The transcription factor ERG regulates super-enhancers associated with an endothelial-specific gene expression program

V Kalna, Y Yang, CR Peghaire, K Frudd… - Circulation …, 2019 - Am Heart Assoc
V Kalna, Y Yang, CR Peghaire, K Frudd, R Hannah, AV Shah, L Osuna Almagro, JJ Boyle
Circulation research, 2019Am Heart Assoc
Rationale: The ETS (E-26 transformation-specific) transcription factor ERG (ETS-related
gene) is essential for endothelial homeostasis, driving expression of lineage genes and
repressing proinflammatory genes. Loss of ERG expression is associated with diseases
including atherosclerosis. ERG's homeostatic function is lineage-specific, because aberrant
ERG expression in cancer is oncogenic. The molecular basis for ERG lineage-specific
activity is unknown. Transcriptional regulation of lineage specificity is linked to enhancer …
Rationale:
The ETS (E-26 transformation-specific) transcription factor ERG (ETS-related gene) is essential for endothelial homeostasis, driving expression of lineage genes and repressing proinflammatory genes. Loss of ERG expression is associated with diseases including atherosclerosis. ERG’s homeostatic function is lineage-specific, because aberrant ERG expression in cancer is oncogenic. The molecular basis for ERG lineage-specific activity is unknown. Transcriptional regulation of lineage specificity is linked to enhancer clusters (super-enhancers).
Objective:
To investigate whether ERG regulates endothelial-specific gene expression via super-enhancers.
Methods and Results:
Chromatin immunoprecipitation with high-throughput sequencing in human umbilical vein endothelial cells showed that ERG binds 93% of super-enhancers ranked according to H3K27ac, a mark of active chromatin. These were associated with endothelial genes such as DLL4 (Delta-like protein 4), CLDN5 (claudin-5), VWF (von Willebrand factor), and CDH5 (VE-cadherin). Comparison between human umbilical vein endothelial cell and prostate cancer TMPRSS2 (transmembrane protease, serine-2):ERG fusion-positive human prostate epithelial cancer cell line (VCaP) cells revealed distinctive lineage-specific transcriptome and super-enhancer profiles. At a subset of endothelial super-enhancers (including DLL4 and CLDN5), loss of ERG results in significant reduction in gene expression which correlates with decreased enrichment of H3K27ac and MED (Mediator complex subunit)-1, and reduced recruitment of acetyltransferase p300. At these super-enhancers, co-occupancy of GATA2 (GATA-binding protein 2) and AP-1 (activator protein 1) is significantly lower compared with super-enhancers that remained constant following ERG inhibition. These data suggest distinct mechanisms of super-enhancer regulation in endothelial cells and highlight the unique role of ERG in controlling a core subset of super-enhancers. Most disease-associated single nucleotide polymorphisms from genome-wide association studies lie within noncoding regions and perturb transcription factor recognition sequences in relevant cell types. Analysis of genome-wide association studies data shows significant enrichment of risk variants for cardiovascular disease and other diseases, at ERG endothelial enhancers and super-enhancers.
Conclusions:
The transcription factor ERG promotes endothelial homeostasis via regulation of lineage-specific enhancers and super-enhancers. Enrichment of cardiovascular disease-associated single nucleotide polymorphisms at ERG super-enhancers suggests that ERG-dependent transcription modulates disease risk.
Am Heart Assoc