Elsevier

Neuroscience

Volume 147, Issue 2, 29 June 2007, Pages 373-387
Neuroscience

Cellular neuroscience
Licensing regulators Geminin and Cdt1 identify progenitor cells of the mouse CNS in a specific phase of the cell cycle

https://doi.org/10.1016/j.neuroscience.2007.03.050Get rights and content

Abstract

Nervous system formation integrates control of cellular proliferation and differentiation and is mediated by multipotent neural progenitor cells that become progressively restricted in their developmental potential before they give rise to differentiated neurons and glial cells. Evidence from different experimental systems suggests that Geminin is a candidate molecule linking proliferation and differentiation during nervous system development. We show here that Geminin and its binding partner Cdt1 are expressed abundantly by neural progenitor cells during early mouse neurogenesis. Their expression levels decline at late developmental stages and become undetectable upon differentiation. Geminin and Cdt1 expressing cells also express Sox2 while no overlap is detected with cells expressing markers of a differentiated neuronal phenotype. A fraction of radial glial cells expressing RC2 and Pax6 are also immunoreactive for Geminin and Cdt1. The majority of the Geminin and Cdt1 expressing cell populations appears to be distinct from fate-restricted precursor cells expressing Mash1 or Neurogenin2. Bromo-deoxy-uridine (BrdU) incorporation experiments reveal a cell cycle specific expression in neural progenitor cells, with Geminin being present from S to M phase, while Cdt1 expression characterizes progenitor cells in G1 phase. Furthermore, in vitro differentiation of adult neurosphere cultures shows downregulation of Geminin/Cdt1 in the differentiated state, in line with our data showing that Geminin is present in neural progenitor cells of the CNS during mouse embryogenesis and adulthood and becomes downregulated upon cell fate specification and differentiation. This suggests a role for Geminin in the formation and maintenance of the neural progenitor cells.

Section snippets

Plasmids

We cloned by PCR the full-length open reading frames of mouse Geminin and Cdt1 into EcoRI/BamHI and BamHI/HindIII sites of pBluescript KS, respectively, using the following set of primers: Geminin, sense 5′-CGGAATTCTGAAAAATGAATCTCAGTATG-3′; anti-sense 5′-GCGGATCCATAGCCCCAGGATTCAAACCTG-3′; Cdt1, sense 5′-GTGGATCCACTACCTTTTTGGCGCCATG-3′; anti-sense 5′-GGTAAGCTTCTCTGTGACAGGAGG-3′ (Xouri et al., 2004).

In situ hybridization

Non-radioactive in situ hybridization was performed using 12 μm fresh-frozen sections of embryonic

Distribution of Geminin and Cdt1 mRNA in the developing CNS

In order to gain an insight into the role of Geminin and Cdt1 in mammalian neurogenesis, we initially characterized their spatial and temporal expression patterns in the CNS during mouse embryonic development, by in situ hybridization (ISH) on E11.5 (data not shown), E12.5 and E15.5 mouse embryos (Fig. 1).

At E12.5, Geminin mRNA can be detected relatively uniformly in the VZ of the cerebral cortex and the ganglionic eminences of the forebrain (Fig. 1A). More posteriorly, it is expressed in the

Discussion

Geminin was first described as a bifunctional protein, regulating both DNA replication and neural cell fate acquisition (Kroll et al 1998, McGarry and Kirschner 1998). Multiple roles have been attributed to Geminin during neurogenesis using different experimental systems (Quinn et al 2001, Del Bene et al 2004, Luo et al 2004, Seo et al 2005a). We undertook an analysis of Geminin and Cdt1 expression during mouse development and in adult brain using different markers to characterize the various

Conclusion

In summary, we have thoroughly defined the expression profiles of Geminin and Cdt1 during mouse neurogenesis. We have demonstrated that both proteins characterize the proliferating neurogenic regions of the developing CNS and that they delineate a specific population of neural precursor cells not yet committed toward a neuronal subtype, providing a potential new tool to further discriminate proliferating and immature cells as they progress from neural stem cells to transit amplifying progenitor

Acknowledgments

We would like to thank Dr. Jordane Malaterre and Mr. Sebastian Dworkin for help and advice and Dr. P. Liodis for critical reading of the manuscript. We are also thankful to Dr. V. Pachnis and Dr D. Kioussis for their support and critical reading of the manuscript. This work was supported by the Karatheodori program of the University of Patras and the Association for International Cancer Research, UK to S. Taraviras.

References (59)

  • P. Malatesta et al.

    Neuronal or glial progeny: regional differences in radial glia fate

    Neuron

    (2003)
  • S. Matsumoto et al.

    Brg1 is required for murine neural stem cell maintenance and gliogenesis

    Dev Biol

    (2006)
  • T.J. McGarry et al.

    Geminin, an inhibitor of DNA replication, is degraded during mitosis

    Cell

    (1998)
  • J.P. Misson et al.

    Identification of radial glial cells within the developing murine central nervous system: studies based upon a new immunohistochemical marker

    Brain Res Dev Brain Res

    (1988)
  • M. Nieto et al.

