101 research outputs found

    YAP contributes to DNA methylation remodeling upon mouse embryonic stem cell differentiation

    Get PDF
    The Yes-associated protein YAP, one of the major effectors of the Hippo pathway together with its related protein TAZ, mediates a range of cellular processes from proliferation and death to morphogenesis. YAP and TAZ regulate a large number of target genes, acting as co-activators of DNA-binding transcription factors or as negative regulators of transcription by interacting with the nucleosome remodeling and histone deacetylase complexes. YAP is expressed in self-renewing embryonic stem cells (ESCs), although it is still debated whether it plays any crucial roles in the control of either stemness or differentiation. Here we show that the transient downregulation of YAP in mouse ESCs perturbs cellular homeostasis, leading to the inability to differentiate properly. Bisulfite genomic sequencing revealed that this transient knockdown caused a genome-wide alteration of the DNA methylation remodeling that takes place during the early steps of differentiation, suggesting that the phenotype we observed might be due to the dysregulation of some of the mechanisms involved in regulation of ESC exit from pluripotency. By gene expression analysis we identified two molecules which could have a role in the altered genome-wide methylation profile: the long non-coding RNA Ephemeron, whose rapid upregulation is crucial for ESCs transition into epiblast, and the methyltransferase-like protein Dnmt3l, which, during the embryo development, cooperates with Dnmt3a and Dnmt3b to contribute to the de novo DNA methylation that governs early steps of ESC differentiation. These data suggest a new role for YAP in the governance of the epigenetic dynamics of exit from pluripotency

    Protein aggregation of the p63 transcription factor underlies severe skin fragility in AEC syndrome.

    Get PDF
    The p63 gene encodes a master regulator of epidermal commitment, development, and differentiation. Heterozygous mutations in the C-terminal domain of the p63 gene can cause ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome, a life-threatening disorder characterized by skin fragility and severe, long-lasting skin erosions. Despite deep knowledge of p63 functions, little is known about mechanisms underlying disease pathology and possible treatments. Here, we show that multiple AEC-associated p63 mutations, but not those causative of other diseases, lead to thermodynamic protein destabilization, misfolding, and aggregation, similar to the known p53 gain-of-function mutants found in cancer. AEC mutant proteins exhibit impaired DNA binding and transcriptional activity, leading to dominant negative effects due to coaggregation with wild-type p63 and p73. Importantly, p63 aggregation occurs also in a conditional knock-in mouse model for the disorder, in which the misfolded p63 mutant protein leads to severe epidermal defects. Variants of p63 that abolish aggregation of the mutant proteins are able to rescue p63's transcriptional function in reporter assays as well as in a human fibroblast-to-keratinocyte conversion assay. Our studies reveal that AEC syndrome is a protein aggregation disorder and opens avenues for therapeutic intervention.This work was supported by Telethon Grants GGP09230 and GGP16235 (to C.M.), ERA-Net Research Program on Rare Diseases (ERARE-2) Skin-Dev (C.M.), Italian Association for Cancer Research Grant IG2011-N.11369 (to C.M.), Fondation Dind-Cottier pour la recherche sur la peau (C.M.), DFG Grant DO 545/8-1 (to V.D.), the Centre for Biomolecular Magnetic Resonance, and the Cluster of Excellence Frankfurt (Macromolecular Complexes). P.G. is supported by a Lichtenberg Professorship of the Volkswagen Foundation. C.R. is a PhD student in molecular oncology at the European School of Molecular Medicine

    Loss of p53 activates thyroid hormone via type 2 deiodinase and enhances DNA damage

    Get PDF
    : The Thyroid Hormone (TH) activating enzyme, type 2 Deiodinase (D2), is functionally required to elevate the TH concentration during cancer progression to advanced stages. However, the mechanisms regulating D2 expression in cancer still remain poorly understood. Here, we show that the cell stress sensor and tumor suppressor p53 silences D2 expression, thereby lowering the intracellular THs availability. Conversely, even partial loss of p53 elevates D2/TH resulting in stimulation and increased fitness of tumor cells by boosting a significant transcriptional program leading to modulation of genes involved in DNA damage and repair and redox signaling. In vivo genetic deletion of D2 significantly reduces cancer progression and suggests that targeting THs may represent a general tool reducing invasiveness in p53-mutated neoplasms

    Direct targets of Klf5 transcription factor contribute to the maintenance of mouse embryonic stem cell undifferentiated state

