Abstract

Genomic imprinting refers to the mono-allelic and parent-specific expression of a subset of genes. While long recognized for their role in embryonic development, imprinted genes have recently emerged as important modulators of postnatal physiology, notably through hypothalamus-driven functions. Here, using mouse models of loss, gain and parental inversion of expression, we report that the paternally expressed Zdbf2 gene controls neonatal growth in mice, in a dose-sensitive but parent-of-origin-independent manner. We further found that Zdbf2-KO neonates failed to fully activate hypothalamic circuits that stimulate appetite, and suffered milk deprivation and diminished circulating Insulin Growth Factor 1 (IGF-1). Consequently, only half of Zdbf2-KO pups survived the first days after birth and those surviving were smaller. This study demonstrates that precise imprinted gene dosage is essential for vital physiological functions at the transition from intra- to extra-uterine life, here the adaptation to oral feeding and optimized body weight gain.

Data availability

All data generated or analysed during this study are included in the manuscript and supporting files.RNA-Seq data have been deposited in GEO under accession code GSE153265

The following data sets were generated

Article and author information

Author details

  1. Juliane Glaser

    Department of Genetics and Developmental Biology, Institut Curie, Paris, France
    Competing interests
    No competing interests declared.
  2. Julian Iranzo

    Department of Genetics and Developmental Biology, Institut Curie, Paris, France
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-9369-2530
  3. Maud Borensztein

    Department of Genetics and Developmental Biology, Institut Curie, Paris, France
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-4378-5018
  4. Mattia Marinucci

    Department of Genetics and Developmental Biology, Institut Curie, Paris, France
    Competing interests
    No competing interests declared.
  5. Angelica Gualtieri

    Centre for Endocrinology, Queen Mary University of London, London, United Kingdom
    Competing interests
    No competing interests declared.
  6. Colin Jouhanneau

    Animal Transgenesis Platform, Institut Curie, Paris, France
    Competing interests
    No competing interests declared.
  7. Aurélie Teissandier

    Department of Genetics and Developmental Biology, Institut Curie, Paris, France
    Competing interests
    No competing interests declared.
  8. Carles Gaston-Massuet

    Centre for Endocrinology, Queen Mary University of London, London, United Kingdom
    Competing interests
    No competing interests declared.
  9. Deborah Bourc'his

    Department of Genetics and Developmental Biology, Institut Curie, Paris, France
    For correspondence
    deborah.bourchis@curie.fr
    Competing interests
    Deborah Bourc'his, Reviewing editor, eLife.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-9499-7291

Funding

FP7 Ideas: European Research Council (ERC-Cog EpiRepro)

  • Aurélie Teissandier
  • Deborah Bourc'his

Fondation Bettencourt Schueller

  • Deborah Bourc'his

Ligue Contre le Cancer

  • Juliane Glaser

BTL Charity (GN417/2238)

  • Angelica Gualtieri
  • Carles Gaston-Massuet

Action Medical Research (GN2272)

  • Angelica Gualtieri
  • Carles Gaston-Massuet

DIM Biotherapies

  • Juliane Glaser

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Joshua M Brickman, University of Copenhagen, Denmark

Ethics

Animal experimentation: All experimentation was approved by the Animal Care and Use Committee of the Institut Curie (agreement number: C 75-05-18) and adhered to European and National Regulation for the Protection of Vertebrate Animals used for Experimental and other Scientific Purposes (Directive 86/609 and 2010/63)

Version history

  1. Received: December 10, 2020
  2. Preprint posted: January 30, 2021 (view preprint)
  3. Accepted: January 19, 2022
  4. Accepted Manuscript published: January 20, 2022 (version 1)
  5. Version of Record published: February 2, 2022 (version 2)

Copyright

© 2022, Glaser et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 1,229
    Page views
  • 229
    Downloads
  • 5
    Citations

Article citation count generated by polling the highest count across the following sources: Crossref, PubMed Central, Scopus.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Juliane Glaser
  2. Julian Iranzo
  3. Maud Borensztein
  4. Mattia Marinucci
  5. Angelica Gualtieri
  6. Colin Jouhanneau
  7. Aurélie Teissandier
  8. Carles Gaston-Massuet
  9. Deborah Bourc'his
(2022)
The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates
eLife 11:e65641.
https://doi.org/10.7554/eLife.65641

Share this article

https://doi.org/10.7554/eLife.65641

Further reading

    1. Chromosomes and Gene Expression
    2. Genetics and Genomics
    Erandi Velazquez-Miranda, Ming He
    Insight

    Endothelial cell subpopulations are characterized by unique gene expression profiles, epigenetic landscapes and functional properties.

    1. Cell Biology
    2. Chromosomes and Gene Expression
    Monica Salinas-Pena, Elena Rebollo, Albert Jordan
    Research Article

    Histone H1 participates in chromatin condensation and regulates nuclear processes. Human somatic cells may contain up to seven histone H1 variants, although their functional heterogeneity is not fully understood. Here, we have profiled the differential nuclear distribution of the somatic H1 repertoire in human cells through imaging techniques including super-resolution microscopy. H1 variants exhibit characteristic distribution patterns in both interphase and mitosis. H1.2, H1.3, and H1.5 are universally enriched at the nuclear periphery in all cell lines analyzed and co-localize with compacted DNA. H1.0 shows a less pronounced peripheral localization, with apparent variability among different cell lines. On the other hand, H1.4 and H1X are distributed throughout the nucleus, being H1X universally enriched in high-GC regions and abundant in the nucleoli. Interestingly, H1.4 and H1.0 show a more peripheral distribution in cell lines lacking H1.3 and H1.5. The differential distribution patterns of H1 suggest specific functionalities in organizing lamina-associated domains or nucleolar activity, which is further supported by a distinct response of H1X or phosphorylated H1.4 to the inhibition of ribosomal DNA transcription. Moreover, H1 variants depletion affects chromatin structure in a variant-specific manner. Concretely, H1.2 knock-down, either alone or combined, triggers a global chromatin decompaction. Overall, imaging has allowed us to distinguish H1 variants distribution beyond the segregation in two groups denoted by previous ChIP-Seq determinations. Our results support H1 variants heterogeneity and suggest that variant-specific functionality can be shared between different cell types.