Hypoxia inducible factors regulate pluripotency and proliferation in human embryonic stem cells cultured at reduced oxygen tensions

Human embryonic stem (hES) cells are routinely cultured under atmospheric, 20% oxygen tensions but are derived from embryos which reside in a 3–5% oxygen (hypoxic) environment. Maintenance of oxygen homeostasis is critical to ensure sufficient levels for oxygen-dependent processes. This study investigates the importance of specific hypoxia inducible factors (HIFs) in regulating the hypoxic responses of hES cells. We report that culture at 20% oxygen decreased hES cell proliferation and resulted in a significantly reduced expression of SOX2, NANOG and POU5F1 (OCT4) mRNA as well as POU5F1 protein compared with hypoxic conditions. HIF1A protein was not expressed at 20% oxygen and displayed only a transient, nuclear localisation at 5% oxygen. HIF2A (EPAS1) and HIF3A displayed a cytoplasmic localisation during initial hypoxic culture but translocated to the nucleus following long-term culture at 5% oxygen and were significantly upregulated compared with cells cultured at 20% oxygen. Silencing of HIF2A resulted in a significant decrease in both hES cell proliferation and POU5F1, SOX2 and NANOG protein expression while the early differentiation marker, SSEA1, was concomitantly increased. HIF3A upregulated HIF2A and prevented HIF1A expression with the knockdown of HIF3A resulting in the reappearance of HIF1A protein. In summary, these data demonstrate that a low oxygen tension is preferential for the maintenance of a highly proliferative, pluripotent population of hES cells. While HIF3A was found to regulate the expression of both HIF1A and HIF2A, it is HIF2A which regulates hES cell pluripotency as well as proliferation under hypoxic conditions.

[1]  L. Chodosh,et al.  Differential Regulation of the Transcriptional Activities of Hypoxia-Inducible Factor 1 Alpha (HIF-1α) and HIF-2α in Stem Cells , 2006, Molecular and Cellular Biology.

[2]  R. Conaway,et al.  Multiple Splice Variants of the Human HIF-3α Locus Are Targets of the von Hippel-Lindau E3 Ubiquitin Ligase Complex* , 2003, The Journal of Biological Chemistry.

[3]  Brian Keith,et al.  Differential Roles of Hypoxia-Inducible Factor 1α (HIF-1α) and HIF-2α in Hypoxic Gene Regulation , 2003, Molecular and Cellular Biology.

[4]  M. M. Bradford A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. , 1976, Analytical biochemistry.

[5]  A. Caplan,et al.  Cultivation of rat marrow‐derived mesenchymal stem cells in reduced oxygen tension: Effects on in vitro and in vivo osteochondrogenesis , 2001, Journal of cellular physiology.

[6]  M. Ivan,et al.  HIFα Targeted for VHL-Mediated Destruction by Proline Hydroxylation: Implications for O2 Sensing , 2001, Science.

[7]  D. Kaufman,et al.  Activation of Hypoxic Response in Human Embryonic Stem Cell–Derived Embryoid Bodies , 2008, Experimental biology and medicine.

[8]  R. Roberts,et al.  Low O2 tensions and the prevention of differentiation of hES cells. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[9]  E. Coonen,et al.  Effect of oxygen concentration on human in-vitro fertilization and embryo culture. , 1999, Human reproduction.

[10]  P. Ratcliffe,et al.  Independent function of two destruction domains in hypoxia‐inducible factor‐α chains activated by prolyl hydroxylation , 2001, The EMBO journal.

[11]  H. Schöler,et al.  Octamer binding proteins confer transcriptional activity in early mouse embryogenesis. , 1989, The EMBO journal.

[12]  R. Nesbitt,et al.  Acta obstetrica et gynecologica scandinavica , 1962 .

[13]  J. Thomson,et al.  Derivation of human embryonic stem cells in defined conditions , 2006, Nature Biotechnology.

[14]  G. Seidel,et al.  Reduced oxygen tension and EDTA improve bovine zygote development in a chemically defined medium. , 2000, Journal of animal science.

[15]  G. Semenzato TARGETING HIF-1 FOR CANCER THERAPY , 2003 .

[16]  R. Wenger,et al.  Cellular adaptation to hypoxia: O2‐sensing protein hydroxylases, hypoxia‐inducible transcription factors, and O2‐regulated gene expression , 2002, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[17]  Kyu-Won Kim,et al.  Hypoxia-inducible factor (HIF-1)α: its protein stability and biological functions , 2004, Experimental & Molecular Medicine.

[18]  J. Pouysségur,et al.  HIF-1: master and commander of the hypoxic world. A pharmacological approach to its regulation by siRNAs. , 2004, Biochemical pharmacology.

[19]  B Fischer,et al.  Oxygen tension in the oviduct and uterus of rhesus monkeys, hamsters and rabbits. , 1993, Journal of reproduction and fertility.

[20]  J. Thomson,et al.  Embryonic stem cell lines derived from human blastocysts. , 1998, Science.

[21]  P. Carmeliet,et al.  Loss of HIF-2α and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice , 2002, Nature Medicine.

[22]  G. Semenza,et al.  Defective vascularization of HIF-1alpha-null embryos is not associated with VEGF deficiency but with mesenchymal cell death. , 1999, Developmental biology.

[23]  Feng Zhao,et al.  Hypoxia enhances proliferation and tissue formation of human mesenchymal stem cells. , 2007, Biochemical and biophysical research communications.

[24]  L. Huang,et al.  Regulation of hypoxia-inducible factor 1α is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway , 1998 .

[25]  Xin Zhang,et al.  Expression and functional analysis of G1 to S regulatory components reveals an important role for CDK2 in cell cycle regulation in human embryonic stem cells , 2009, Oncogene.

[26]  David J. Anderson,et al.  Culture in Reduced Levels of Oxygen Promotes Clonogenic Sympathoadrenal Differentiation by Isolated Neural Crest Stem Cells , 2000, The Journal of Neuroscience.

[27]  R. Talbot,et al.  Transcriptome alterations due to physiological normoxic (2% O2) culture of human embryonic stem cells. , 2008, Regenerative medicine.

[28]  Jong-Ho Cha,et al.  Hypoxia-inducible factor-1 alpha inhibits self-renewal of mouse embryonic stem cells in Vitro via negative regulation of the leukemia inhibitory factor-STAT3 pathway. , 2007, The Journal of biological chemistry.

[29]  J. Shay,et al.  Hypoxia-Inducible Factor 1 Mediates Upregulation of Telomerase (hTERT) , 2004, Molecular and Cellular Biology.

[30]  G. Semenza,et al.  A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation , 1992, Molecular and cellular biology.

[31]  G. Goodall,et al.  Hypoxia-inducible Factor-1 (cid:1) mRNA Contains an Internal Ribosome Entry Site That Allows Efficient Translation during Normoxia and Hypoxia , 2022 .

[32]  G. Semenza,et al.  HIF-1 and human disease: one highly involved factor. , 2000, Genes & development.

[33]  John D Gordan,et al.  HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. , 2007, Cancer cell.

[34]  H. Kuo,et al.  A reduced oxygen tension (5%) is not beneficial for maintaining human embryonic stem cells in the undifferentiated state with short splitting intervals. , 2008, Human reproduction.

[35]  R. Schultz,et al.  Effects of oxygen tension on gene expression in preimplantation mouse embryos. , 2006, Fertility and sterility.

[36]  A. Harvey,et al.  Oxygen-Regulated Gene Expression in Bovine Blastocysts1 , 2004, Biology of reproduction.

[37]  Brian Keith,et al.  HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth. , 2006, Genes & development.

[38]  V. Erdmann,et al.  Differentiating the functional role of hypoxia‐inducible factor (HIF)‐1α and HIF‐2α (EPAS‐1) by the use of RNA interference: erythropoietin is a HIF‐2α target gene in Hep3B and Kelly cells , 2004 .

[39]  A C Simpson,et al.  Effect of oxygen concentration on in-vitro development of preimplantation sheep and cattle embryos. , 1990, Journal of reproduction and fertility.

[40]  R. Roberts,et al.  Identification of oxygen-sensitive transcriptional programs in human embryonic stem cells. , 2008, Stem cells and development.

[41]  Till Acker,et al.  Loss of HIF-2α and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice , 2002, Nature Medicine.

[42]  A. Simpson,et al.  Physiologic oxygen enhances human embryonic stem cell clonal recovery and reduces chromosomal abnormalities. , 2006, Cloning and stem cells.

[43]  E. Rankin,et al.  Hypoxia-inducible factor-2 (HIF-2) regulates hepatic erythropoietin in vivo. , 2007, The Journal of clinical investigation.

[44]  H. Leese,et al.  Protection against reactive oxygen species during mouse preimplantation embryo development: Role of EDTA, oxygen tension, catalase, superoxide dismutase and pyruvate , 2001, Molecular reproduction and development.

[45]  Yukihiro Kondo,et al.  Expression and Characterization of Hypoxia-Inducible Factor (HIF)-3α in Human Kidney: Suppression of HIF-Mediated Gene Expression by HIF-3α , 2001 .

[46]  G. Semenza Targeting HIF-1 for cancer therapy , 2003, Nature Reviews Cancer.

[47]  S. Lindenberg,et al.  The impact of oxygen tension on developmental competence of post‐thaw human embryos , 2005, Acta obstetricia et gynecologica Scandinavica.

[48]  Lei Xiong,et al.  Hypoxia‐driven proliferation of embryonic neural stem/progenitor cells – role of hypoxia‐inducible transcription factor‐1α , 2008, The FEBS journal.

[49]  B. Kovačič,et al.  Influence of atmospheric versus reduced oxygen concentration on development of human blastocysts in vitro: a prospective study on sibling oocytes. , 2008, Reproductive biomedicine online.

[50]  M. Stojkovic,et al.  A role for NANOG in G1 to S transition in human embryonic stem cells through direct binding of CDK6 and CDC25A , 2009, The Journal of cell biology.