2 and SMAD6. These studies underlined different mechanisms employed by Fra-1 for the repression of these genes, embodying the complexity of Fra-1 transcriptional regulation mechanisms

    Multiple Fra-1-bound enhancers showing different molecular and functional features can cooperate to repress gene transcription

    No full text
    Abstract Background How transcription factors (TFs) down-regulate gene expression remains ill-understood, especially when they bind to multiple enhancers contacting the same gene promoter. In particular, it is not known whether they exert similar or significantly different molecular effects at these enhancers. Results To address this issue, we used a particularly well-suited study model consisting of the down-regulation of the TGFB2 gene by the TF Fra-1 in Fra-1-overexpressing cancer cells, as Fra-1 binds to multiple enhancers interacting with the TGFB2 promoter. We show that Fra-1 does not repress TGFB2 transcription via reducing RNA Pol II recruitment at the gene promoter but by decreasing the formation of its transcription-initiating form. This is associated with complex long-range chromatin interactions implicating multiple molecularly and functionally heterogeneous Fra-1-bound transcriptional enhancers distal to the TGFB2 transcriptional start site. In particular, the latter display differential requirements upon the presence and the activity of the lysine acetyltransferase p300/CBP. Furthermore, the final transcriptional output of the TGFB2 gene seems to depend on a balance between the positive and negative effects of Fra-1 at these enhancers. Conclusion Our work unveils complex molecular mechanisms underlying the repressive actions of Fra-1 on TGFB2 gene expression. This has consequences for our general understanding of the functioning of the ubiquitous transcriptional complex AP-1, of which Fra-1 is the most documented component for prooncogenic activities. In addition, it raises the general question of the heterogeneity of the molecular functions of TFs binding to different enhancers regulating the same gene

    YTHDC1 regulates distinct post-integration steps of HIV-1 replication and is important for viral infectivity

    No full text
    International audienceBackground The recent discovery of the role of m 6 A methylation in the regulation of HIV-1 replication unveiled a novel layer of regulation for HIV gene expression. This epitranscriptomic modification of HIV-1 RNAs is under the dynamic control of specific writers and erasers. In addition, cytoplasmic readers of the m 6 A mark are recruited to the modified viral RNAs and regulate HIV-1 replication. Yet, little is known about the effects of m 6 A writers and readers on the biogenesis of HIV-1 RNAs. Results We showed that the METTL3/14 m 6 A methyltransferase complex and the m 6 A YTHDF2 cytoplasmic writer down regulates the abundance of HIV-1 RNAs in infected cells. We also identified the m 6 A nuclear writer YTHDC1 as a novel regulator of HIV-1 transcripts. In HIV-1 producer cells, we showed that knocking down YTHDC1 increases the levels of unspliced and incompletely spliced HIV-1 RNAs, while levels of multiply spliced transcripts remained unaffected. In addition, we observed that depletion of YTHDC1 has no effect on the nuclear cytoplasmic distribution of viral transcripts. YTHDC1 binds specifically to HIV-1 transcripts in a METTL3-dependent manner. Knocking down YTHDC1 reduces the expression of Env and Vpu viral proteins in producer cells and leads to the incorporation of unprocessed Env gp160 in virus particles, resulting in the decrease of their infectivity. Conclusions Our findings indicate that, by controlling HIV-1 RNA biogenesis and protein expression, the m 6 A nuclear reader YTHDC1 is required for efficient production of infectious viral particles. Graphical Abstrac

    The histone variant macroH2A1.1 regulates RNA polymerase II-paused genes within defined chromatin interaction landscapes

    No full text
    International audienceThe histone variant macroH2A1.1 plays a role in cancer development and metastasis. To determine the underlying molecular mechanisms, we mapped the genome-wide localization of endogenous macroH2A1.1 in the human breast cancer cell line MDA-MB-231. We demonstrate that macroH2A1.1 specifically binds to active promoters and enhancers in addition to facultative heterochromatin. Selective knock down of macroH2A1.1 deregulates the expression of hundreds of highly active genes. Depending on the chromatin landscape, macroH2A1.1 acts through two distinct molecular mechanisms. The first mitigates excessive transcription by binding over domains including the promoter and the gene body. The second stimulates expression of RNA polymerase II (Pol II)-paused genes, including genes regulating mammary tumor cell migration. In contrast to the first mechanism, macroH2A1.1 specifically associates with the transcription start site of Pol II-paused genes. These processes occur in a predefined local 3D genome landscape, but do not require rewiring of enhancer-promoter contacts. We thus propose that macroH2A1.1 serves as a transcriptional modulator with a potential role in assisting the conversion of promoter-locked Pol II into a productive, elongating Pol II

