49
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      MyT1 Counteracts the Neural Progenitor Program to Promote Vertebrate Neurogenesis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          The generation of neurons from neural stem cells requires large-scale changes in gene expression that are controlled to a large extent by proneural transcription factors, such as Ascl1. While recent studies have characterized the differentiation genes activated by proneural factors, less is known on the mechanisms that suppress progenitor cell identity. Here, we show that Ascl1 induces the transcription factor MyT1 while promoting neuronal differentiation. We combined functional studies of MyT1 during neurogenesis with the characterization of its transcriptional program. MyT1 binding is associated with repression of gene transcription in neural progenitor cells. It promotes neuronal differentiation by counteracting the inhibitory activity of Notch signaling at multiple levels, targeting the Notch1 receptor and many of its downstream targets. These include regulators of the neural progenitor program, such as Hes1, Sox2, Id3, and Olig1. Thus, Ascl1 suppresses Notch signaling cell-autonomously via MyT1, coupling neuronal differentiation with repression of the progenitor fate.

          Graphical Abstract

          Highlights

          • MyT1 promotes neurogenesis downstream Ascl1

          • MyT1 represses Notch1 receptor and many of its downstream target genes

          • MyT1 represses Hes1 expression by direct DNA binding and competition with RBPJ

          • Ascl1 suppresses Notch signaling cell-autonomously while promoting differentiation

          Abstract

          Vasconcelos et al. find that the transcription factor MyT1 promotes neuronal differentiation downstream the proneural factor Ascl1. MyT1 represses the transcription of Notch signaling components and targets genes, including important regulators of the neural progenitor program. Thus, Ascl1 activates differentiation genes while repressing the progenitor program via MyT1.

          Related collections

          Most cited references42

          • Record: found
          • Abstract: found
          • Article: not found

          Direct conversion of fibroblasts to functional neurons by defined factors

          Cellular differentiation and lineage commitment are considered robust and irreversible processes during development. Recent work has shown that mouse and human fibroblasts can be reprogrammed to a pluripotent state with a combination of four transcription factors. This raised the question of whether transcription factors could directly induce other defined somatic cell fates, and not only an undifferentiated state. We hypothesized that combinatorial expression of neural lineage-specific transcription factors could directly convert fibroblasts into neurons. Starting from a pool of nineteen candidate genes, we identified a combination of only three factors, Ascl1, Brn2, and Myt1l, that suffice to rapidly and efficiently convert mouse embryonic and postnatal fibroblasts into functional neurons in vitro. These induced neuronal (iN) cells express multiple neuron-specific proteins, generate action potentials, and form functional synapses. Generation of iN cells from non-neural lineages could have important implications for studies of neural development, neurological disease modeling, and regenerative medicine.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            The cell biology of neurogenesis.

            During the development of the mammalian central nervous system, neural stem cells and their derivative progenitor cells generate neurons by asymmetric and symmetric divisions. The proliferation versus differentiation of these cells and the type of division are closely linked to their epithelial characteristics, notably, their apical-basal polarity and cell-cycle length. Here, we discuss how these features change during development from neuroepithelial to radial glial cells, and how this transition affects cell fate and neurogenesis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Signalling downstream of activated mammalian Notch.

              Notch belongs to a family of transmembrane proteins that are widely conserved from flies to vertebrates and are thought to be involved in cell-fate decisions. In Drosophila, the Suppressor of hairless (Su(H)) gene and genes of the Enhancer of split (E(Spl)) complex, which encode proteins of the basic helix-loop-helix type have been implicated in the Notch signalling pathway. Mammalian homologues of E(Spl), such as the mouse Hairy enhancer of split (HES-1), have been isolated. Both HES-1 and the intracellular domain of murine Notch (mNotch) are able to block MyoD-induced myogenesis. Here we show that activated forms of mNotch associate with the human analogue of Su(H), KBF2/RBP-J kappa (refs 8,9) and act as transcriptional activators through the KBF2-binding sites of the HES-1 promoter.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Rep
                Cell Rep
                Cell Reports
                Cell Press
                2211-1247
                04 October 2016
                04 October 2016
                04 October 2016
                : 17
                : 2
                : 469-483
                Affiliations
                [1 ]Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
                [2 ]Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
                [3 ]Department of Cell and Molecular Biology, Ludwig Institute for Cancer Research, Karolinska Institutet, 17177 Stockholm, Sweden
                Author notes
                []Corresponding author dscastro@ 123456igc.gulbenkian.pt
                [4]

                Lead Contact

                Article
                S2211-1247(16)31246-3
                10.1016/j.celrep.2016.09.024
                5067283
                27705795
                f2a8c5ee-274d-4d81-bcf8-c1ceeb26b379
                © 2016 The Author(s)

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 24 February 2016
                : 12 July 2016
                : 9 September 2016
                Categories
                Article

                Cell biology
                myt1,ascl1,neurogenesis,notch signaling,hes1,transcription,rbpj
                Cell biology
                myt1, ascl1, neurogenesis, notch signaling, hes1, transcription, rbpj

                Comments

                Comment on this article