↓ Skip to main content

PLOS

Convergence of Hypoxia and TGFβ Pathways on Cell Cycle Regulation in Human Hematopoietic Stem/Progenitor Cells

Overview of attention for article published in PLOS ONE, March 2014
Altmetric Badge

Mentioned by

blogs
1 blog
twitter
1 X user

Citations

dimensions_citation
48 Dimensions

Readers on

mendeley
65 Mendeley
Title
Convergence of Hypoxia and TGFβ Pathways on Cell Cycle Regulation in Human Hematopoietic Stem/Progenitor Cells
Published in
PLOS ONE, March 2014
DOI 10.1371/journal.pone.0093494
Pubmed ID
Authors

Albertus T. J. Wierenga, Edo Vellenga, Jan Jacob Schuringa

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 (TGFβ) 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 TGFβ 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 TGFβ, suggesting an auto- or paracrine role of TGFβ 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 TGFβ. Finally, the Gα-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.

X Demographics

X Demographics

The data shown below were collected from the profile of 1 X user who shared this research output. Click here to find out more about how the information was compiled.
Mendeley readers

Mendeley readers

The data shown below were compiled from readership statistics for 65 Mendeley readers of this research output. Click here to see the associated Mendeley record.

Geographical breakdown

Country Count As %
Netherlands 1 2%
Italy 1 2%
Belgium 1 2%
Unknown 62 95%

Demographic breakdown

Readers by professional status Count As %
Student > Ph. D. Student 13 20%
Researcher 11 17%
Student > Bachelor 7 11%
Student > Master 6 9%
Student > Doctoral Student 4 6%
Other 7 11%
Unknown 17 26%
Readers by discipline Count As %
Agricultural and Biological Sciences 16 25%
Biochemistry, Genetics and Molecular Biology 15 23%
Medicine and Dentistry 5 8%
Engineering 3 5%
Mathematics 2 3%
Other 5 8%
Unknown 19 29%