File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Human placental exosomes induce maternal systemic immune tolerance by reprogramming circulating monocytes

TitleHuman placental exosomes induce maternal systemic immune tolerance by reprogramming circulating monocytes
Authors
KeywordsMaternal immune tolerance
Monocyte
PD-L1
Placental exosome
PTEN
T cell
Issue Date2022
Citation
Journal of Nanobiotechnology, 2022, v. 20 n. 1, article no. 86 How to Cite?
AbstractBackground: The maternal immune system needs to tolerate the semi-allogeneic fetus in pregnancy. The adaptation occurs locally at the maternal–fetal interface as well as systemically through the maternal circulation. Failure to tolerate the paternal antigens may result in pregnancy complications, such as pregnancy loss and pre-eclampsia. However, the mechanism that regulates maternal immune tolerance, especially at the systemic level, is still an enigma. Here we report that the first-trimester placenta-derived exosomes (pEXOs) contribute to maternal immune tolerance by reprogramming the circulating monocytes. Results: pEXOs predominantly target monocytes and pEXO-educated monocytes exhibit an immunosuppressive phenotype as demonstrated by reduced expression of marker genes for monocyte activation, T-cell activation and antigen-process/presentation at the transcriptomic level. They also have a greater propensity towards M2 polarization when compared to the monocytes without pEXO treatment. The inclusion of pEXOs in a monocyte-T-cell coculture model significantly reduces proliferation of the T helper cells and cytotoxic T cells and elevates the expansion of regulatory T cells. By integrating the microRNAome of pEXO and the transcriptomes of pEXO-educated monocytes as well as various immune cell functional assays, we demonstrate that the pEXO-derived microRNA miR-29a-3p promotes the expression of programmed cell death ligand-1, a well-known surface receptor that suppresses the adaptive immune system, by down-regulation of phosphatase and tensin homolog in monocytes. Conclusions: This is the first report to show how human pEXO directly regulates monocyte functions and its molecular mechanism during early pregnancy. The results uncover the importance of pEXO in regulating the maternal systemic immune response during early pregnancy by reprogramming circulating monocytes. The study provides the basis for understanding the regulation of maternal immune tolerance to the fetal allograft.
Persistent Identifierhttp://hdl.handle.net/10722/313216
PubMed Central ID
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorBAI, K-
dc.contributor.authorLee, CL-
dc.contributor.authorLiu, X-
dc.contributor.authorLI, J-
dc.contributor.authorCao, D-
dc.contributor.authorZhang, L-
dc.contributor.authorHu, D-
dc.contributor.authorLi, H-
dc.contributor.authorHou, Y-
dc.contributor.authorXu, Y-
dc.contributor.authorKan, SYA-
dc.contributor.authorCheung, KW-
dc.contributor.authorNg, EHY-
dc.contributor.authorYeung, WSB-
dc.contributor.authorChiu, CN-
dc.date.accessioned2022-06-06T05:47:45Z-
dc.date.available2022-06-06T05:47:45Z-
dc.date.issued2022-
dc.identifier.citationJournal of Nanobiotechnology, 2022, v. 20 n. 1, article no. 86-
dc.identifier.urihttp://hdl.handle.net/10722/313216-
dc.description.abstractBackground: The maternal immune system needs to tolerate the semi-allogeneic fetus in pregnancy. The adaptation occurs locally at the maternal–fetal interface as well as systemically through the maternal circulation. Failure to tolerate the paternal antigens may result in pregnancy complications, such as pregnancy loss and pre-eclampsia. However, the mechanism that regulates maternal immune tolerance, especially at the systemic level, is still an enigma. Here we report that the first-trimester placenta-derived exosomes (pEXOs) contribute to maternal immune tolerance by reprogramming the circulating monocytes. Results: pEXOs predominantly target monocytes and pEXO-educated monocytes exhibit an immunosuppressive phenotype as demonstrated by reduced expression of marker genes for monocyte activation, T-cell activation and antigen-process/presentation at the transcriptomic level. They also have a greater propensity towards M2 polarization when compared to the monocytes without pEXO treatment. The inclusion of pEXOs in a monocyte-T-cell coculture model significantly reduces proliferation of the T helper cells and cytotoxic T cells and elevates the expansion of regulatory T cells. By integrating the microRNAome of pEXO and the transcriptomes of pEXO-educated monocytes as well as various immune cell functional assays, we demonstrate that the pEXO-derived microRNA miR-29a-3p promotes the expression of programmed cell death ligand-1, a well-known surface receptor that suppresses the adaptive immune system, by down-regulation of phosphatase and tensin homolog in monocytes. Conclusions: This is the first report to show how human pEXO directly regulates monocyte functions and its molecular mechanism during early pregnancy. The results uncover the importance of pEXO in regulating the maternal systemic immune response during early pregnancy by reprogramming circulating monocytes. The study provides the basis for understanding the regulation of maternal immune tolerance to the fetal allograft.-
dc.languageeng-
dc.relation.ispartofJournal of Nanobiotechnology-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectMaternal immune tolerance-
dc.subjectMonocyte-
dc.subjectPD-L1-
dc.subjectPlacental exosome-
dc.subjectPTEN-
dc.subjectT cell-
dc.titleHuman placental exosomes induce maternal systemic immune tolerance by reprogramming circulating monocytes-
dc.typeArticle-
dc.identifier.emailLee, CL: kcllee@hku.hk-
dc.identifier.emailNg, EHY: nghye@hku.hk-
dc.identifier.emailYeung, WSB: wsbyeung@hku.hk-
dc.identifier.emailChiu, CN: pchiucn@hku.hk-
dc.identifier.authorityLee, CL=rp02515-
dc.identifier.authorityNg, EHY=rp00426-
dc.identifier.authorityYeung, WSB=rp00331-
dc.identifier.authorityChiu, CN=rp00424-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.1186/s12951-022-01283-2-
dc.identifier.pmid35180876-
dc.identifier.pmcidPMC8857816-
dc.identifier.scopuseid_2-s2.0-85124933917-
dc.identifier.hkuros333236-
dc.identifier.volume20-
dc.identifier.issue1-
dc.identifier.spagearticle no. 86-
dc.identifier.epagearticle no. 86-
dc.identifier.isiWOS:000757835500001-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats