Abstract

Vascular adhesion protein-1 (VAP-1) is an inflammation-inducible adhesion molecule and a primary amine oxidase involved in immune cell trafficking. Leukocyte extravasation into tissues is mediated by adhesion molecules expressed on endothelial cells and pericytes. Pericytes play a major role in the angiogenesis and vascularization of cycling endometrium. However, the functional properties of pericytes in the human endometrium are not known. Here we show that pericytes surrounding the spiral arterioles in midluteal human endometrium constitutively express VAP-1. We first characterize these pericytes and demonstrate that knockdown of VAP-1 perturbed their biophysical properties and compromised their contractile, migratory, adhesive and clonogenic capacities. Furthermore, we show that loss of VAP-1 disrupts pericyte-uterine natural killer cell interactions in vitro. Taken together, the data not only reveal that endometrial pericytes represent a cell population with distinct biophysical and functional properties but also suggest a pivotal role for VAP-1 in regulating the recruitment of innate immune cells in human endometrium. We posit that VAP-1 could serve as a potential biomarker for pregnancy pathologies caused by a compromised perivascular environment prior to conception.

Highlights

  • The human endometrium undergoes iterative cycles of growth, differentiation and shedding during the reproductive years

  • This study focuses on Vascular adhesion protein-1 (VAP-1) and its relationship with the biophysical properties of endometrial pericytes

  • We report that VAP-1 is highly expressed on the pericytes around the spiral arterioles in the human endometrium

Read more

Summary

Introduction

The human endometrium undergoes iterative cycles of growth, differentiation and shedding during the reproductive years. This highly dynamic tissue is dependent on mesenchymal stem/progenitor cells (MSCs) that bestow its regeneration capacity during each menstrual cycle (Gargett et al, 2015). During the midluteal window of implantation, SUSD2+ cells reside around the emerging spiral arterioles and are characterized by the expression of genes encoding prototypic pericyte markers, including PDGFRB, CD146, neural/glial antigen 2 (CSPG4), and α-smooth muscle actin (ACTA2) (Armulik et al, 2011; Murakami et al, 2014; Ferland-Mccollough et al, 2017). Compared to (SUSD2−) endometrial stromal cells (EnSCs), the SUSD2+ cell fraction is enriched in clonal MSCs (Murakami et al, 2013; Lucas et al, 2016), indicating that they are integral to the perivascular stem cell niche in human endometrium (Santamaria et al, 2018)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call