8 research outputs found

    The epigenetic regulator RINF (CXXC5) maintains SMAD7 expression in human immature erythroid cells and sustains red blood cells expansion

    Full text link
    The gene CXXC5, encoding a Retinoid-Inducible Nuclear Factor (RINF), is located within a region at 5q31.2 commonly deleted in myelodysplastic syndrome (MDS) and adult acute myeloid leukemia (AML). RINF may act as an epigenetic regulator and has been proposed as a tumor suppressor in hematopoietic malignancies. However, functional studies in normal hematopoiesis are lacking, and its mechanism of action is unknow. Here, we evaluated the consequences of RINF silencing on cytokineinduced erythroid differentiation of human primary CD34+ progenitors. We found that RINF is expressed in immature erythroid cells and that RINF-knockdown accelerated erythropoietin-driven maturation, leading to a significant reduction (~45%) in the number of red blood cells (RBCs), without affecting cell viability. The phenotype induced by RINF-silencing was TGFβ-dependent and mediated by SMAD7, a TGFβ- signaling inhibitor. RINF upregulates SMAD7 expression by direct binding to its promoter and we found a close correlation between RINF and SMAD7 mRNA levels both in CD34+ cells isolated from bone marrow of healthy donors and MDS patients with del(5q). Importantly, RINF knockdown attenuated SMAD7 expression in primary cells and ectopic SMAD7 expression was sufficient to prevent the RINF knockdowndependent erythroid phenotype. Finally, RINF silencing affects 5’-hydroxymethylation of human erythroblasts, in agreement with its recently described role as a Tet2- anchoring platform in mouse. Altogether, our data bring insight into how the epigenetic factor RINF, as a transcriptional regulator of SMAD7, may fine-tune cell sensitivity to TGFβ superfamily cytokines and thus play an important role in both normal and pathological erythropoiesis

    Functional Characterization of the Nuclear Factor RINF During Normal and Tumoral Hematopoiesis

