31 research outputs found

    Tunicate cytostatic factor TC14-3 induces a polycomb group gene and histone modification through Ca2+ binding and protein dimerization

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>As many invertebrate species have multipotent cells that undergo cell growth and differentiation during regeneration and budding, many unique and interesting homeostatic factors are expected to exist in those animals. However, our understanding of such factors and global mechanisms remains very poor. Single zooids of the tunicate, <it>Polyandrocarpa </it><it>misakiensis</it>, can give off as many as 40 buds during the life span. Bud development proceeds by means of transdifferentiation of very limited number of cells and tissues. TC14-3 is one of several different but closely related polypeptides isolated from <it>P. misakiensis</it>. It acts as a cytostatic factor that regulates proliferation, adhesion, and differentiation of multipotent cells, although the molecular mechanism remains uncertain. The Polycomb group (PcG) genes are involved in epigenetic control of genomic activity in mammals. In invertebrates except <it>Drosophila</it>, PcG and histone methylation have not been studied so extensively, and genome-wide gene regulation is poorly understood.</p> <p>Results</p> <p>When Phe<sup>65 </sup>of TC14-3 was mutated to an acidic amino acid, the resultant mutant protein failed to dimerize. The replacement of Thr<sup>69 </sup>with Arg<sup>69 </sup>made dimers unstable. When Glu<sup>106 </sup>was changed to Gly<sup>106</sup>, the resultant mutant protein completely lost Ca<sup>2+ </sup>binding. All these mutant proteins lacked cytostatic activity, indicating the requirement of protein dimerization and calcium for the activity. <it>Polyandrocarpa </it><it>Eed</it>, a component of PcG, is highly expressed during budding, like TC14-3. When wild-type and mutant TC14-3s were applied in vivo and in vitro to <it>Polyandrocarpa </it>cells, only wild-type TC14-3 could induce <it>Eed </it>without affecting histone methyltransferase gene expression. Eed-expressing cells underwent trimethylation of histone H3 lysine27. <it>PmEed </it>knockdown by RNA interference rescued cultured cells from the growth-inhibitory effects of TC14-3.</p> <p>Conclusion</p> <p>These results show that in <it>P. misakiensis</it>, the cytostatic activity of TC14-3 is mediated by <it>PmEed </it>and resultant histone modification, and that the gene expression requires both the protein dimerization and Ca<sup>2+</sup>-binding of TC14-3. This system consisting of a humoral factor, PcG, and histone methylation would contribute to the homeostatic regulation of cell growth and terminal differentiation of invertebrate multipotent cells.</p

    BVES regulates c-Myc stability via PP2A and suppresses 1 colitis-induced 2 tumorigenesis

    Get PDF
    Objective Blood vessel epicardial substance (BVES) is a tight junction-associated protein that regulates epithelial-mesenchymal states and is underexpressed in epithelial malignancy. However, the functional impact of BVES loss on tumourigenesis is unknown. Here we define the in vivo role of BVES in colitis-associated cancer (CAC), its cellular function and its relevance to patients with IBD. Design We determined BVES promoter methylation status using an Infinium HumanMethylation450 array screen of patients with UC with and without CAC. We also measured BVES mRNA levels in a tissue microarray consisting of normal colons and CAC samples. Bves−/− and wild-type mice (controls) were administered azoxymethane (AOM) and dextran sodium sulfate (DSS) to induce tumour formation. Last, we used a yeast twohybrid screen to identify BVES interactors and performed mechanistic studies in multiple cell lines to define how BVES reduces c-Myc levels. Results BVES mRNA was reduced in tumours from patients with CAC via promoter hypermethylation. Importantly, BVES promoter hypermethylation was concurrently present in distant non-malignant-appearing mucosa. As seen in human patients, Bves was underexpressed in experimental inflammatory carcinogenesis, and Bves−/− mice had increased tumour multiplicity and degree of dysplasia after AOM/DSS administration. Molecular analysis of Bves−/− tumours revealed Wnt activation and increased c-Myc levels. Mechanistically, we identified a new signalling pathway whereby BVES interacts with PR61α, a protein phosphatase 2A regulatory subunit, to mediate c-Myc destruction. Conclusion Loss of BVES promotes inflammatory tumourigenesis through dysregulation of Wnt signalling and the oncogene c-Myc. BVES promoter methylation status may serve as a CAC biomarker
    corecore