The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates

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...

Full description

Bibliographic Details
Main Authors: Juliane Glaser, Julian Iranzo, Maud Borensztein, Mattia Marinucci, Angelica Gualtieri, Colin Jouhanneau, Aurélie Teissandier, Carles Gaston-Massuet, Deborah Bourc'his
Format: Article
Language:English
Published: eLife Sciences Publications Ltd 2022-01-01
Series:eLife
Subjects:
Online Access:https://elifesciences.org/articles/65641
_version_ 1811227539404750848
author Juliane Glaser
Julian Iranzo
Maud Borensztein
Mattia Marinucci
Angelica Gualtieri
Colin Jouhanneau
Aurélie Teissandier
Carles Gaston-Massuet
Deborah Bourc'his
author_facet Juliane Glaser
Julian Iranzo
Maud Borensztein
Mattia Marinucci
Angelica Gualtieri
Colin Jouhanneau
Aurélie Teissandier
Carles Gaston-Massuet
Deborah Bourc'his
author_sort Juliane Glaser
collection DOAJ
description 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.
first_indexed 2024-04-12T09:44:18Z
format Article
id doaj.art-4e59277598dd4ccbbd4bcaaa22690b54
institution Directory Open Access Journal
issn 2050-084X
language English
last_indexed 2024-04-12T09:44:18Z
publishDate 2022-01-01
publisher eLife Sciences Publications Ltd
record_format Article
series eLife
spelling doaj.art-4e59277598dd4ccbbd4bcaaa22690b542022-12-22T03:37:59ZengeLife Sciences Publications LtdeLife2050-084X2022-01-011110.7554/eLife.65641The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonatesJuliane Glaser0https://orcid.org/0000-0001-6745-6924Julian Iranzo1https://orcid.org/0000-0002-9369-2530Maud Borensztein2https://orcid.org/0000-0002-4378-5018Mattia Marinucci3Angelica Gualtieri4Colin Jouhanneau5Aurélie Teissandier6Carles Gaston-Massuet7Deborah Bourc'his8https://orcid.org/0000-0001-9499-7291Institut Curie, PSL Research University, INSERM, CNRS, Paris, FranceInstitut Curie, PSL Research University, INSERM, CNRS, Paris, FranceInstitut Curie, PSL Research University, INSERM, CNRS, Paris, FranceInstitut Curie, PSL Research University, INSERM, CNRS, Paris, FranceCentre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United KingdomInstitut Curie, PSL Research University, Animal Transgenesis Platform, Paris, FranceInstitut Curie, PSL Research University, INSERM, CNRS, Paris, FranceCentre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United KingdomInstitut Curie, PSL Research University, INSERM, CNRS, Paris, FranceGenomic 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.https://elifesciences.org/articles/65641genomic imprintingdevelopmentgrowthhypothalamus
spellingShingle Juliane Glaser
Julian Iranzo
Maud Borensztein
Mattia Marinucci
Angelica Gualtieri
Colin Jouhanneau
Aurélie Teissandier
Carles Gaston-Massuet
Deborah Bourc'his
The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates
eLife
genomic imprinting
development
growth
hypothalamus
title The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates
title_full The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates
title_fullStr The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates
title_full_unstemmed The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates
title_short The imprinted Zdbf2 gene finely tunes control of feeding and growth in neonates
title_sort imprinted zdbf2 gene finely tunes control of feeding and growth in neonates
topic genomic imprinting
development
growth
hypothalamus
url https://elifesciences.org/articles/65641
work_keys_str_mv AT julianeglaser theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT julianiranzo theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT maudborensztein theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT mattiamarinucci theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT angelicagualtieri theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT colinjouhanneau theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT aurelieteissandier theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT carlesgastonmassuet theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT deborahbourchis theimprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT julianeglaser imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT julianiranzo imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT maudborensztein imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT mattiamarinucci imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT angelicagualtieri imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT colinjouhanneau imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT aurelieteissandier imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT carlesgastonmassuet imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates
AT deborahbourchis imprintedzdbf2genefinelytunescontroloffeedingandgrowthinneonates