    TASOR epigenetic repressor cooperates with a CNOT1 RNA degradation pathway to repress HIV

    No full text
    International audienceThe Human Silencing Hub (HUSH) complex constituted of TASOR, MPP8 and Periphilin recruits the histone methyl-transferase SETDB1 to spread H3K9me3 repressive marks across genes and transgenes in an integration site-dependent manner. The deposition of these repressive marks leads to heterochromatin formation and inhibits gene expression, but the underlying mechanism is not fully understood. Here, we show that TASOR silencing or HIV-2 Vpx expression, which induces TASOR degradation, increases the accumulation of transcripts derived from the HIV-1 LTR promoter at a post-transcriptional level. Furthermore, using a yeast 2-hybrid screen, we identify new TASOR partners involved in RNA metabolism including the RNA deadenylase CCR4-NOT complex scaffold CNOT1. TASOR and CNOT1 synergistically repress HIV expression from its LTR. Similar to the RNA-induced transcriptional silencing complex found in fission yeast, we show that TASOR interacts with the RNA exosome and RNA Polymerase II, predominantly under its elongating state. Finally, we show that TASOR facilitates the association of RNA degradation proteins with RNA polymerase II and is detected at transcriptional centers. Altogether, we propose that HUSH operates at the transcriptional and post-transcriptional levels to repress HIV proviral expression

    The PDK1 Inhibitor Dichloroacetate Controls Cholesterol Homeostasis Through the ERK5/MEF2 Pathway

    Get PDF
    Abstract Controlling cholesterol levels is a major challenge in human health, since hypercholesterolemia can lead to serious cardiovascular disease. Drugs that target carbohydrate metabolism can also modify lipid metabolism and hence cholesterol plasma levels. In this sense, dichloroacetate (DCA), a pyruvate dehydrogenase kinase (PDK) inhibitor, augments usage of the glycolysis-produced pyruvate in the mitochondria increasing oxidative phosphorylation (OXPHOS). In several animal models, DCA decreases plasma cholesterol and triglycerides. Thus, DCA was used in the 70 s to treat diabetes mellitus, hyperlipoproteinemia and hypercholesterolemia with satisfactory results. However, the mechanism of action remained unknown and we describe it here. DCA increases LDLR mRNA and protein levels as well as LDL intake in several cell lines, primary human hepatocytes and two different mouse models. This effect is mediated by transcriptional activation as evidenced by H3 acetylation on lysine 27 on the LDLR promoter. DCA induces expression of the MAPK ERK5 that turns on the transcription factor MEF2. Inhibition of this ERK5/MEF2 pathway by genetic or pharmacological means decreases LDLR expression and LDL intake. In summary, our results indicate that DCA, by inducing OXPHOS, promotes ERK5/MEF2 activation leading to LDLR expression. The ERK5/MEF2 pathway offers an interesting pharmacological target for drug development

    The PDK1 Inhibitor Dichloroacetate Controls Cholesterol Homeostasis Through the ERK5/MEF2 Pathway

    No full text
    Controlling cholesterol levels is a major challenge in human health, since hypercholesterolemia can lead to serious cardiovascular disease. Drugs that target carbohydrate metabolism can also modify lipid metabolism and hence cholesterol plasma levels. In this sense, dichloroacetate (DCA), a pyruvate dehydrogenase kinase (PDK) inhibitor, augments usage of the glycolysis-produced pyruvate in the mitochondria increasing oxidative phosphorylation (OXPHOS). In several animal models, DCA decreases plasma cholesterol and triglycerides. Thus, DCA was used in the 70 s to treat diabetes mellitus, hyperlipoproteinemia and hypercholesterolemia with satisfactory results. However, the mechanism of action remained unknown and we describe it here. DCA increases LDLR mRNA and protein levels as well as LDL intake in several cell lines, primary human hepatocytes and two different mouse models. This effect is mediated by transcriptional activation as evidenced by H3 acetylation on lysine 27 on the LDLR promoter. DCA induces expression of the MAPK ERK5 that turns on the transcription factor MEF2. Inhibition of this ERK5/MEF2 pathway by genetic or pharmacological means decreases LDLR expression and LDL intake. In summary, our results indicate that DCA, by inducing OXPHOS, promotes ERK5/MEF2 activation leading to LDLR expression. The ERK5/MEF2 pathway offers an interesting pharmacological target for drug development
    corecore