The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination

Preterm birth (PTB) represents 15 million births every year worldwide and is frequently associated with maternal/fetal infections and inflammation, inducing neuroinflammation. This neuroinflammation is mediated by microglial cells, which are brain-resident macrophages that release cytotoxic molecule...

Full description

Bibliographic Details
Main Authors: Cécile Morin, David Guenoun, Irvin Sautet, Valérie Faivre, Zsolt Csaba, Leslie Schwendimann, Pierrette Young-Ten, Juliette Van Steenwinckel, Pierre Gressens, Cindy Bokobza
Format: Article
Language:English
Published: MDPI AG 2022-04-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/23/9/4867
_version_ 1797504511269404672
author Cécile Morin
David Guenoun
Irvin Sautet
Valérie Faivre
Zsolt Csaba
Leslie Schwendimann
Pierrette Young-Ten
Juliette Van Steenwinckel
Pierre Gressens
Cindy Bokobza
author_facet Cécile Morin
David Guenoun
Irvin Sautet
Valérie Faivre
Zsolt Csaba
Leslie Schwendimann
Pierrette Young-Ten
Juliette Van Steenwinckel
Pierre Gressens
Cindy Bokobza
author_sort Cécile Morin
collection DOAJ
description Preterm birth (PTB) represents 15 million births every year worldwide and is frequently associated with maternal/fetal infections and inflammation, inducing neuroinflammation. This neuroinflammation is mediated by microglial cells, which are brain-resident macrophages that release cytotoxic molecules that block oligodendrocyte differentiation, leading to hypomyelination. Some preterm survivors can face lifetime motor and/or cognitive disabilities linked to periventricular white matter injuries (PWMIs). There is currently no recommendation concerning the mode of delivery in the case of PTB and its impact on brain development. Many animal models of induced-PTB based on LPS injections exist, but with a low survival rate. There is a lack of information regarding clinically used pharmacological substances to induce PTB and their consequences on brain development. Mifepristone (RU-486) is a drug used clinically to induce preterm labor. This study aims to elaborate and characterize a new model of induced-PTB and PWMIs by the gestational injection of RU-486 and the perinatal injection of pups with IL-1beta. A RU-486 single subcutaneous (s.c.) injection at embryonic day (E)18.5 induced PTB at E19.5 in pregnant OF1 mice. All pups were born alive and were adopted directly after birth. IL-1beta was injected intraperitoneally from postnatal day (P)1 to P5. Animals exposed to both RU-486 and IL-1beta demonstrated microglial reactivity and subsequent PWMIs. In conclusion, the s.c. administration of RU-486 induced labor within 24 h with a high survival rate for pups. In the context of perinatal inflammation, RU-486 labor induction significantly decreases microglial reactivity in vivo but did not prevent subsequent PWMIs.
first_indexed 2024-03-10T04:05:37Z
format Article
id doaj.art-aeaea988e1b94f61a8afad11c8971349
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-10T04:05:37Z
publishDate 2022-04-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-aeaea988e1b94f61a8afad11c89713492023-11-23T08:23:26ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672022-04-01239486710.3390/ijms23094867The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent HypomyelinationCécile Morin0David Guenoun1Irvin Sautet2Valérie Faivre3Zsolt Csaba4Leslie Schwendimann5Pierrette Young-Ten6Juliette Van Steenwinckel7Pierre Gressens8Cindy Bokobza9NeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FranceNeuroDiderot, Inserm UMR-1141, Hôpital Robert Debré, Université de Paris, 75019 Paris, FrancePreterm birth (PTB) represents 15 million births every year worldwide and is frequently associated with maternal/fetal infections and inflammation, inducing neuroinflammation. This neuroinflammation is mediated by microglial cells, which are brain-resident macrophages that release cytotoxic molecules that block oligodendrocyte differentiation, leading to hypomyelination. Some preterm survivors can face lifetime motor and/or cognitive disabilities linked to periventricular white matter injuries (PWMIs). There is currently no recommendation concerning the mode of delivery in the case of PTB and its impact on brain development. Many animal models of induced-PTB based on LPS injections exist, but with a low survival rate. There is a lack of information regarding clinically used pharmacological substances to induce PTB and their consequences on brain development. Mifepristone (RU-486) is a drug used clinically to induce preterm labor. This study aims to elaborate and characterize a new model of induced-PTB and PWMIs by the gestational injection of RU-486 and the perinatal injection of pups with IL-1beta. A RU-486 single subcutaneous (s.c.) injection at embryonic day (E)18.5 induced PTB at E19.5 in pregnant OF1 mice. All pups were born alive and were adopted directly after birth. IL-1beta was injected intraperitoneally from postnatal day (P)1 to P5. Animals exposed to both RU-486 and IL-1beta demonstrated microglial reactivity and subsequent PWMIs. In conclusion, the s.c. administration of RU-486 induced labor within 24 h with a high survival rate for pups. In the context of perinatal inflammation, RU-486 labor induction significantly decreases microglial reactivity in vivo but did not prevent subsequent PWMIs.https://www.mdpi.com/1422-0067/23/9/4867preterm birthinduced labormifepristoneRU-486microgliaoligodendrocyte maturation
spellingShingle Cécile Morin
David Guenoun
Irvin Sautet
Valérie Faivre
Zsolt Csaba
Leslie Schwendimann
Pierrette Young-Ten
Juliette Van Steenwinckel
Pierre Gressens
Cindy Bokobza
The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination
International Journal of Molecular Sciences
preterm birth
induced labor
mifepristone
RU-486
microglia
oligodendrocyte maturation
title The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination
title_full The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination
title_fullStr The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination
title_full_unstemmed The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination
title_short The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination
title_sort impact of mouse preterm birth induction by ru 486 on microglial activation and subsequent hypomyelination
topic preterm birth
induced labor
mifepristone
RU-486
microglia
oligodendrocyte maturation
url https://www.mdpi.com/1422-0067/23/9/4867
work_keys_str_mv AT cecilemorin theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT davidguenoun theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT irvinsautet theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT valeriefaivre theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT zsoltcsaba theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT leslieschwendimann theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT pierretteyoungten theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT juliettevansteenwinckel theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT pierregressens theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT cindybokobza theimpactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT cecilemorin impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT davidguenoun impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT irvinsautet impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT valeriefaivre impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT zsoltcsaba impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT leslieschwendimann impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT pierretteyoungten impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT juliettevansteenwinckel impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT pierregressens impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination
AT cindybokobza impactofmousepretermbirthinductionbyru486onmicroglialactivationandsubsequenthypomyelination