    No full text
    L’hématopoïèse regroupe l'ensemble des mécanismes qui assurent le renouvellement continu et régulé des cellules sanguines, à partir des cellules souches hématopoïétiques. La différenciation des cellules souches en cellules matures est un phénomène finement orchestré par divers signaux (facteurs de croissance, hormones, cytokines et microenvironnement médullaire) capables de stimuler la prolifération de cellules quiescentes ainsi que leur engagement dans diverses voies de la maturation hématopoïétique. Ces processus sont généralement régulés via l’activation de facteurs de transcription, mais également par des mécanismes épigénétiques. Le gène RINF/CXXC5 a initialement été décrit comme essentiel pour les processus de différenciation granulocytaire normale. Toutefois, son implication dans d’autres voies de l’hématopoïèse n’avait pas été étudiée. Afin de définir son rôle dans la différenciation et l’engagement vers des lignages hématopoïétiques autres que la voie granulocytaire, notamment érythroïde et monocytaire, sa contribution fonctionnelle dans des lignées hématopoïétiques et dans des cellules primaires CD34+ de moelle osseuse (donneurs sains) a été étudiée par une approche de perte ou gain de fonction. Ces expériences, dans des modèles de la voie granulocytaire (lignée NB4 et HL60 traitées par les rétinoïdes) et de la voie érythroïde (lignées K562 et UT7/GM traitées par l’hémine ou l’EPO) démontrent l’implication de RINF dans la maturation terminale de ces deux lignages. Ces données ont ensuite été validées dans des progéniteurs hématopoïétiques (tests clonogéniques) où l’expression de RINF favorise la différenciation granuleuse et interfère négativement avec la différenciation érythroïde, sans impacter la voie monocytaire. En effet, l’extinction de son expression diminue le nombre des colonies granuleuses et augmente le nombre de colonies érythroïdes. Des dérégulations au niveau de l’expression des facteurs ou co-facteurs de trancription qui régulent l’hématopoïèse peuvent aboutir à des hémopathies, telles que les Leucémies Aiguës Myéloïdes. Ces pathologies sont la résultante de l’association d’une augmentation de la prolifération cellulaire et d’un blocage des processus de différenciation. Au vu de son rôle important au cours de la granulopoïèse et de l’érythropoïèse, l’hypothèse que des dérégulations de RINF pourraient intervenir dans les processus de leucémogenèse a été testée par l’étude de son niveau d’expression dans les différentes Leucémies Aiguës Myéloïdes. Ainsi, il a été démontré que parmi les patients dont le niveau d’expression de RINF est le plus élevé, le pronostic vital est mauvais, associé à une résistance aux traitements chimiothérapeutiques. L’ensemble de ces données a permis d’aboutir à la conclusion que des dérégulations de l’expression de RINF pourraient contribuer au processus de leucémogenèse, et qu’ainsi RINF pourrait être une potentielle cible thérapeutique dans le cadre des LAM. D’un point de vue moléculaire, le mode d’action de la protéine RINF reste à ce jour du domaine de l’hypothèse, mais la présence d’un doigt de zinc de type CXXC lui permettrait d’intervenir dans des mécanismes des régulations épigénétiques, tels que la méthylation de l’ADN. Une meilleure compréhension des mécanismes régulés par la protéine pourrait permettre à terme une meilleure compréhension des régulations de l’hématopoïèse, voire des processus de leucémogenèse.During hematopoiesis, hematopoietic stem cells (HSC) differentiation is orchestred by different signals, able to stimulate cell proliferation of quiescent cells, and their commitment in the different hematopoietic lineages. These process are regulated by transcription factors activation, as well by epigenetic mecanisms. By a microarray approach, we have identified a novel retinoid-responsive gene (CXXC5) encoding a Retinoid-Inducible Nuclear Factor (RINF) that plays an essential role during in vitro human hematopoiesis. Indeed, expression studies and gene silencing experiments both demonstrate RINF requirement during in vitro terminal differentiation of myeloid leukemia cells (NB4, HL60), but also during normal myelopoiesis of bone marrow progenitors (CD34+ HSPC cells in presence of cytokines). In the present study, we demonstrate that in cell lines, RINF overexpression provokes an earlier myeloid differentiation under retinoids treatement and slow-downs erythroid maturation induced by hemin whereas its down-regulation accelerates erythroid terminal differentiation. In normal CD34+ HSCP, we demonstrated that RINF down regulation (1) promotes differentiation in erythroid lineage at the expense of granulocyte lineage, and (2) accelerates terminal erythroid differentiation. Overexpression, contribute to promote myeloid pathway even though cells are in erythroid conditions. Because of its role during hematopoiesis regulation and its gene localization in 5q31.2, we investigated CXXC5/RINF expression in primary human acute myeloid leukemia (AML) cells derived from 594 patients. A wide variation in CXXC5/RINF mRNA levels was observed in the immature leukemic myeloblasts. Furthermore, patients with low-risk cytogenetic abnormalities showed significantly lower levels compared to patients with high-risk abnormalities, and high RINF/CXXC5/ mRNA levels were associated with decreased overall survival for patients receiving intensive chemotherapy for newly diagnosed AML. CXXC5/RINF knockdown in AML cell lines caused increased susceptibility to chemotherapy-induced apoptosis, and regulation of apoptosis also seemed to differ between primary human AML cells with high and low RINF expression. The association with adverse prognosis together with the antiapoptotic effect of CXXC5/RINF suggests that targeting of CXXC5/RINF should be considered as a possible therapeutic strategy, especially in high-risk patients who show increased expression in AML cells compared with normal hematopoietic cells