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>A growing body of evidence has shown that Krüppel-like transcription factors play a crucial role in maintaining embryonic stem cell (ESC) pluripotency and in governing ESC fate decisions. Krüppel-like factor 5 (Klf5) appears to play a critical role in these processes, but detailed knowledge of the molecular mechanisms of this function is still not completely addressed.</p> <p>Results</p> <p>By combining genome-wide chromatin immunoprecipitation and microarray analysis, we have identified 161 putative primary targets of Klf5 in ESCs. We address three main points: (1) the relevance of the pathways governed by Klf5, demonstrating that suppression or constitutive expression of single Klf5 targets robustly affect the ESC undifferentiated phenotype; (2) the specificity of Klf5 compared to factors belonging to the same family, demonstrating that many Klf5 targets are not regulated by Klf2 and Klf4; and (3) the specificity of Klf5 function in ESCs, demonstrated by the significant differences between Klf5 targets in ESCs compared to adult cells, such as keratinocytes.</p> <p>Conclusions</p> <p>Taken together, these results, through the definition of a detailed list of Klf5 transcriptional targets in mouse ESCs, support the important and specific functional role of Klf5 in the maintenance of the undifferentiated ESC phenotype.</p> <p>See: <url>http://www.biomedcental.com/1741-7007/8/125</url></p

    ECMO for COVID-19 patients in Europe and Israel

    Get PDF
    Since March 15th, 2020, 177 centres from Europe and Israel have joined the study, routinely reporting on the ECMO support they provide to COVID-19 patients. The mean annual number of cases treated with ECMO in the participating centres before the pandemic (2019) was 55. The number of COVID-19 patients has increased rapidly each week reaching 1531 treated patients as of September 14th. The greatest number of cases has been reported from France (n = 385), UK (n = 193), Germany (n = 176), Spain (n = 166), and Italy (n = 136) .The mean age of treated patients was 52.6 years (range 16–80), 79% were male. The ECMO configuration used was VV in 91% of cases, VA in 5% and other in 4%. The mean PaO2 before ECMO implantation was 65 mmHg. The mean duration of ECMO support thus far has been 18 days and the mean ICU length of stay of these patients was 33 days. As of the 14th September, overall 841 patients have been weaned from ECMO support, 601 died during ECMO support, 71 died after withdrawal of ECMO, 79 are still receiving ECMO support and for 10 patients status n.a. . Our preliminary data suggest that patients placed on ECMO with severe refractory respiratory or cardiac failure secondary to COVID-19 have a reasonable (55%) chance of survival. Further extensive data analysis is expected to provide invaluable information on the demographics, severity of illness, indications and different ECMO management strategies in these patients

    Role of p63 in skin biology: its function in cell proliferation and differentiation

    Get PDF
    p63, a p53 family member, plays an essential role in epidermal development controlling several biological functions, some of which remain poorly understood. Using a global gene expression analysis, I contributed to the identification of novel downstream targets and signaling pathways regulated by p63. We firstly identified a set of genes not specifically expressed in epidermis (non-epidermal genes) that are indirectly suppressed by p63. In parallel, p63 sustains Bone Morphogenetic Protein (BMP) signaling by directly suppressing transcription of the inhibitory Smad7, and by inducing Bmp7. In the absence of p63, BMP signaling is compromised and leads to ectopic expression of the non-epidermal genes in vitro and in vivo. Reactivation of BMP signaling by exogenous stimuli suppresses ectopic expression of non-epidermal genes in the absence of p63. These data indicate that p63 prevents ectopic expression of non-epidermal genes by a mechanism involving activation of BMP signaling. In our genome-wide analysis we also observed that many cell cycle genes were positively regulated by p63. I found that loss of p63 in keratinocytes causes cell cycle arrest both in vitro and in vivo. Thus, I investigated how cell cycle genes are affected by p63. I identified a new mechanism through which p63 favors cell cycle progression repressing two members of a microRNA family: miR-34a and miR-34c. In the absence of p63, I observed increased levels of miR-34a and miR-34c in primary mouse keratinocytes and in mouse embryonic skin. p63 directly binds to p53-consensus sites in both miR-34a and miR-34c gene promoters and inhibits their activity. Keratinocytes are arrested in the G1-phase of the cell cycle in the absence of p63 and have reduced levels of two cell cycle regulators, cyclin D1 and cyclin-dependent kinase 4 (Cdk4), that are known targets of miR-34. Importantly, concomitant downregulation of miR-34a and miR-34c in the absence of p63 restores cell cycle progression and the expression of cyclin D1 and Cdk4. These data demonstrate that p63 sustains cell cycle progression in keratinocytes not only by previously defined mechanisms, such as repression of the CDK inhibitor p21 Cip1/Waf1, but also by directly repressing components of the miR-34 family. In conclusion, during my PhD program I uncovered two novel mechanisms through which p63 regulates cell cycle progression and tissue identity in epidermis

    p63 in Squamous Cell Carcinoma of the Skin: More Than a Stem Cell/Progenitor Marker

    No full text
    The p63 gene is often overexpressed in squamous cell carcinomas; however, how its overexpression contributes to tumor formation and expansion is still incompletely understood. Devos et al. report the development of a versatile mouse model demonstrating that p63 facilitates squamous cell carcinoma formation in skin and providing an excellent tool to dissect the relevance of its downstream signaling pathways in tumorigenesis
    corecore