10 research outputs found

    E4F1-mediated control of pyruvate dehydrogenase activity is essential for skin homeostasis.

    Get PDF
    The multifunctional protein E4 transcription factor 1 (E4F1) is an essential regulator of epidermal stem cell (ESC) maintenance. Here, we found that E4F1 transcriptionally regulates a metabolic program involved in pyruvate metabolism that is required to maintain skin homeostasis. E4F1 deficiency in basal keratinocytes resulted in deregulated expression of dihydrolipoamide acetyltransferase (Dlat), a gene encoding the E2 subunit of the mitochondrial pyruvate dehydrogenase (PDH) complex. Accordingly, E4f1 knock-out (KO) keratinocytes exhibited impaired PDH activity and a redirection of the glycolytic flux toward lactate production. The metabolic reprogramming of E4f1 KO keratinocytes associated with remodeling of their microenvironment and alterations of the basement membrane, led to ESC mislocalization and exhaustion of the ESC pool. ShRNA-mediated depletion of Dlat in primary keratinocytes recapitulated defects observed upon E4f1 inactivation, including increased lactate secretion, enhanced activity of extracellular matrix remodeling enzymes, and impaired clonogenic potential. Altogether, our data reveal a central role for Dlat in the metabolic program regulated by E4F1 in basal keratinocytes and illustrate the importance of PDH activity in skin homeostasis

    identification of new signaling pathways involving the multifunctional protein E4F1 in stem cell homeostasis

    No full text
    La protéine E4F1 a été identifiée comme une cible cellulaire de l'onco-protéine virale E1A au cours de l'infection par l'adénovirus de type V. Bien que très peu étudiée jusqu'à présent, les travaux du laboratoire d'accueil indiquent cependant qu'E4F1 est une protéine multifonctionnelle ayant à la fois un rôle de facteur de transcription mais fonctionnant également comme une E3 ligase vis-à-vis d'autres régulateurs transcriptionnels tels que les membres de la famille p53. E4F1 se situe au carrefour de plusieurs voies de signalisation fréquemment altérées au cours de la progression tumorale, notamment les voies impliquant les suppresseurs de tumeurs Rb et p53. In vivo, E4F1 est impliqué dans l'homéostasie des cellules souches à travers différents mécanismes encore non élucidés. L'objectif de mon projet de thèse est d'identifier et de caractériser les voies de signalisation impliquant E4F1 dans l'homéostasie des cellules souches. Pour répondre à cette question, j'utilise diverses techniques : Histologie, IHC,FACS, QPCR et des tests clonogénique sur des cellules issues des épidermes de souris E4F1 WT et KO.E4F1 protein has been first identified in the 80's as a cellular target of the adenoviral oncoprotein E1A. E4F1 is an atypical multifunctional protein with two different activities. In fact, it can acts as a transcriptional factor thanks to its DNA binding domain and exhibits either positive or negative transcriptional activities depending on the promoter context. In addition to its transcriptional activities, E4F1 is also an atypical E3-ligase for other transcription factors, including the p53 tumor suppressor. A growing body of evidence suggests that the multifunctional protein E4F1 is involved in signaling pathways that play essential roles during normal development and tumorigenesis. Moreover, we have shown that skin defect is one of the most striking phenotype of E4F1 conditional knockout mice. E4F1 inactivation in the entire skin or in the basal compartment of the epidermis (K5 specific knockout mice) induces skin homeostasis defects, as evidenced by transient hyperplasia in the interfollicular epithelium and alteration of keratinocyte differentiation, followed by loss of cellularity in the epidermis and severe skin ulcerations. E4F1 depletion alters clonogenic activity of epidermal stem cells (ESCs) ex vivo and ends in exhaustion of the ESC pool in vivo, indicating that the lesions observed in the E4F1 mutant skin result, at least in part, from cell-autonomous alterations in ESC maintenance. The clonogenic potential of E4F1 KO ESCs is partially rescued by Bmi1 overexpression, or by Ink4a/Arf overexpression or p53 depletion. In additions, in vivo, skin phenotype of E4F1 KO mice is also delayed in animals with Ink4a/Arf and E4F1 compound gene deficiencies. Taking together, our data show the essential role of E4F1 protein into Bmi1-Arf-p53 pathway to regulate ESC-dependent skin homeostasis