    Proximity Profiling of the CFTR Interaction Landscape in Response to Orkambi

    No full text
    Deletion of phenylalanine 508 (∆F508) of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) anion channel protein is the leading cause of Cystic Fibrosis (CF). Here, we report the analysis of CFTR and ∆F508-CFTR interactomes using BioID (proximity-dependent biotin identification), a technique that can also detect transient associations. We identified 474 high-confidence CFTR proximity-interactors, 57 of which have been previously validated, with the remainder representing novel interaction space. The ∆F508 interactome, comprising 626 proximity-interactors was markedly different from its wild type counterpart, with numerous alterations in protein associations categorized in membrane trafficking and cellular stress functions. Furthermore, analysis of the ∆F508 interactome in cells treated with Orkambi identified several interactions that were altered as a result of this drug therapy. We examined two candidate CFTR proximity interactors, VAPB and NOS1AP, in functional assays designed to assess surface delivery and overall chloride efflux. VAPB depletion impacted both CFTR surface delivery and chloride efflux, whereas NOS1AP depletion only affected the latter. The wild type and ∆F508-CFTR interactomes represent rich datasets that could be further mined to reveal additional candidates for the functional rescue of ∆F508-CFTR

    Expression of the potential therapeutic target CXXC5 in primary acute myeloid leukemia cells -high expression is associated with adverse prognosis as well as altered intracellular signaling and transcriptional regulation

    Get PDF
    International audienceThe CXXC5 gene encodes a transcriptional activator with a zinc-finger domain, and high expression in human acute myeloid leukemia (AML) cells is associated with adverse prognosis. We now characterized the biological context of CXXC5 expression in primary human AML cells. The global gene expression profile of AML cells derived from 48 consecutive patients was analyzed; cells with high and low CXXC5 expression then showed major differences with regard to extracellular communication and intracellular signaling. We observed significant differences in the phosphorylation status of several intracellular signaling mediators (CREB, PDK1, SRC, STAT1, p38, STAT3, rpS6) that are important for PI3K-Akt-mTOR signaling and/or transcriptional regulation. High CXXC5 expression was also associated with high mRNA expression of several stem cell-associated transcriptional regulators, the strongest associations being with WT1, GATA2, RUNX1, LYL1, DNMT3, SPI1, and MYB. Finally, CXXC5 knockdown in human AML cell lines caused significantly increased expression of the potential tumor suppressor gene TSC22 and genes encoding the growth factor receptor KIT, the cytokine Angiopoietin 1 and the selenium-containing glycoprotein Selenoprotein P. Thus, high CXXC5 expression seems to affect several steps in human leukemogenesis, including intracellular events as well as extracellular communication. INTRODUCTION CXXC5 is a retinoid-responsive gene localized to the 5q31.3 chromosomal region [1] and encoding a retinoid-inducible nuclear factor (RINF) [2] that is a protein containing a CXXC-type zinc-finger domain and acting as a transcription regulator [3]. Expression studies as well as gene silencing experiments suggest that CXXC5 is important in normal myelopoiesis [2] and for differentiation of endothelial cells [3]. Furthermore, we recently described that CXXC5 is expressed in primary acute myeloid leukemia (AML) cells; this expression shows a wide variation between patients and high levels are associated with an adverse prognosis and resistance to chemotherapy-induced apoptosis [4]. Another study recently confirmed our observations and CXXC5 expression was then of independent prognostic significance in multivariate analyses after adjustmen

    ARID1a Associates with Lymphoid-Restricted Transcription Factors and Has an Essential Role in T Cell Development

