Abstract
An optimal culture system for human pluripotent stem cells should be fully defined and free of animal components. To date, most xeno-free culture systems require human feeder cells and/or highly complicated culture media that contain activators of the fibroblast growth factor (FGF) and transforming growth factor-β (TGFβ) signaling pathways, and none provide for replacement of FGF/TGFβ ligands with chemical compounds. The Wnt/β-catenin signaling pathway plays an important role in mouse embryonic stem cells in leukemia inhibitory factor-independent culture; however, the role of Wnt/β-catenin signaling in human pluripotent stem cell is still poorly understood and controversial because of the dual role of Wnts in proliferation and differentiation. Building on our previous investigations of small molecules modulating Wnt/β-catenin signaling in mouse embryonic stem cells, we identified a compound, ID-8, that could support Wnt-induced human embryonic stem cell proliferation and survival without differentiation. Dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) is the target of the small molecule ID-8. Its role in human pluripotent cell renewal was confirmed by DYRK knockdown in human embryonic stem cells. Using Wnt and the DYRK inhibitor ID-8, we have developed a novel and simple chemically defined xeno-free culture system that allows for long-term expansion of human pluripotent stem cells without FGF or TGFβ activation. These culture conditions do not include xenobiotic supplements, serum, serum replacement, or albumin. Using this culture system, we have shown that several human pluripotent cell lines maintained pluripotency (>20 passages) and a normal karyotype and still retained the ability to differentiate into derivatives of all three germ layers. This Wnt-dependent culture system should provide a platform for complete replacement of growth factors with chemical compounds.
Keywords: DYRK, Wnt, Human embryonic stem cells, Human induced pluripotent stem cells
Introduction
The goal of regenerative medicine is to repair or replace damaged or diseased tissues or organs. In the past decade, since the first human embryonic stem cells (hESCs) were described [1, 2], there has been remarkable progress toward clinical applications of hESC-derived cellular therapeutics. However, further improvements toward the economical production of large quantities of either hESCs or induced pluripotent stem cells (iPSCs) [3, 4] cultured in fully defined xeno-free conditions remain an important research goal for therapeutic applications. Although several feeder- and xeno-free culture conditions have been reported [5–7], these conditions require complex culture media or many human-derived protein components. In particular, to meet good manufacturing practice (GMP) standards, replacing such components with small molecules would provide significant advantages. The development of well-defined xeno-free culture conditions is contingent upon our understanding of the key signaling pathways involved in hESC self-renewal. The extrinsic factors regulating hESC maintenance and early differentiation events seem to differ from those of mouse embryonic stem cells (mESCs) and to date are incompletely understood [8], but activation of basic fibroblast growth factor (bFGF) and transforming growth factor-β (TGFβ)/Activin/Nodal signaling forms the cornerstone of most systems for hESC propagation [9, 10].
Wnt signaling plays important roles throughout development [11]. Wnt signaling is important in stem cell biology; however, there is no consensus as to whether Wnt signaling is important for differentiation of stem/progenitor cells or proliferation and maintenance of potency (pluripotency or multipotency) [12, 13]. Wnt/β-catenin signaling has been demonstrated to maintain pluripotency in mESCs in leukemia inhibitory factor (LIF)-independent culture, but it is dispensable in LIF-dependent culture [14–16]. In hESCs, it has been reported that Wnt/β-catenin signaling maintains cells in the undifferentiated state [12, 17, 18]; however, it has also been reported that Wnt/β-catenin signaling induces differentiation [19, 20]. Thus, the dichotomous behavior of Wnt/β-catenin signaling in controlling both proliferation and differentiation of hESCs has been unclear and has fueled enormous controversy concerning the role of Wnt signaling in maintenance of pluripotency and induction of differentiation.
Using a chemical genomic approach, we have previously identified small molecule chemical compounds that modulate Wnt/β-catenin signaling and developed a model that rationalizes these divergent behaviors as a result of differential transcriptional coactivator binding to β-catenin in various stem cell types, including mESCs [21–23]. Modulation of Wnt/β-catenin signaling provided long-term mESC self-renewal in defined culture conditions without LIF [22, 24]. Therefore, we investigated whether orchestration of Wnt/β-catenin signaling with a small molecule modulator would allow for the long-term growth and maintenance of hESCs under defined xeno-free conditions.
Materials and Methods
Cells and Cell Culture
The hESC lines HES2 [2], HES3 [2], H1 [1], and H9 [1] and the human fetal dermal fibroblast-derived iPSC line [25] were maintained using standard cell culture methodology. For enzymatic bulk expansion, the cells were cultured on mitotically arrested mouse embryonic fibroblast (MEF) feeder cell layers in Dulbecco's modified Eagle's medium/Ham's F-12 medium (DMEM/F-12; Sigma D6421; Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com) supplemented with 20% knockout serum replacement (KSR; Invitrogen, Carlsbad, CA, http://www.invitrogen.com), l-glutamine, nonessential amino acid, and 4 ng/ml recombinant bFGF2 (fibroblast growth factor 2 [FGF2]; Peprotech, Rocky Hill, NJ, http://www.peprotech.com), as previously described. For feeder-free culture, the MEF feeder layer was removed via separation from embryonic stem cell clumps by sedimentation under gravity during passage, and cells were cultured in MEF-conditioned medium (MEF-CM) on 30-fold diluted Matrigel (BD Biosciences, San Diego, CA, http://www.bdbiosciences.com)-coated culture dishes.
The replating assay was previously described [26]. Briefly, the hESCs in feeder-free culture were dissociated completely with 0.05% trypsin-EDTA (Invitrogen) and seeded at 104 cells per well in Matrigel-coated 6-well culture plates and cultured in MEF-CM. Various concentrations of Wnt3a (purchased from Peprotech or prepared in our laboratory on the basis of a previous report [27]; the activity of the purified Wnt3a was confirmed using the super TOPFLASH assay [28]), IQ-1, ID-8, and/or ICG-001 (all synthesized in our laboratory; identity and purity confirmed by proton nuclear magnetic resonance and liquid chromatography/mass spectrometry [LC/MS]) were supplemented into the culture media at the onset of seeding and then continuously until the end of culturing. For all assays, the cell and colony morphology were examined under a microscope, and the replating efficiency was examined by counting the number of colonies after 7 days of culture.
For xeno-free, feeder-free culture, the cells were passaged onto 2 μg/cm2 human fibronectin (from human foreskin fibroblasts; Sigma-Aldrich F2518), 10–20 μg/cm2 human laminin (from human fibroblasts; Sigma-Aldrich L4544), and 10 μg/cm2 human vitronectin (Recombinant Vitoronectin; Invitrogen PHE0011)-coated culture dishes and cultured in DMEM/F-12 (Sigma D6421) supplemented with l-glutamine, nonessential amino acids, insulin-transferrin-selenite (ITS) (Sigma-Aldrich I1884; dosage twice as high as manufacturer's instructions), 1 μg/ml l-ascorbic acid, 20–25 ng/ml Wnt3a, and 500 nM ID-8, with or without 4 ng/ml bFGF. The culture medium was changed daily. Some of the colonies detached from the edge of the dish after 5–7 days of culture. At this time or when colonies became confluent, cells were treated with 0.5 mg/ml GRGDTP peptide (AnaSpec, Fremont, CA, http://www.anaspec.com) or 1 μg/ml echistatin (Sigma-Aldrich) and incubated at 37°C for 15–45 minutes until all colonies started to detach at the edge region. The colonies were detached and partially dissociated into clumps by gentle pipetting, and then the clumps were washed with culture medium, collected by centrifugation, and seeded onto new dishes at a 1:2 or 3 split ratio.
For examination of cell attachment, subconfluent cells were dissociated as clumps, and a part of the solution containing cell clumps was dissociated into single cells with 0.25% trypsin-EDTA for cell counting. The clumps were seeded into 24-well plates with an estimated 5,000-cell clumps per well. After overnight culture, nonattached cells were washed away with phosphate-buffered saline (PBS), attached cells were dissociated with 0.25% trypsin-EDTA, and then the attached cell number was examined. For examination of cell population growth, the cells were seeded into 24-well plates with an estimated 2,000- or 5,000-cell clumps per well, and the cells were dissociated and counted every 24 hours.
Experiments using hESCs and hiPSCs in this study have been approved by University of Southern California Stem Cell Research Oversight Committee (No. 2008-6-1).
Quantitative Polymerase Chain Reaction Analysis
Total RNA was isolated from hESCs cultured in feeder-free conditions with or without Wnt3a and ID-8 using an RNeasy micro kit (Qiagen, Hilden, Germany, http://www1.qiagen.com). Reverse transcription was performed with Omniscript (Qiagen), and quantitative reverse transcription–polymerase chain reaction (PCR) was then performed using each gene-specific primer/probe mix (TaqMan Gene Expression Assays), TaqMan 2 × Master Mix, and the ABI Prism 7900 Sequence Detection system (Applied Biosystems, Foster City, CA, http://www.appliedbiosystems.com) according to the manufacturer's protocol. The PCR data were analyzed by the Δ/Δ CT method and normalized with Cycrophilin A (PPIA) expression with RQ Manager software (Applied Biosystems).
Identification of ID-8 Target
The cells were washed twice with 5 ml of ice-old PBS, harvested by scraping into 1 ml of PBS containing protease inhibitor cocktail (Calbiochem, San Diego, CA, http://www.emdbiosciences.com) and collected by centrifugation. The cell pellet was frozen and stored in liquid nitrogen. A total of 2 g of cell pellets were collected from 20 dishes each of 15-cm diameter. The pellets were thawed on ice and then suspended in 20 ml of chilled M-Per buffer (Pierce, Rockford, IL, http://www.piercenet.com) containing 1 mM dithiothreitol (DTT) and the protease inhibitor cocktail. The cell suspension was mixed and lysed on a nutating mixer for 30 minutes at 4°C. The lysate was then centrifuged at 10,000 rpm for 30 minutes at 4°C, and the supernatant was divided into two equal aliquots in 50-ml centrifuge tubes. Dimethyl sulfoxide (DMSO) control was added to one tube, and 100 μM ID-8 was added to the other tube. The tubes were incubated for 1 hour on a nutator at 4°C. Four hundred microliters of a 50% slurry of streptavidin-Sepharose (GE Healthcare Life Sciences, Piscataway, NJ, http://www.gelifesciences.com) was pre-equilibrated in M-Per buffer and then added to 10 ml of 50 μM biotinylated ID-8 and incubated for 1 hour at 4°C. The Sepharose beads were then washed extensively with M-Per to remove unbound biotinylated ID-8. The streptavidin-Sepharose bound with biotinylated ID-8 was mixed with the DMSO- or free ID-8-treated cell lysates described above. The mixture was then incubated on a nutating mixer for 4 hours at 4°C. The Sepharose beads, containing proteins bound to biotinylated ID-8, were washed three times with 1 ml of M-Per buffer and then mixed with 2× Laemmli Sample Buffer and boiled for 5 minutes. The proteins and Sepharose were then separated using Illustra MicroSpin Columns (GE Healthcare Life Sciences). Bound proteins eluted in Laemmli Buffer were subjected to SDS-polyacrylamide gel electrophoresis (PAGE) and silver stained (Invitrogen). The specific bands decreased by free competitor ID-8 were analyzed by LC/MS (LTQ-XL; Thermo Scientific). The elution was also analyzed by Western blotting with specific antibodies against dual-specificity tyrosine phosphorylation-regulated kinase 1a (DYRK1a) (2771; Cell Signaling Technology, Beverly, MA, http://www.cellsignal.com), DYRK2 (ab37912; Abcam, Cambridge, MA, http://www.abcam.com), DYRK3 (sc-66868; Santa Cruz Biotechnology Inc., Santa Cruz, CA, http://www.scbt.com), DYRK4 (ab37911; Abcam), or HIP2K (sc-100383; Santa Cruz Biotechnology).
