Frontiers in Cardiovascular Medicine (Jun 2022)

Deciphering Pro-angiogenic Transcription Factor Profiles in Hypoxic Human Endothelial Cells by Combined Bioinformatics and in vitro Modeling

  • Arne Schmidt,
  • Arne Schmidt,
  • Arne Schmidt,
  • Maximilian Fuchs,
  • Maximilian Fuchs,
  • Stevan D. Stojanović,
  • Stevan D. Stojanović,
  • Chunguang Liang,
  • Kevin Schmidt,
  • Kevin Schmidt,
  • Kevin Schmidt,
  • Mira Jung,
  • Ke Xiao,
  • Ke Xiao,
  • Ke Xiao,
  • Jan Weusthoff,
  • Jan Weusthoff,
  • Jan Weusthoff,
  • Annette Just,
  • Angelika Pfanne,
  • Jörg H. W. Distler,
  • Thomas Dandekar,
  • Jan Fiedler,
  • Jan Fiedler,
  • Jan Fiedler,
  • Thomas Thum,
  • Thomas Thum,
  • Thomas Thum,
  • Meik Kunz,
  • Meik Kunz,
  • Meik Kunz

DOI
https://doi.org/10.3389/fcvm.2022.877450
Journal volume & issue
Vol. 9

Abstract

Read online

BackgroundConstant supply of oxygen is crucial for multicellular tissue homeostasis and energy metabolism in cardiac tissue. As a first response to acute hypoxia, endothelial cells (ECs) promote recruitment and adherence of immune cells to the dysbalanced EC barrier by releasing inflammatory mediators and growth factors, whereas chronic hypoxia leads to the activation of a transcription factor (TF) battery, that potently induces expression of growth factors and cytokines including platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF). We report a hypoxia-minded, targeted bioinformatics approach aiming to identify and validate TFs that regulate angiogenic signaling.ResultsA comprehensive RNA-Seq dataset derived from human ECs subjected to normoxic or hypoxic conditions was selected to identify significantly regulated genes based on (i) fold change (normoxia vs. hypoxia) and (ii) relative abundancy. Transcriptional regulation of this gene set was confirmed via qPCR in validation experiments where HUVECs were subjected to hypoxic conditions for 24 h. Screening the promoter and upstream regulatory elements of these genes identified two TFs, KLF5 and SP1, both with a potential binding site within these regions of selected target genes. In vitro, siRNA experiments confirmed SP1- and KLF5-mediated regulation of identified hypoxia-sensitive endothelial genes. Next to angiogenic signaling, we also validated the impact of TFs on inflammatory signaling, both key events in hypoxic sensing. Both TFs impacted on inflammatory signaling since endogenous repression led to increased NF-κB signaling. Additionally, SP1 silencing eventuated decreased angiogenic properties in terms of proliferation and tube formation.ConclusionBy detailed in silico analysis of promoter region and upstream regulatory elements for a list of hypoxia-sensitive genes, our bioinformatics approach identified putative binding sites for TFs of SP or KLF family in vitro. This strategy helped to identify TFs functionally involved in human angiogenic signaling and therefore serves as a base for identifying novel RNA-based drug entities in a therapeutic setting of vascularization.

Keywords