    No full text
    Maturation of lymphoid cells is controlled by the action of stage and lineage-restricted transcription factors working in concert with the general transcription and chromatin remodeling machinery to regulate gene expression. To better understand this functional interplay, we used Biotin Identification in human embryonic kidney cells to identify proximity interaction partners for GATA3, TCF7 (TCF1), SPI1, HLF, IKZF1, PAX5, ID1, and ID2. The proximity interaction partners shared among the lineage-restricted transcription factors included ARID1a, a BRG1-associated factor complex component. CUT&RUN analysis revealed that ARID1a shared binding with TCF7 and GATA3 at a substantial number of putative regulatory elements in mouse T cell progenitors. In support of an important function for ARID1a in lymphocyte development, deletion of Arid1a in early lymphoid progenitors in mice resulted in a pronounced developmental arrest in early T cell development with a reduction of CD4+CD8+ cells and a 20-fold reduction in thymic cellularity. Exploring gene expression patterns in DN3 cells from Wt and Arid1a-deficient mice suggested that the developmental block resided in the DN3a to DN3b transition, indicating a deficiency in β-selection. Our work highlights the critical importance of functional interactions between stage and lineage-restricted factors and the basic transcription machinery during lymphocyte differentiation

    A SARS-CoV-2 – host proximity interactome

    No full text
    Posté sur BioRxiv le 4 septembre 2020 https://www.biorxiv.org/content/10.1101/2020.09.03.282103v1.article-infoViral replication is dependent on interactions between viral polypeptides and host proteins. Identifying virus-host protein interactions can thus uncover unique opportunities for interfering with the virus life cycle via novel drug compounds or drug repurposing. Importantly, many viral-host protein interactions take place at intracellular membranes and poorly soluble organelles, which are difficult to profile using classical biochemical purification approaches. Applying proximity-dependent biotinylation (BioID) with the fast-acting miniTurbo enzyme to 27 SARS-CoV-2 proteins in a lung adenocarcinoma cell line (A549), we detected 7810 proximity interactions (7382 of which are new for SARS-CoV-2) with 2242 host proteins (results available at covid19interactome.org). These results complement and dramatically expand upon recent affinity purification-based studies identifying stable host-virus protein complexes, and offer an unparalleled view of membrane-associated processes critical for viral production. Host cell organellar markers were also subjected to BioID in parallel, allowing us to propose modes of action for several viral proteins in the context of host proteome remodelling. In summary, our dataset identifies numerous high confidence proximity partners for SARS-CoV-2 viral proteins, and describes potential mechanisms for their effects on specific host cell functions

    The epigenetic regulator RINF (CXXC5) maintains <I>SMAD7</I> expression in human immature erythroid cells and sustains red blood cell expansion

    No full text
    International audienceThe gene CXXC5, encoding a retinoid-inducible nuclear factor (RINF), is located within a region at 5q31.2 commonly deleted in myelodysplastic syndrome and adult acute myeloid leukemia. RINF may act as an epigenetic regulator and has been proposed as a tumor suppressor in hematopoietic malignancies. However, functional studies in normal hematopoiesis are lacking, and its mechanism of action is unknown. Here, we evaluated the consequences of RINF silencing on cytokine-induced erythroid differentiation of human primary CD34+ progenitors. We found that RINF is expressed in immature erythroid cells and that RINF-knockdown accelerated erythropoietin-driven maturation, leading to a significant reduction (~45%) in the number of red blood cells, without affecting cell viability. The phenotype induced by RINF-silencing was dependent on tumor growth factor b (TGFb) and mediated by SMAD7, a TGFb-signaling inhibitor. RINF upregulates SMAD7 expression by direct binding to its promoter and we found a close correlation between RINF and SMAD7 mRNA levels both in CD34+ cells isolated from bone marrow of healthy donors and myelodysplastic syndrome patients with del(5q). Importantly, RINF knockdown attenuated SMAD7 expression in primary cells and ectopic SMAD7 expression was sufficient to prevent the RINF knockdown-dependent erythroid phenotype. Finally, RINF silencing affects 5’-hydroxymethylation of human erythroblasts, in agreement with its recently described role as a TET2-anchoring platform in mouse. Collectively, our data bring insight into how the epigenetic factor RINF, as a transcriptional regulator of SMAD7, may fine-tune cell sensitivity to TGFb superfamily cytokines and thus play an important role in both normal and pathological erythropoiesis
    corecore