HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth - PubMed (original) (raw)

Comparative Study

HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth

Kelly L Covello et al. Genes Dev. 2006.

Abstract

The division, differentiation, and function of stem cells and multipotent progenitors are influenced by complex signals in the microenvironment, including oxygen availability. Using a genetic "knock-in" strategy, we demonstrate that targeted replacement of the oxygen-regulated transcription factor HIF-1alpha with HIF-2alpha results in expanded expression of HIF-2alpha-specific target genes including Oct-4, a transcription factor essential for maintaining stem cell pluripotency. We show that HIF-2alpha, but not HIF-1alpha, binds to the Oct-4 promoter and induces Oct-4 expression and transcriptional activity, thereby contributing to impaired development in homozygous Hif-2alpha KI/KI embryos, defective hematopoietic stem cell differentiation in embryoid bodies, and large embryonic stem cell (ES)-derived tumors characterized by altered cellular differentiation. Furthermore, loss of HIF-2alpha severely reduces the number of embryonic primordial germ cells, which require Oct-4 expression for survival and/or maintenance. These results identify Oct-4 as a HIF-2alpha-specific target gene and indicate that HIF-2alpha can regulate stem cell function and/or differentiation through activation of Oct-4, which in turn contributes to HIF-2alpha's tumor promoting activity.

PubMed Disclaimer

Figures

Figure 1.

Figure 1.

Disrupted development in Hif-2α KI embryos. (A_–_F) Between E10.5 and E13.5, ∼50% of Hif-2α KI/+ heterozygous embryos demonstrated a range of phenotypes. At E13.5, Hif-2α KI/+ heterozygous yolk sacs and embryos display hemorrhaging (B, arrows) and gross abnormalities (C), compared with wild-type (WT) controls (A). (C) Several embryos disintegrated upon dissection, suggesting they were dead or severely abnormal. At E10.5, Hif-2α KI/+ heterozygous embryos displayed phenotypes ranging from subtle hemorrhaging (E, arrow) to cardiac effusion (F, asterisk). A wild-type (WT) E10.5 embryo control is shown in D. (G_–_K) Comparison of wild-type (WT) (G,H) and homozygous Hif-2α KI/KI (I) and presumed homozygous Hif-2α KI/? (J,K) E7.5 embryos. Genotypes of embryos in J and K were inferred from morphological similarity to confirmed Hif-2α KI/KI embryos (shown in I) and frequency (10% of total embryos). Hif-2α KI/KI embryos displayed improper formation of the epiblast (ep, arrows in H,J,K), and embryonic (em) and extraembryonic (ex) cavities, compared with wild type (WT). (L–P) Whole-mount RNA in situ hybridization on E7.5 embryos using probes for the mesodermal marker Brachyury (Bry) (L,M) and visceral endodermal marker α-fetoprotein (AFP) (N–P). In contrast to wild-type (WT) littermate controls (L,N), which display normal mesoderm migration through the primitive streak (bracket in L), Hif-2α KI/KI embryos exhibit patterning defects (M) consistent with abnormal gastrulation. Similarly, AFP expression is restricted to the extraembryonic visceral endoderm in wild-type embryos (arrow in N), but maintained in embryonic visceral endoderm in Hif-2α KI/KI embryos (O,P).

Figure 2.

Figure 2.

Increased Oct-4, Vegf, and Tgf-α expression in Hif-2α KI/KI embryos. (A) Genotypes of individual E7.5 embryos were determined by PCR on genomic DNA and quantitative RT–PCR analysis of Hif-1α, Hif-2α, and Pgk transcripts. Transcript levels were normalized to 18S rRNA transcripts in each sample. Data are expressed as the ratio of target gene expression in each sample relative to a known wild-type (WT) litter (n = 8) that was stage matched. (B) Average levels of Hif-1α and Hif-2α mRNA in embryos pooled by genotype. (C) Average levels of Oct-4, Vegf, and Tgf-α mRNA in embryos pooled by genotype. (D–F) Whole-mount RNA in situ hybridization for Oct-4 mRNA demonstrated that Oct-4 is expressed in the embryonic epiblast in both wild-type (WT) and homozygous Hif-2α KI/KI E7.5 embryos.

Figure 3.

Figure 3.

