Cargando…

Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models

Obesity is prevalent among women of reproductive age and is associated with increased risk of developing multiple pregnancy disorders. Pregnancy must induce immune tolerance to avoid fetal rejection, while obesity can cause chronic inflammation through activating the immune system. Impaired maternal...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Yanhong, Chen, Jiajia, Lin, Yikong, Xu, Ling, Sang, Yifei, Li, Dajin, Du, Meirong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8219966/
https://www.ncbi.nlm.nih.gov/pubmed/34177956
http://dx.doi.org/10.3389/fimmu.2021.694077
_version_ 1783711054962884608
author Li, Yanhong
Chen, Jiajia
Lin, Yikong
Xu, Ling
Sang, Yifei
Li, Dajin
Du, Meirong
author_facet Li, Yanhong
Chen, Jiajia
Lin, Yikong
Xu, Ling
Sang, Yifei
Li, Dajin
Du, Meirong
author_sort Li, Yanhong
collection PubMed
description Obesity is prevalent among women of reproductive age and is associated with increased risk of developing multiple pregnancy disorders. Pregnancy must induce immune tolerance to avoid fetal rejection, while obesity can cause chronic inflammation through activating the immune system. Impaired maternal immuno-tolerance leads to pregnancy failure, such as recurrent spontaneous abortion (RSA), one of the most common complications during early pregnancy. How does maternal immune response change under obesity stress in normal pregnancy and RSA? In turn, is obesity affected by different gestational statuses? Limited information is presently available now. Our study investigated pregnancy outcomes and maternal immune responses in two murine models (normal pregnancy and spontaneous abortion models) after obesity challenge with a high-fat diet (HFD). Abortion-prone mice fed HFD had significantly higher weight gains during pregnancy than normal pregnant mice with HFD feeding. Nonetheless, the embryo implantation and resorption rates were comparable between HFD and normal chow diet (NCD)-fed mice in each model. Evaluation of immune cell subsets showed HFD-induced obesity drove the upregulation of activated NK cell-activating receptor (NKp46)(+) NK cells and pro-inflammatory macrophages (MHCII(high) Mφ) as well as CD4(+) and CD8(+) T cells in the normal pregnancy group. However, in the abortion-prone group, relative more immature NK cells with decreased activity phenotypes were found in obese mice. Moreover, there were increased DCreg (CD11b(high) DC) cells and decreased CD4(+) and CD8(+) T cells detected in the HFD abortion-prone mice relative to those fed the NCD diet. Our findings reveal how pregnancy obesity and maternal immune regulation are mutually influenced. It is worth noting that the abortion-prone model where active maternal immune status was intensified by obesity, in turn stimulated an overcompensation response, leading to an over-tolerized immune status, and predisposing to potential risks of perinatal complications.
format Online
Article
Text
id pubmed-8219966
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-82199662021-06-24 Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models Li, Yanhong Chen, Jiajia Lin, Yikong Xu, Ling Sang, Yifei Li, Dajin Du, Meirong Front Immunol Immunology Obesity is prevalent among women of reproductive age and is associated with increased risk of developing multiple pregnancy disorders. Pregnancy must induce immune tolerance to avoid fetal rejection, while obesity can cause chronic inflammation through activating the immune system. Impaired maternal immuno-tolerance leads to pregnancy failure, such as recurrent spontaneous abortion (RSA), one of the most common complications during early pregnancy. How does maternal immune response change under obesity stress in normal pregnancy and RSA? In turn, is obesity affected by different gestational statuses? Limited information is presently available now. Our study investigated pregnancy outcomes and maternal immune responses in two murine models (normal pregnancy and spontaneous abortion models) after obesity challenge with a high-fat diet (HFD). Abortion-prone mice fed HFD had significantly higher weight gains during pregnancy than normal pregnant mice with HFD feeding. Nonetheless, the embryo implantation and resorption rates were comparable between HFD and normal chow diet (NCD)-fed mice in each model. Evaluation of immune cell subsets showed HFD-induced obesity drove the upregulation of activated NK cell-activating receptor (NKp46)(+) NK cells and pro-inflammatory macrophages (MHCII(high) Mφ) as well as CD4(+) and CD8(+) T cells in the normal pregnancy group. However, in the abortion-prone group, relative more immature NK cells with decreased activity phenotypes were found in obese mice. Moreover, there were increased DCreg (CD11b(high) DC) cells and decreased CD4(+) and CD8(+) T cells detected in the HFD abortion-prone mice relative to those fed the NCD diet. Our findings reveal how pregnancy obesity and maternal immune regulation are mutually influenced. It is worth noting that the abortion-prone model where active maternal immune status was intensified by obesity, in turn stimulated an overcompensation response, leading to an over-tolerized immune status, and predisposing to potential risks of perinatal complications. Frontiers Media S.A. 2021-06-09 /pmc/articles/PMC8219966/ /pubmed/34177956 http://dx.doi.org/10.3389/fimmu.2021.694077 Text en Copyright © 2021 Li, Chen, Lin, Xu, Sang, Li and Du https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Li, Yanhong
Chen, Jiajia
Lin, Yikong
Xu, Ling
Sang, Yifei
Li, Dajin
Du, Meirong
Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models
title Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models
title_full Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models
title_fullStr Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models
title_full_unstemmed Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models
title_short Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models
title_sort obesity challenge drives distinct maternal immune response changes in normal pregnant and abortion-prone mouse models
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8219966/
https://www.ncbi.nlm.nih.gov/pubmed/34177956
http://dx.doi.org/10.3389/fimmu.2021.694077
work_keys_str_mv AT liyanhong obesitychallengedrivesdistinctmaternalimmuneresponsechangesinnormalpregnantandabortionpronemousemodels
AT chenjiajia obesitychallengedrivesdistinctmaternalimmuneresponsechangesinnormalpregnantandabortionpronemousemodels
AT linyikong obesitychallengedrivesdistinctmaternalimmuneresponsechangesinnormalpregnantandabortionpronemousemodels
AT xuling obesitychallengedrivesdistinctmaternalimmuneresponsechangesinnormalpregnantandabortionpronemousemodels
AT sangyifei obesitychallengedrivesdistinctmaternalimmuneresponsechangesinnormalpregnantandabortionpronemousemodels
AT lidajin obesitychallengedrivesdistinctmaternalimmuneresponsechangesinnormalpregnantandabortionpronemousemodels
AT dumeirong obesitychallengedrivesdistinctmaternalimmuneresponsechangesinnormalpregnantandabortionpronemousemodels