    The study of the determinants controlling Arpp19 phosphatase-inhibitory activity reveals an Arpp19/PP2A-B55 feedback loop

    No full text
    International audienceAbstract Arpp19 is a potent PP2A-B55 inhibitor that regulates this phosphatase to ensure the stable phosphorylation of mitotic/meiotic substrates. At G2-M, Arpp19 is phosphorylated by the Greatwall kinase on S67. This phosphorylated Arpp19 form displays a high affinity to PP2A-B55 and a slow dephosphorylation rate, acting as a competitor of PP2A-B55 substrates. The molecular determinants conferring slow dephosphorylation kinetics to S67 are unknown. PKA also phosphorylates Arpp19. This phosphorylation performed on S109 is essential to maintain prophase I-arrest in Xenopus oocytes although the underlying signalling mechanism is elusive. Here, we characterize the molecular determinants conferring high affinity and slow dephosphorylation to S67 and controlling PP2A-B55 inhibitory activity of Arpp19. Moreover, we show that phospho-S109 restricts S67 phosphorylation by increasing its catalysis by PP2A-B55. Finally, we discover a double feed-back loop between these two phospho-sites essential to coordinate the temporal pattern of Arpp19-dependent PP2A-B55 inhibition and Cyclin B/Cdk1 activation during cell division

    The Na/K-ATPase Oxidant Amplification Loop Regulates Aging

    No full text
    As aging involves oxidant injury, we examined the role of the recently described Na/K-ATPase oxidant amplification loop (NKAL). First, C57Bl6 old mice were given a western diet to stimulate oxidant injury or pNaKtide to antagonize the NKAL. The western diet accelerated functional and morphological evidence for aging whereas pNaKtide attenuated these changes. Next, human dermal fibroblasts (HDFs) were exposed to different types of oxidant stress in vitro each of which increased expression of senescence markers, cell-injury, and apoptosis as well as stimulated the NKAL. Further stimulation of the NKAL with ouabain augmented cellular senescence whereas treatment with pNaKtide attenuated it. Although N-Acetyl Cysteine and Vitamin E also ameliorated overall oxidant stress to a similar degree as pNaKtide, the pNaKtide produced protection against senescence that was substantially greater than that seen with either antioxidant. In particular, pNaKtide appeared to specifically ameliorate nuclear oxidant stress to a greater degree. These data demonstrate that the NKAL is intimately involved in the aging process and may serve as a target for anti-aging interventions

    Transcription factor E4F1 is essential for epidermal stem cell maintenance and skin homeostasis

    Get PDF
    A growing body of evidence suggests that the multifunctional protein E4F1 is involved in signaling pathways that play essential roles during normal development and tumorigenesis. We generated E4F1 conditional knockout mice to address E4F1 functions in vivo in newborn and adult skin. E4F1 inactivation in the entire skin or in the basal compartment of the epidermis induces skin homeostasis defects, as evidenced by transient hyperplasia in the interfollicular epithelium and alteration of keratinocyte differentiation, followed by loss of cellularity in the epidermis and severe skin ulcerations. E4F1 depletion alters clonogenic activity of epidermal stem cells (ESCs) ex vivo and ends in exhaustion of the ESC pool in vivo, indicating that the lesions observed in the E4F1 mutant skin result, at least in part, from cell-autonomous alterations in ESC maintenance. The clonogenic potential of E4F1 KO ESCs is rescued by Bmi1 overexpression or by Ink4a/Arf or p53 depletion. Skin phenotype of E4F1 KO mice is also delayed in animals with Ink4a/Arf and E4F1 compound gene deficiencies. Our data identify a regulatory axis essential for ESC-dependent skin homeostasis implicating E4F1 and the Bmi1–Arf–p53 pathway
    corecore