Increased Vegf and Tgf-α expression in Hif-2α KI/KI 3.5 d EBs. (A) Schematic of HIF-1α control knock-in allele, in which a c-Myc-tagged Hif-1α cDNA was targeted to the first coding exon of the murine Hif-1α locus in ES cells. A detailed description is presented in Supplementary Figure 2. (B) Extracts from homozygous Hif-1α KI/KI and Hif-2α KI/KI ES cells were immunoprecipitated with c-Myc epitope antibodies and then analyzed on Western blots using c-Myc antibodies. Western blot analysis using AKT antibodies confirms that each sample in the immunoprecipitation experiment had equal amounts of input protein. (N) Normoxia (21% O2); (H) hypoxia (4 h at 1.5% O2). (C) Quantitative RT–PCR analysis in 3.5-d EBs of Alda A transcripts in wild-type (WT) and homozygous Hif-1α KI/KI, Hif-2α KI/KI, and _Hif-1α_−/− EBs. Fold change indicates increases in mRNA in EBs grown at 3.0% O2 relative to EBs grown at 21% O2. Transcript levels were normalized to 18S rRNA transcripts in each sample. (D) Real-time PCR analysis in 3.5-d EBs cultured at 21% O2 indicates increased Tgf-α, Vegf, and Hif-2α mRNA in two independently derived homozygous Hif-2α KI/KI clones. Changes in expression are specific to the Hif-2α KI allele, as Hif-1α KI/KI clones and Hif-1α−/− EBs behave similarly to wild type. (*) p < 0.005.

Figure 4.

Figure 4.

Expanded HIF-2α expression promotes Oct-4 expression and activity in 3.5-d EBs, and HIF-2α protein directly binds the Oct-4 promoter. (A) Real-time PCR analysis of Oct-4 demonstrates increased Oct-4 mRNA in normoxic 3.5-d EBs generated from two separately derived homozygous Hif-2α KI/KI clones. Increased Oct-4 expression is specific to the Hif-2α KI/KI EBs, as Hif-1α KI/KI and _Hif-1α_−/− EBs behave similarly to wild type (WT). (*) p < 0.005. (B) Western blot analysis shows increased Oct-4 protein in independent homozygous Hif-2α KI/KI EB clones. Levels were normalized to the AKT loading control, and fold change was determined by densitometry. (C) Real-time PCR analysis for mesodermal markers Fgf-4 and Bry mRNA demonstrates elevation of Oct-4 target genes specifically in independent homozygous Hif-2α KI/KI EBs. (*) p < 0.005. (D) Schematic diagram of the Oct-4 promoter displaying the relative position of putative HREs, as well as regions of notable sequence conservation between mouse and human (CR1–CR4). (E) ChIP on 293 cells shows that HIF-2α binds the putative HREs in CR4 and CR3 of the Oct-4 promoter. PCR products are obtained only when immunoprecipitated with HIF-2α and not when immunoprecipitated with an isotype control nonspecific antibody. No PCR product was observed in the HIF-2α ChIP using prim ers between CR2 and CR1 of the Oct-4 gene. Furthermore, HIF-1α does not appear to bind these regions in the Oct-4 promoter. (F) Transient transfection of Oct-4 promoter reporters into 786-O WT-8 renal carcinoma cells demonstrates that Oct-4 is induced by hypoxia (1.5% O2), but this induction is abolished when the HREs in CR4 and CR3 are removed. Normoxic basal expression was unchanged when comparing full-length Oct-4 promoter constructs (GOF-9) and Oct-4 promoter constructs with the HREs deleted (GOF-6).

Figure 5.

Figure 5.

Reversal of Hif-2α KI hematopoietic phenotypes by TGF-α and Oct-4 inhibition. (A) Normoxic day 9 Hif-2α KI/KI EBs form fewer hematopoietic CFUs than wild-type (WT) EBs. Numbers of CFU-E, CFU-M, CFU-G, and CFU-GM, CFU-GEMM progenitors are shown. Homozygous Hif-2α KI/KI EBs form more secondary EBs, compared with wild type. (B) Comparison of total CFU formation in EBs derived from ES cells of different genotype, expressed as a percentage of wild type (WT). Note the statistically significant decrease in the ability of homozygous Hif-2α KI/KI ES cells to form CFUs. This phenotype is specific to the two independent homozygous Hif-2α KI/KI clones, as Hif-1α KI/KI clones, _Hif-1α_−/− EBs, and Hif-2α KI/KI + HIF-2α shRNA clones behave similarly to wild type. (*) p < 0.005. (C) Western blot analysis for phosphorylated EGFR revealed inhibition of TGF-α activity by PD153035 in wild-type (WT) and in homozygous Hif-2α KI EBs. Hif-2α KI/KI EBs exhibited hyperphosphorylation of EGFR, consistent with increased TGF-α activity. Reduction in EGFR phosphorylation in wild-type EBs correlated to a decrease in total CFU formation. In contrast, reduction of EGFR phosphorylation in Hif-2α KI/KI EBs correlated to increased CFU formation. Restoration of CFU formation in Hif-2α KI/KI EBs was most notable in samples displaying nearly wild-type levels of EGFR phosphorylation. Total CFU formation is expressed as a percentage of that obtained with EBs derived from wild-type ES cells. (*) p < 0.005. (D) Western blot analysis indicating variable knock-down of Oct-4 protein and activity (Fgf-4 RNA expression) in EBs derived from homozygous Hif-2α KI/KI ES cell clones expressing Oct-4 shRNAs. EBs derived from Hif-2α KI/KI ES clones with Oct-4 protein levels similar to wild type (WT) displayed reduced expression of Fgf-4 RNA, and partial restoration of CFU formation compared with Hif-2α KI/KI. (*) p < 0.005. (E) EBs derived from Hif-2α KI/KI ES clones doubly inhibited for Oct-4 and TGF-α show even greater restoration of CFU formation and reduced secondary EB formation compared with Hif-2α KI/KI.

