Myc represses primitive endoderm differentiation in pluripotent stem cells - PubMed (original) (raw)

Myc represses primitive endoderm differentiation in pluripotent stem cells

Keriayn N Smith et al. Cell Stem Cell. 2010.

Abstract

The generation of induced pluripotent stem cells (iPSCs) provides a novel method to facilitate investigations into the mechanisms that control stem cell pluripotency and self-renewal. Myc has previously been shown to be critical for murine embryonic stem cell (mESC) maintenance, while also enhancing directed reprogramming of fibroblasts by effecting widespread changes in gene expression. Despite several studies identifying in vivo target genes, the precise mechanism by which Myc regulates pluripotency remains unknown. Here we report that codeletion of c- and N-MYC in iPSCs and ESCs results in their spontaneous differentiation to primitive endoderm. We show that Myc sustains pluripotency through repression of the primitive endoderm master regulator GATA6, while also contributing to cell cycle control by regulation of the mir-17-92 miRNA cluster. Our findings demonstrate the indispensable requirement for c- or N-myc in pluripotency beyond proliferative and metabolic control.

Copyright 2010 Elsevier Inc. All rights reserved.

PubMed Disclaimer

Figures

Figure 1

Figure 1. Deletion of c- and N-myc in iPSCs results in loss of self-renewal

(A) Derivation of double knockout c-myc; N-myc miPSCs by transfection of CreGFP and isolation by FACS. (B) Locus map of targeted alleles and genotype analysis. Genotype analysis was performed on genomic DNA isolated from parental c-MYCfl/fl;N-MYCfl/fl, CreGFP- (Flox) miPSCs, and CreGFP+ (dKO) miPSCs. Amplicon lengths and corresponding primer sets corresponding to their position at the c-MYC and N-MYC loci are indicated. (C) c-MYCfl/fl;N-MYCfl/fl miPSCs transfected with CreGFP were FACS-isolated to separate dKO and Flox cells. GFP- (Flox) and GFP+ (dKO) cells were then plated in mESC medium for 3 days. Left panels; phase contrast images of dKO and Flox cells on gelatin. Middle and right panels; images of dKO and Flox cells following alkaline phosphatase staining on gelatin and mouse embryo fibroblast feeders, respectively. Scale bar, 100μm. (D) Quantitative analysis of alkaline phosphatase staining for wild-type miPSCs, Flox and dKO cells. n>150, for each condition. (E) Left panel; cell cycle profiles of propidium iodide stained Flox and dKO cells obtained by flow cytometric analyses. Right panel; % of Flox and dKO cells in G1-, S- and G2/M-phases of the cell cycle as determined by flow cytometry analysis. (F) Immunostaining demonstrates dKO cells remain proliferative, compared to Flox cells, by BrdU incorporation after labeling for 24 hrs. Scale bar, 100μm. (See also Figure S1).

Figure 2

Figure 2. Deletion of c- and N-myc in iPSCs results in differentiation to primitive endoderm

(A) qRT-PCR of c-myc, N-myc, endoderm (Gata6 and Foxa2), mesoderm (Brachyury), and primitive ectoderm (Fgf5) markers indicate endoderm differentiation in dKO cells compared to Flox cells. Cells were cultured in the presence of LIF and experiments were performed in triplicate, normalized to GAPDH and represented as mean ± s.d. (B) Immunostaining for c-myc, N-myc, pluripotency markers, Nanog and SSEA-1 and endoderm markers, FoxA2 and Gata4, reveals the spontaneous differentiation to endoderm following loss of Myc in miPSCs cultured in LIF. Scale bar, 100 μm. (C) qRT-PCR examining Nanog, Gata6, Foxa2, Sox17, Brachyury, and Fgf5 transcripts in miPSCs cultured in LIF (iPS), and during embryoid body differentiation (Flox, dKO) indicates that the loss of Myc predisposes miPSCs to primitive endoderm differentiation. Experiments were performed in triplicate, normalized to GAPDH and represented as mean ± s.d. (D) qRT-PCR analysis of endoderm markers, Gata6, Foxa2, and Sox17; mesendoderm marker, Brachyury; primitive ectoderm marker, Fgf5; and ectoderm marker Otx2, 4 days after LIF removal with and without retinoic acid. iPS represents control miPSCs cultured in the presence of LIF. Triplicate experiments were performed, normalized to GAPDH and represented as mean ± s.d. (E) Flox and dKO miPSCs and mESCs expressing β-galactosidase were injected into blastocyst stage C57BL/6 embryos, transferred into recipient females and allowed to develop until E14.5. LacZ staining was then performed on fixed, whole embryos. The number of blastocysts injected, the number of chimeras generated and the % of chimeras generated are indicated. (See also Figure S2).

