Please use this identifier to cite or link to this item: https://hdl.handle.net/2440/132879
Citations
Scopus Web of Science® Altmetric
?
?
Type: Journal article
Title: Macrophages exert homeostatic actions in pregnancy to protect against preterm birth and fetal inflammatory injury
Author: Gomez-Lopez, N.
Garcia-Flores, V.
Chin, P.Y.
Groome, H.M.
Bijland, M.T.
Diener, K.R.
Romero, R.
Robertson, S.A.
Citation: JCI Insight, 2021; 6(19):e146089-1-e146089-22
Publisher: American Society for Clinical Investigation
Issue Date: 2021
ISSN: 2379-3708
2379-3708
Statement of
Responsibility: 
Nardhy Gomez-Lopez, Valeria Garcia-Flores, Peck Yin Chin, Holly M. Groome, Melanie T. Bijland, Kerrilyn R. Diener, Roberto Romero, and Sarah A. Robertson
Abstract: Macrophages are commonly thought to contribute to the pathophysiology of preterm labor by amplifying inflammation — but a protective role has not previously been considered to our knowledge. We hypothesized that given their antiinflammatory capability in early pregnancy, macrophages exert essential roles in maintenance of late gestation and that insufficient macrophages may predispose individuals to spontaneous preterm labor and adverse neonatal outcomes. Here, we showed that women with spontaneous preterm birth had reduced CD209⁺CD206⁺ expression in alternatively activated CD45⁺CD14⁺ICAM3⁻ macrophages and increased TNF expression in proinflammatory CD45⁺CD14⁺CD80⁺HLA-DR⁺ macrophages in the uterine decidua at the materno-fetal interface. In Cd11b(DTR/DTR) mice, depletion of maternal CD11b⁺ myeloid cells caused preterm birth, neonatal death, and postnatal growth impairment, accompanied by uterine cytokine and leukocyte changes indicative of a proinflammatory response, while adoptive transfer of WT macrophages prevented preterm birth and partially rescued neonatal loss. In a model of intra-amniotic inflammation–induced preterm birth, macrophages polarized in vitro to an M2 phenotype showed superior capacity over nonpolarized macrophages to reduce uterine and fetal inflammation, prevent preterm birth, and improve neonatal survival. We conclude that macrophages exert a critical homeostatic regulatory role in late gestation and are implicated as a determinant of susceptibility to spontaneous preterm birth and fetal inflammatory injury.
Keywords: Macrophages
Rights: © 2021, Gomez-Lopez et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.
DOI: 10.1172/jci.insight.146089
Grant ID: http://purl.org/au-research/grants/nhmrc/1140916
http://purl.org/au-research/grants/nhmrc/1012386
Published version: http://dx.doi.org/10.1172/jci.insight.146089
Appears in Collections:Medicine publications

Files in This Item:
File Description SizeFormat 
hdl_132879.pdfPublished Version8.52 MBAdobe PDFView/Open


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.