14 research outputs found

    The transcriptional landscape of hematopoietic stem cell ontogeny

    Get PDF
    Transcriptome analysis of adult hematopoietic stem cells (HSCs) and their progeny has revealed mechanisms of blood differentiation and leukemogenesis, but a similar analysis of HSC development is lacking. Here, we acquired the transcriptomes of developing HSCs purified from >2,500 murine embryos and adult mice. We found that embryonic hematopoietic elements clustered into three distinct transcriptional states characteristic of the definitive yolk sac, HSCs undergoing specification, and definitive HSCs. We applied a network-biology-based analysis to reconstruct the gene regulatory networks of sequential stages of HSC development and functionally validated candidate transcriptional regulators of HSC ontogeny by morpholino-mediated knockdown in zebrafish embryos. Moreover, we found that HSCs from in vitro differentiated embryonic stem cells closely resemble definitive HSCs, yet lack a Notch-signaling signature, likely accounting for their defective lymphopoiesis. Our analysis and web resource will enhance efforts to identify regulators of HSC ontogeny and facilitate the engineering of hematopoietic specification

    CHMP1A encodes an essential regulator of BMI1-INK4A in cerebellar development

    Get PDF
    Charged multivesicular body protein 1A (CHMP1A; also known as chromatin-modifying protein 1A) is a member of the ESCRT-III (endosomal sorting complex required for transport-III) complex but is also suggested to localize to the nuclear matrix and regulate chromatin structure. Here, we show that loss-of-function mutations in human CHMP1A cause reduced cerebellar size (pontocerebellar hypoplasia) and reduced cerebral cortical size (microcephaly). CHMP1A-mutant cells show impaired proliferation, with increased expression of INK4A, a negative regulator of stem cell proliferation. Chromatin immunoprecipitation suggests loss of the normal INK4A repression by BMI in these cells. Morpholino-based knockdown of zebrafish chmp1a resulted in brain defects resembling those seen after bmi1a and bmi1b knockdown, which were partially rescued by INK4A ortholog knockdown, further supporting links between CHMP1A and BMI1-mediated regulation of INK4A. Our results suggest that CHMP1A serves as a critical link between cytoplasmic signals and BMI1-mediated chromatin modifications that regulate proliferation of central nervous system progenitor cells

    Editorial: Zebrafish Epigenetics

    Get PDF
    A key area of focus in the field of epigenetics pertains the comprehension of the functional relevance of the epigenetic mechanisms occurring during embryogenesis to shape normal developmental trajectories and adult phenotypes (Atlasi and Stunnenberg, 2017; Skvortsova et al., 2018; Cavalieri, 2021; Marchione et al., 2021). Several lines of evidence highlighted that the small freshwater cyprinid Danio rerio, commonly known as zebrafish, is an excellent vertebrate model for research purposes in the field of epigenetics (Huang et al., 2013; Balasubramanian et al., 2019; Horsfield, 2019; Cavalieri, 2020). The general strengths of zebrafish over concurrent models are well known: ease of husbandry and maintenance in laboratory, high fecundity, external fertilization, short life cycle and generation time. Beyond this, the increasing popularity of zebrafish for epigenetic research purposes is due to two main reasons. First, components of the epigenetic machinery have been widely characterized in zebrafish, showing overall conservation with mammals (Howe et al., 2013; Cavalieri and Spinelli, 2017). No less important, zebrafish embryos are optically translucent and relatively permeable to a wide range of compounds, allowing non-invasive live imaging of morphogenesis and phenotypes following exposure to environmental stressors that challenge the epigenome (Godinho, 2011; Ali et al., 2014). Altogether, these benefits also make zebrafish an outstanding model for large-scale screening of potential therapeutics targeting epigenetic regulatory mechanisms

    Ikaros Induces Quiescence and T-Cell Differentiation in a Leukemia Cell Line

    No full text
    Ikaros is a hematopoietic cell-specific zinc finger DNA binding protein that plays an important role in lymphocyte development. Genetic disruption of Ikaros results in T-cell transformation. Ikaros null mice develop leukemia with 100% penetrance. It has been hypothesized that Ikaros controls gene expression through its association with chromatin remodeling complexes. The development of leukemia in Ikaros null mice suggests that Ikaros has the characteristics of a tumor suppressor gene. In this report, we show that the introduction of Ikaros into an established mouse Ikaros null T leukemia cell line leads to growth arrest at the G(0)/G(1) stage of the cell cycle. This arrest is associated with up-regulation of the cell cycle-dependent kinase inhibitor p27(kip1), the induction of expression of T-cell differentiation markers, and a global and specific increase in histone H3 acetylation status. These studies provide strong evidence that Ikaros possesses the properties of a bona fide tumor suppressor gene for the T-cell lineage and offer insight into the mechanism of Ikaros's tumor suppressive activity

    Direct Recruitment of Polycomb Repressive Complex 1 to Chromatin by Core Binding Transcription Factors

    No full text
    Polycomb repressive complexes (PRCs) play key roles in developmental epigenetic regulation. Yet the mechanisms that target PRCs to specific loci in mammalian cells remain incompletely understood. In this study we show that Bmi1, a core component of Polycomb Repressive Complex 1 (PRC1), binds directly to the Runx1/CBFβ transcription factor complex. Genome-wide studies in megakaryocytic cells demonstrate significant chromatin occupancy overlap between the PRC1 core component Ring1b and Runx1/CBFβ and functional regulation of a considerable fraction of commonly bound genes. Bmi1/Ring1b and Runx1/CBFβ deficiencies generate partial phenocopies of one another in vivo. We also show that Ring1b occupies key Runx1 binding sites in primary murine thymocytes and that this occurs via PRC2-independent mechanisms. Genetic depletion of Runx1 results in reduced Ring1b binding at these sites in vivo. These findings provide evidence for site-specific PRC1 chromatin recruitment by core binding transcription factors in mammalian cells.National Institutes of Health (U.S.) (Grant U54-CA112967)National Institutes of Health (U.S.) (Grant R01-GM089903)National Science Foundation (U.S.) (Award DB1-0821391)National Institutes of Health (U.S.) (P30-ES002109
    corecore