Figure 6.

Figure 6.

HIF-2α mediates tumor growth through up-regulation of Oct-4. (A) Gross appearance of teratomas resected from nude mice 21 d after subcutaneous injection with willd-type (WT), Hif-2α KI/KI ES cells, and Hif-2α KI/KI ES cells with Oct-4 shRNA knock-down (_Hif-2α KI/KI_-Oct-4 shRNA). (B) Final teratoma masses of wild-type (WT), Hif-2α KI/KI, and _Hif-2α KI/KI_-Oct-4 shRNA teratomas formed after 22 d of growth. Error bars measure standard error of the mean. (*) p = 0.012. (C) Western blot analysis of Oct-4 and β-catenin protein in wild-type (WT), Hif-2α KI/KI, and _Hif-2α KI/KI_-Oct-4 shRNA teratomas. Data are representative of three independent teratomas per genotype. (D) Ki67 staining of wild-type (WT), Hif-2α KI/KI, and _Hif-2α KI/KI_-Oct-4 shRNA teratoma sections. Final magnifications are 200×. (E) Nanog staining of wild type (WT), Hif-2α KI/KI, and Hif-2α KI/KI with Oct-4 shRNA knock-down teratoma sections.

Figure 7.

Figure 7.

HIF-2α is required for Oct-4 expression and normal PGC numbers. (A) Real-time PCR analysis demonstrates increased hypoxic induction (1.5% O2 for 16 h) of Oct-4 mRNA in wild-type (WT) and _Hif-1α_−/− 3.5-d EBs, which is diminished in _Hif-2α_−/− EBs. (B_–_D) PGCs in E8.5 embryos revealed by AP staining. Arrows point to representative PGCs in C and D. In contrast to wild-type (WT) and heterozygous Hif-2α+/− littermate controls (C), somite-matched _Hif-2α_−/− embryos exhibit reduced numbers of PGCs (D). (B) Quantification of PGC numbers in somite-matched E8.5 (s7–s11) embryos. (E,F) PGCs in E12.5 genital ridges dissected from embryos revealed by AP staining. (G) Proposed model for regulation of Oct-4 by HIF-2α. In PGCs, the Oct-4 locus is expressed and regulated by HIF-2α. The Oct-4 locus is also expressed in ES cells; however, HIF-2α activity is normally restricted or repressed in these cells (C.-J. Hu, S. Iyer, A. Sataur, K.L. Covello and M.C. Simon, in prep.). In differentiating somatic cells, the Oct-4 locus adopts a closed conformation and is not expressed. At this point, HIF-2α regulates other target genes (e.g., Vegf) without modulating Oct-4 levels. In homozygous Hif-2α KI/KI embryos, EBs, and teratomas, expanded HIF-2α expression results in up-regulation of Oct-4 and other targets. HIF-2α-mediated expression of an inappropriately derepressed Oct-4 locus in transformed cells could similarly modulate cancer stem cell identity and tumor progression.

Similar articles

Cited by

References

    1. Acker T, Diez-Juan A, Aragones J, Tjwa M, Brusselmans K, Moons L, Fukumura D, Moreno-Murciano M.P, Herbert J.M, Burger A, et al. Genetic evidence for a tumor suppressor role of HIF-2α. Cancer Cell. 2005;8:131–141. - PubMed
    1. Adelman D.M, Maltepe E, Simon M.C. Multilineage embryonic hematopoiesis requires hypoxic ARNT activity. Genes & Dev. 1999;13:2478–2483. - PMC - PubMed
    1. Boiani M, Eckardt S, Scholer H.R, McLaughlin K.J. Oct4 distribution and level in mouse clones: Consequences for pluripotency. Genes & Dev. 2002;16:1209–1219. - PMC - PubMed
    1. Boyer L.A, Lee T.I, Cole M.F, Johnstone S.E, Levine S.S, Zucker J.P, Guenther M.G, Kumar R.M, Murray H.L, Jenner R.G, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005;122:947–956. - PMC - PubMed
    1. Cheng L, Thomas A, Roth L.M, Zheng W, Michael H, Karim F.W. OCT4: A novel biomarker for dysgerminoma of the ovary. Am. J. Surg. Pathol. 2004;28:1341–1346. - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources