[HTML][HTML] Loss of FOXF1 expression promotes human lung-resident mesenchymal stromal cell migration via ATX/LPA/LPA1 signaling axis

P Cao, NM Walker, RR Braeuer, S Mazzoni-Putman… - Scientific Reports, 2020 - nature.com
P Cao, NM Walker, RR Braeuer, S Mazzoni-Putman, Y Aoki, K Misumi, DS Wheeler, R Vittal…
Scientific Reports, 2020nature.com
Abstract Forkhead box F1 (FOXF1) is a lung embryonic mesenchyme-associated
transcription factor that demonstrates persistent expression into adulthood in mesenchymal
stromal cells. However, its biologic function in human adult lung-resident mesenchymal
stromal cells (LR-MSCs) remain to be elucidated. Here, we demonstrate that FOXF1
expression acts as a restraint on the migratory function of LR-MSCs via its role as a novel
transcriptional repressor of autocrine motility-stimulating factor Autotaxin (ATX). Fibrotic …
Abstract
Forkhead box F1 (FOXF1) is a lung embryonic mesenchyme-associated transcription factor that demonstrates persistent expression into adulthood in mesenchymal stromal cells. However, its biologic function in human adult lung-resident mesenchymal stromal cells (LR-MSCs) remain to be elucidated. Here, we demonstrate that FOXF1 expression acts as a restraint on the migratory function of LR-MSCs via its role as a novel transcriptional repressor of autocrine motility-stimulating factor Autotaxin (ATX). Fibrotic human LR-MSCs demonstrated lower expression of FOXF1 mRNA and protein, compared to non-fibrotic controls. RNAi-mediated FOXF1 silencing in LR-MSCs was associated with upregulation of key genes regulating proliferation, migration, and inflammatory responses and significantly higher migration were confirmed in FOXF1-silenced LR-MSCs by Boyden chamber. ATX is a secreted lysophospholipase D largely responsible for extracellular lysophosphatidic acid (LPA) production, and was among the top ten upregulated genes upon Affymetrix analysis. FOXF1-silenced LR-MSCs demonstrated increased ATX activity, while mFoxf1 overexpression diminished ATX expression and activity. The FOXF1 silencing-induced increase in LR-MSC migration was abrogated by genetic and pharmacologic targeting of ATX and LPA1 receptor. Chromatin immunoprecipitation analyses identified three putative FOXF1 binding sites in the 1.5 kb ATX promoter which demonstrated transcriptional repression of ATX expression. Together these findings identify FOXF1 as a novel transcriptional repressor of ATX and demonstrate that loss of FOXF1 promotes LR-MSC migration via the ATX/LPA/LPA1 signaling axis.
nature.com