Convergence of hypoxia and TGFß pathways on cell cycle regulation in human hematopoietic stem/progenitor cells

Authors:
Wierenga AT, Vellenga E, Schuringa JJ
In:
Source: PLoS ONE
Publication Date: (2014)
Issue: 9(3): e93494
Research Area:
Immunotherapy / Hematology
Basic Research
Cells used in publication:
UT7
Species: human
Tissue Origin: blood
CD34+ cell, human
Species: human
Tissue Origin: blood
OCI-AML3
Species: human
Tissue Origin: blood
Mononuclear, cord blood, human
Species: human
Tissue Origin: blood
Abstract
Although it has been shown that HIF1 and 2 fulfill essential roles within the hematopoietic system and in the regulation of HSC fate, little is currently known about the specific mechanisms that are involved. We identified transcriptome changes induced by hypoxia, constitutively active HIF1(P402/564) and HIF2(P405/531) in human cord blood CD34+ cells. Thus, we were able to identify common hypoxia-HIF1-HIF2 gene signatures, but we also identified specific target genes that were exclusively regulated by HIF1, HIF2 or hypoxia. Geneset enrichment analysis (GSEA) revealed that, besides known pathways associated with "hypoxia-induced signaling", also significant enrichment for the Transforming Growth Factor beta (TGFß) pathway was observed within the hypoxia/HIF1/HIF2 transcriptomes. One of the most significantly upregulated genes in both gene sets was the cyclin dependent kinase inhibitor CDKN1C (p57kip2). Combined hypoxia treatment or HIF overexpression together with TGFß stimulation resulted in enhanced expression of CDKN1C and enhanced cell cycle arrest within the CD34+/CD38- stem cell compartment. Interestingly, we observed that CD34+ cells cultured under hypoxic conditions secreted high levels of latent TGFß, suggesting an auto- or paracrine role of TGFß in the regulation of quiescence of these cells. However, knockdown of SMAD4 could not rescue the hypoxia induced cell cycle arrest, arguing against direct effects of hypoxia-induced secreted TGFß. Finally, the Ga-coupled receptor GTPase RGS1 was identified as a HIF-dependent hypoxia target that dampens SDF1-induced migration and signal transduction in human CD34+ stem/progenitor cells.