Figure 3

Figure 3. Conditional activation of c- or N-myc, but not L-myc, is sufficient to maintain pluripotency in dKO cells

(A) Flox and dKO cell morphology after transfection with c-mycER, N-mycER or L-mycER in the presence or absence of 4OHT for 3 days. Scale bar, 100 μm. (B) Quantitation of alkaline phosphatase staining (Figure S3) showing the % positive colonies versus the % negative colonies. n>300 for each condition. Error bars represent mean ± s.d. from triplicate experiments. (C) Activation of 4OHT-inducible N-mycER inhibits the activation of FoxA2 and Sox17 transcript as determined by qRT-PCR. Experiments were performed in triplicate, normalized to GAPDH and represented as mean ± s.d. The data are representative of multiple experiments where either c- or N-mycER expressing cell lines were used. (See also Figure S3).

Figure 4

Figure 4. c-myc regulates the miR-17-92 cluster to control the cell cycle

(A) c-myc6×9e10 binds to the upstream regulatory region of the miR-17-92 cluster in ChIP-Chip assays; *p<0.001. (B) Independent validation of ChIP-Chip analysis with ChIP-qPCR using the AB2.1 c-mycΔ/6×9e10 cell line. In control samples, ChIP-qPCR was carried out using chromatin immunoprecipitated by the 9e10 antibody with the c-mycΔ/Δ cell line and control IgG with the c-mycΔ/6×9e10 cell line. (C) miR-20a transcript is down-regulated upon deletion of c- and N-MYC in miPSCs. (D) Activation of c-mycER with 4OHT in mESCs increases miR-20a transcript over basal levels. Experiments were performed in triplicate, normalized to GAPDH and represented as mean ± s.d. (E) Increased expression of the mir-17-92 target, Rb2/p130 upon deletion of Myc in dKO cells. Flox and dKO cells were immunostained with an antibody for Rb2/p130. DNA, DAPI staining. Scale bar, 100 μm. (F) Myc targets miR-17 and miR-20a regulate Rb2. miPSCs were transfected with a firefly luciferase reporter containing a Rb2-UTR under a CMV promoter and either a scrambled control, miR-17 precursor, or a miR-20a precursor. Luciferase assays were performed in triplicate and normalized to a Renilla luciferase control. Data are representative of multiple experiments, *p<0.05; **p<0.01. (G) Cell cycle profiles (flow cytometry) comparing untransfected wild-type miPSCs (black bars), cells transfected with a non-targeting control (blue bars) or, cells transfected with an antisense oligonucleotide against miR-17 (red bars). % of cells in G1-, S-, and G2/M-phases are shown 48 hours after transfection. Similar results were obtained in mESCs (data not shown). (H) Immunostaining monitoring the incorporation of BrdU after labeling for 24 hrs demonstrates reduced cellular proliferation of cells upon transfection of antisense oligonucleotide inhibitors against miR-17. Assay was performed in triplicate and is represented as mean ± s.d., *p<0.05. (See also Figure S4).