    Neural bHLH genes control the neuronal versus glial fate decision in cortical progenitors

    Neuron

    (2001)
  • H. Nishitani et al.

    The human licensing factor for DNA replication Cdt1 accumulates in G1 and is destabilized after initiation of S-phase

    J Biol Chem

    (2001)
  • L. Pevny et al.

    SOX genes and neural progenitor identity

    Curr Opin Neurobiol

    (2005)
  • L. Pevny et al.

    The stem-cell menagerie

    Trends Neurosci

    (2003)
  • S. Saxena et al.

    Geminin-Cdt1 balance is critical for genetic stability

    Mutat Res

    (2005)
  • Y. Sun et al.

    Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanisms

    Cell

    (2001)
  • J.J. Taylor et al.

    Tcf- and Vent-binding sites regulate neural-specific geminin expression in the gastrula embryo

    Dev Biol

    (2006)
  • L. Wilson et al.

    The mechanisms of dorsoventral patterning in the vertebrate neural tube

    Dev Biol

    (2005)
  • S.A. Anthony et al.

    Radial glia serve as neuronal progenitors in all regions of the central nervous system

    Neuron

    (2004)
  • K. Barami et al.

    Hu protein as an early marker of neuronal phenotypic differentiation by subependymal zone cells of the adult songbird forebrain

    J Neurobiol

    (1995)
  • N. Bertrand et al.

    Proneural genes and the specification of neural cell types

    Nat Rev Neurosci

    (2002)
  • O. Britz et al.

    A role for proneural genes in the maturation of cortical progenitor cells

    Cereb Cortex

    (2006)
  • J.P. Brown et al.

    Transient expression of doublecortin during adult neurogenesis

    J Comp Neurol

    (2003)
  • S. Couillard-Despres et al.

    Doublecortin expression levels in adult brain reflect neurogenesis

    Eur J Neurosci

    (2005)
  • F. Del Bene et al.

    Direct interaction of geminin and Six3 in eye development

    Nature

    (2004)
  • Cited by (36)

    • Intrinsic neural stem cell properties define brain hypersensitivity to genotoxic stress

      2022, Stem Cell Reports
      Citation Excerpt :

      To define the consequences of defective replication in cortical development, we investigated the response of early progenitors (herein referred to as NECs) and mid-neurogenesis progenitors (referred to as NPs) to impaired replication licensing by depleting Geminin (GMNN). GMNN is a known inhibitor of DNA replication licensing that is expressed in the proliferating neural stem and progenitor cells and is downregulated upon cell-cycle exit and differentiation (Spella et al., 2007). Previous studies have shown that suppression of GMNN in cultured cells induces aberrant licensing and incomplete genome replication (Huang et al., 2015; Klotz-Noack et al., 2012; Melixetian et al., 2004).

    • Adult Neural Stem Cells and Multiciliated Ependymal Cells Share a Common Lineage Regulated by the Geminin Family Members

      2019, Neuron
      Citation Excerpt :

      Then the DNA fragment corresponding to the GemC1 cDNA size was isolated from an agarose gel and ligated to the pBlueScriptKS backbone, upstream of the T3 promoter sequence. The pBlueScriptKS-Geminin plasmid was a gift from the laboratory of S. Taraviras (Spella et al., 2007). Briefly, in this study, the open reading frame of Geminin was cloned between the EcoRI/BamHI sites of the pBlueScriptKS plasmid, upstream of the T7 promoter sequence.

    • Inactivation of geminin in neural crest cells affects the generation and maintenance of enteric progenitor cells, leading to enteric aganglionosis

      2016, Developmental Biology
      Citation Excerpt :

      NCCs delaminate from the dorsal part of the NT by undergoing an epithelial to mesenchymal transition (EMT). Geminin expression starts as cells enter into S phase, a stage that has been shown to be essential for the efficient formation of the NCCs (Burstyn-Cohen and Kalcheim, 2002; Spella et al., 2007; Xouri et al., 2004). Moreover, a role in EMT and regulation of E-cadherin expression has been attributed to Geminin during early gastrulation (Emmett and O'Shea, 2012), which suggests a potential mechanism for the defects in the generation of early NCCs observed in mice that lack Geminin expression

    • The Geminin and Idas coiled coils preferentially form a heterodimer that Inhibits Geminin function in DNA replication licensing

      2013, Journal of Biological Chemistry
      Citation Excerpt :

      High levels of Idas have been previously reported in the choroid plexus and the cortical hem of the mouse telencephalon (24). In this specific part of the brain Geminin is predominantly expressed by neural progenitors (48), whereas Idas levels become higher in specific differentiated cell types (Pefani et al. (24)). High levels of Idas have also been reported in other developing multiciliated cells (Stubbs et al. (25)).

    • Reduced Geminin levels promote cellular senescence

      2013, Mechanisms of Ageing and Development
    View all citing articles on Scopus
    1

    Present address: Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA (O. Britz); Department of Pharmaceutical Biology, Victorian College of Pharmacy, Monash University, Victoria 3052, Australia (T. Mantamadiotis).

    View full text