[HTML][HTML] Faulty oxygen sensing disrupts angiomotin function in trophoblast cell migration and predisposes to preeclampsia

A Farrell, S Alahari, L Ermini, A Tagliaferro, M Litvack… - JCI insight, 2019 - ncbi.nlm.nih.gov
A Farrell, S Alahari, L Ermini, A Tagliaferro, M Litvack, M Post, I Caniggia
JCI insight, 2019ncbi.nlm.nih.gov
Human placenta development and a successful pregnancy is incumbent upon precise
oxygen-dependent control of trophoblast migration/invasion. Persistent low oxygen leading
to failed trophoblast invasion promotes inadequate spiral artery remodeling, a characteristic
of preeclampsia. Angiomotin (AMOT) is a multifaceted scaffolding protein involved in cell
polarity and migration, yet its upstream regulation and significance in the human placenta
remain unknown. Herein, we show that AMOT is primarily expressed in migratory …
Abstract
Human placenta development and a successful pregnancy is incumbent upon precise oxygen-dependent control of trophoblast migration/invasion. Persistent low oxygen leading to failed trophoblast invasion promotes inadequate spiral artery remodeling, a characteristic of preeclampsia. Angiomotin (AMOT) is a multifaceted scaffolding protein involved in cell polarity and migration, yet its upstream regulation and significance in the human placenta remain unknown. Herein, we show that AMOT is primarily expressed in migratory extravillous trophoblast cells (EVTs) of the intermediate and distal anchoring column. Its expression increases after 10 weeks of gestation when oxygen tension rises and EVT migration/invasion peaks. Time-lapse imaging confirmed that the AMOT 80-kDa isoform promotes migration of trophoblastic JEG3 and HTR-8/SVneo cells. In preeclampsia, however, AMOT expression is decreased and its localization to migratory fetomaternal interface EVTs is disrupted. We demonstrate that Jumonji C domain–containing protein 6 (JMJD6), an oxygen sensor, positively regulates AMOT via oxygen-dependent lysyl hydroxylation. Furthermore, in vitro and ex vivo studies show that transforming growth factor-β (TGF-β) regulates AMOT expression, its interaction with polarity protein PAR6, and its subcellular redistribution from tight junctions to cytoskeleton. Our data reveal an oxygen-and TGF-β–driven migratory function for AMOT in the human placenta, and implicate its deficiency in impaired trophoblast migration that plagues preeclampsia.
ncbi.nlm.nih.gov