Figure 5

Figure 5. c-myc binds and represses the primitive endoderm master regulator, GATA6 and suppresses primitive endoderm differentiation

(A) GATA6 is a Myc-bound target identified from ChIP-Chip assays; *p<0.001. (B) Independent validation of GATA6 as a Myc-bound target by ChIP-qPCR using 8 primer sets to scan the GATA6 region corresponding to the ChIP-Chip analysis shown in (A). Primer set 2 corresponds to the statistically significant region identified by DNA Analytics software (Agilent) in the GATA6 promoter shown in (A) to bind c-myc. Primer sets 5, 7 and 8 also represent regions of significant enrichment and primer sets 1, 3, 4 and 6 correspond to regions not bound by c-myc. (C) c-myc transcript is down-regulated in primitive endoderm. AFP-GFP mESCs were aggregated in the presence of LIF then cultured for 3 days. GFP positive and negative cells were isolated by FACS and analyzed by qRT-PCR. (D) Transcription of GATA6, SOX17 and FOXA2 increases in dKO cells. Flox (GFP-) or dKO (GFP+) cells were isolated by FACS, cultured for 3 days, and nuclear run-on assays performed by labeling nascent nuclear transcripts with biotin-16-UTP. After the isolation of biotinylated transcripts on streptavidin beads, qRT-PCR analysis was performed in triplicate. Gata6, Sox17 and FoxA2 values were normalized to GAPDH and represented as mean ± s.d. (E) Induction of Gata6 transcript by sodium orthovanadate is blocked by activation of c-mycER. mESCs were aggregated in the presence of orthovanadate for 24 hr, in the presence or absence of 4OHT. qRT-PCR analysis was performed in triplicate and values normalized to GAPDH and represented as mean ± s.d. Data are representative of multiple experiments. (F) mESCs carrying a c-mycER transgene or vector alone, were aggregated for 3 days in the presence of LIF, in the presence or absence of 4OHT. Embryoid bodies were probed with antibodies for Nanog or the endoderm marker Gata4 and stained with DAPI. Embryoid bodies were analyzed by confocal microscopy and DIC optics. (G) Sox17-GFP mESCs were transfected with vector alone or with a c-myc expression construct. Cells were then aggregated in the absence of LIF for 3 days. Flow cytometry was used to determine the effect of endoderm differentiation by evaluating the % of GFP+ cells. (H) GATA6 is required for endoderm formation following loss of c- and N-Myc. c-MYCfl/fl;N-MYCfl/fl miPSCs were transfected with CreGFP and Gata6 shRNA or scrambled (scr) shRNA construct. GFP positive and negative cells were FACS sorted, plated and analyzed after 3 days by qRT-PCR. Experiments were performed in triplicate, normalized to GAPDH and represented as mean ± s.d. (I) Myc-deleted and Gata6 knockdown cells have restricted differentiation potential. c-MYCfl/fl;N-MYCfl/fl miPSCs were transfected with CreGFP and Gata6 shRNA or scrambled (scr) shRNA construct. Control miPSCs (iPS) were cultured in LIF, and GFP positive (dKO) and negative cells (Flox) were FACS sorted, aggregated in the absence of LIF to induce differentiation and analyzed after 4 days by qRT-PCR. Experiments were performed in triplicate, normalized to GAPDH and represented as mean ± s.d. (See also Figure S5).

Similar articles

Cited by

References

    1. Aguda BD, Kim Y, Piper-Hunter MG, Friedman A, Marsh CB. MicroRNA regulation of a cancer network: consequences of the feedback loop involving miR-17-92, E2F, and Myc. Proc Natl Acad Sci USA. 2008;105:19678–19683. - PMC - PubMed
    1. Aparicio O, Geisberg JV, Struhl K. Chromatin immunoprecipitation for determining the association of proteins with specific genomic sequences in vivo. Current Protocols in Cell Biology. 2004;17:1–23. - PubMed
    1. Baudino TA, McKay C, Pendevile-Samain H, Nilsson JA, Maclean KH, White EL, Davis AC, Ihle JN, Cleveland JL. c-Myc is essential for vasculogenesis and angiogenesis during development and tumor progression. Genes Dev. 2002;16:2530–43. - PMC - PubMed
    1. Borowiak M, Maehr R, Chen S, Chen AE, Tang W, Fox JL, Schreiber SL, Melton DA. Small molecules efficiently direct differentiation of mouse and human embryonic stem cells. Cell Stem Cell. 2009;4:348–358. - PMC - PubMed
    1. Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, Murray HL, Jenner RG, Gifford DK, Melton DA, Jaenisch R, Young RA. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005;122:947–956. - PMC - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources