16 research outputs found

    TOX3 mutations in breast cancer

    Get PDF
    TOX3 maps to 16q12, a region commonly lost in breast cancers and recently implicated in the risk of developing breast cancer. However, not much is known of the role of TOX3 itself in breast cancer biology. This is the first study to determine the importance of TOX3 mutations in breast cancers. We screened TOX3 for mutations in 133 breast tumours and identified four mutations (three missense, one in-frame deletion of 30 base pairs) in six primary tumours, corresponding to an overall mutation frequency of 4.5%. One potentially deleterious missense mutation in exon 3 (Leu129Phe) was identified in one tumour (genomic DNA and cDNA). Whilst copy number changes of 16q12 are common in breast cancer, our data show that mutations of TOX3 are present at low frequency in tumours. Our results support that TOX3 should be further investigated to elucidate its role in breast cancer biology.Breast Cancer Research Foundation grant; University of Cambridge; Cancer Research UK; Hutchison Whampoa Limited; NIHR Cambridge Biomedical Research Centre; Marie Curie Career Integration Grant; Cancer Research UK [16942]; National Institute for Health Research [NF-SI-0611-10154

    The methyl binding domain 3/nucleosome remodelling and deacetylase complex regulates neural cell fate determination and terminal differentiation in the cerebral cortex.

    Get PDF
    BACKGROUND: Chromatin-modifying complexes have key roles in regulating various aspects of neural stem cell biology, including self-renewal and neurogenesis. The methyl binding domain 3/nucleosome remodelling and deacetylation (MBD3/NuRD) co-repressor complex facilitates lineage commitment of pluripotent cells in early mouse embryos and is important for stem cell homeostasis in blood and skin, but its function in neurogenesis had not been described. Here, we show for the first time that MBD3/NuRD function is essential for normal neurogenesis in mice. RESULTS: Deletion of MBD3, a structural component of the NuRD complex, in the developing mouse central nervous system resulted in reduced cortical thickness, defects in the proper specification of cortical projection neuron subtypes and neonatal lethality. These phenotypes are due to alterations in PAX6+ apical progenitor cell outputs, as well as aberrant terminal neuronal differentiation programmes of cortical plate neurons. Normal numbers of PAX6+ apical neural progenitor cells were generated in the MBD3/NuRD-mutant cortex; however, the PAX6+ apical progenitor cells generate EOMES+ basal progenitor cells in reduced numbers. Cortical progenitor cells lacking MBD3/NuRD activity generate neurons that express both deep- and upper-layer markers. Using laser capture microdissection, gene expression profiling and chromatin immunoprecipitation, we provide evidence that MBD3/NuRD functions to control gene expression patterns during neural development. CONCLUSIONS: Our data suggest that although MBD3/NuRD is not required for neural stem cell lineage commitment, it is required to repress inappropriate transcription in both progenitor cells and neurons to facilitate appropriate cell lineage choice and differentiation programmes.We wish to thank Nicola Reynolds for the help with figures; Aoife O’Shaughnessy for the critical reading of the manuscript; Peter Humphreys, the SCI Biofacility staff and Margaret McLeish for technical assistance; Stephanie Hall and Gerard Evan for access to the Laser Capture Microscope and Nathalie Saurat and members of the BH lab for useful discussions. This work was supported by a Wellcome Trust Senior Fellowship in the Basic Biomedical Sciences awarded to BH and a bourse de formation from the Fonds de la Recherche en Santé Québec awarded to EK.This is the final published version of the article. It was originally published in Neural Development (Knock E, et al., Neural Development, 2015, 10:13, doi:10.1186/s13064-015-0040-z). The final version is available at http://dx.doi.org/10.1186/s13064-015-0040-

    The Methyl-CpG Binding Proteins Mecp2, Mbd2 and Kaiso Are Dispensable for Mouse Embryogenesis, but Play a Redundant Function in Neural Differentiation

    Get PDF
    The precise molecular changes that occur when a neural stem (NS) cell switches from a programme of self-renewal to commit towards a specific lineage are not currently well understood. However it is clear that control of gene expression plays an important role in this process. DNA methylation, a mark of transcriptionally silent chromatin, has similarly been shown to play important roles in neural cell fate commitment in vivo. While DNA methylation is known to play important roles in neural specification during embryonic development, no such role has been shown for any of the methyl-CpG binding proteins (Mecps) in mice.. No evidence for functional redundancy between these genes in embryonic development or in the derivation or maintenance of neural stem cells in culture was detectable. However evidence for a defect in neuronal commitment of triple knockout NS cells was found.Although DNA methylation is indispensable for mammalian embryonic development, we show that simultaneous deficiency of three methyl-CpG binding proteins genes is compatible with apparently normal mouse embryogenesis. Nevertheless, we provide genetic evidence for redundancy of function between methyl-CpG binding proteins in postnatal mice

    The methyl binding domain 3/nucleosome remodelling and deacetylase complex regulates neural cell fate determination and terminal differentiation in the cerebral cortex

    No full text
    Abstract Background Chromatin-modifying complexes have key roles in regulating various aspects of neural stem cell biology, including self-renewal and neurogenesis. The methyl binding domain 3/nucleosome remodelling and deacetylation (MBD3/NuRD) co-repressor complex facilitates lineage commitment of pluripotent cells in early mouse embryos and is important for stem cell homeostasis in blood and skin, but its function in neurogenesis had not been described. Here, we show for the first time that MBD3/NuRD function is essential for normal neurogenesis in mice. Results Deletion of MBD3, a structural component of the NuRD complex, in the developing mouse central nervous system resulted in reduced cortical thickness, defects in the proper specification of cortical projection neuron subtypes and neonatal lethality. These phenotypes are due to alterations in PAX6+ apical progenitor cell outputs, as well as aberrant terminal neuronal differentiation programmes of cortical plate neurons. Normal numbers of PAX6+ apical neural progenitor cells were generated in the MBD3/NuRD-mutant cortex; however, the PAX6+ apical progenitor cells generate EOMES+ basal progenitor cells in reduced numbers. Cortical progenitor cells lacking MBD3/NuRD activity generate neurons that express both deep- and upper-layer markers. Using laser capture microdissection, gene expression profiling and chromatin immunoprecipitation, we provide evidence that MBD3/NuRD functions to control gene expression patterns during neural development. Conclusions Our data suggest that although MBD3/NuRD is not required for neural stem cell lineage commitment, it is required to repress inappropriate transcription in both progenitor cells and neurons to facilitate appropriate cell lineage choice and differentiation programmes

    Neural differentiation of 3KO NS Cells.

    No full text
    <p>NS cells were cultured for 15 days in neuronal differentiation conditions and immunostained for ß-tubulin III (green) and Gfap (red) in panels 1 and 2, Map2 in panels 3 and 4, and for Gad67 (green), a marker of Gabaergic neurons, in panels 5 and 6. Cells were counterstained in Dapi (blue). Panels 1, 3, and 5: wild-type NS cells; panels 2, 4, and 6: 3KO NS cells. B. Quantitation of the number of ß-tubulinIII expressing (white bars) and Gfap-expressing (grey bars) cells in 14 day neural differentiation cultures from wild-type (left) and 3KO (right) NS cell cultures. Error bars represent SEM from three independent experiments. C. Quantitation of the number of ß-tubulinIII positive cells present in NS cell cultures after seven days' neuronal differentiation. Data is shown for three different wild-type and three different 3KO NS cell lines. Error bars represent SEM from at least three independent experiments. The difference seen between the wild-type and 3KO lines is highly statistically significant (p<0.0024, Bonferroni). D. Images of 7 day neural cultures from wild-type (left) or 3KO (right) NS cells, stained for ß-tubulin III (green) and Dapi (blue). E. Relative percentage of ß-tubulin III positive cells present in NS cell cultures of indicated genotype after eight days' neuronal differentiation. Error bars represent SEM from at least four independent fields of cells. All cell lines were derived from E14.5 cortex except those labeled “<i>Zbtb33</i> Flox” and “<i>Zbtb33</i> KO”, which are ES cell-derived NS lines, with the <i>Zbtb33</i> KO line having been derived from the <i>Zbtb33</i> Flox line by Cre-transfection and gene deletion. Hence data for the <i>Zbtb33</i> Flox line is included as an appropriate wild-type control for the <i>Zbtb33</i> -null NS line.</p

    Genetic interaction between Mecps in postnatal animals.

    No full text
    <p>A. Cumulative plot of percentage of Mecp2-null mice (solid black line), Mecp2/Kaiso-double null mice (dotted black line), Mecp2/Mbd2-double null mice (dotted grey line) and Mecp2/Mbd2/Kaiso-triple null mice (solid grey line) surviving over time. All mice used in this analysis were produced from mouse lines that had undergone at least 6 generations of backcrossing to C57Bl/6 mice. B. Statistical analysis of the data pictured in A. N refers to the number of mice in the sample. A two-tailed Mann-Whitney test (<a href="http://faculty.vassar.edu/lowry/utest.html" target="_blank">http://faculty.vassar.edu/lowry/utest.html</a>) was used to calculate p-values. p<sub>Mecp2</sub> gives the p-value as compared to the Mecp2 single-null data, and p<sub>3KO</sub> gives the p-value as compared to the triple null data.</p
    corecore