Knockdown Assay
Several DYRK microRNAs (miRNAs) were designed and lentiviral vector plasmids were constructed using the Block-iT Pol II miR RNAi kit (Invitrogen) according to the manufacturer's protocol. The knockdown efficiencies of the miRNA expression plasmids were evaluated by transient transfection and Western blotting. The most efficient miRNA sequence, antisense, 5′-tcaaggagtcaatttcgtaacg-3′, and sense, 5′-gttacgaatgacctccttg-3′, was used for further lentiviral production.
To produce the miRNA expression lentiviral vectors, the plasmid was cotransfected with packaging plasmids for vesicular stomatitis virus G protein pseudotyped virus into 293T cells with Fugene6 transfection reagent (Roche Applied Science, Indianapolis, IN, http://www.roche-applied-science.com). Viral supernatants were collected and concentrated by ultracentrifugation for 2 hours. Feeder-free H9 cells were infected with the virus in MEF-CM supplemented with 10 μg/ml polybrene. The infected cells were selected and isolated as pools using 0.5–1 μg/ml Blasticidin over 1 week. The infected H9 cell pools were then used for examination of replating efficiency.
Nuclear Purification and Immunoprecipitation
The cells were washed twice with ice-cold PBS, scraped, and harvested following treatment with DMSO control, 500 nM ID-8, or 20 μM ICG-001 for 24 hours. The nuclear fraction was purified using an NE-PER nuclear extraction kit (Pierce, Thermo Scientific). For prevention of protein degradation, 1 mM DTT and a protease inhibitor cocktail (Calbiochem) were supplemented during harvest and processing. A total 5 μg of nuclear protein from each treated sample was then reacted with 1 μg of anti-CREB-binding protein (CBP) antibody (sc-369; Santa Cruz Biotechnology), anti-p300 antibody (sc-584; Santa Cruz Biotechnology), or normal rabbit IgG in 1 ml of immunoprecipitation buffer (25 mM Tris, HCl, pH 7.5, 150 mM NaCl, 5% glycerol, 0.5% Nonidet P40, 1 mM EDTA, 1 mM DTT, and protease inhibitor cocktail) at 4°C overnight. The protein-antibody complex was then reacted with 50 μl of 50% slurry of protein A-agarose beads (Roche Applied Science) on a nutator for 1 hour at 4°C. The bead complex was washed three times with immunoprecipitation buffer, resuspended with 2× Laemmli buffer, and boiled for 5 minutes. The protein and Sepharose were then separated on an Illustra MicroSpin Column (GE Healthcare Life Sciences). Bound proteins eluted in the Laemmli buffer were subjected to SDS-PAGE and transferred onto polyvinylidene difluoride membrane overnight. The coimmunoprecipitated β-catenin was detected by Western blotting with an anti-β-catenin antibody (610153; BD Biosciences). The intensity of the β-catenin Western blotting signal was analyzed using UN-SCAN-IT software (version 5.1, Silk Scientific, Orem, UT, http://www.silkscientific.com) and normalized with the signals in the total nuclear lysate.
Characterization, Karyotype Analysis, and Differentiation Assay
The hESC colonies were fixed with 4% paraformaldehyde (PFA), and alkaline phosphatase (ALP) activity was determined with a Vector Blue Alkaline Phosphatase Substrate Kit (Vector Laboratories, Burlingame, CA, http://www.vectorlabs.com). Immunostaining was carried out with the following primary antibodies: TRA-1–60 and TRA-1–80 (Santa Cruz Biotechnology), anti-stage-specific embryonic antigen (SSEA)-3 and -4 (Developmental Studies Hybridoma Bank, Iowa City, IA, http://www.uiowa.edu/∼dshbwww), GCTM-2 (our laboratory), anti-OCT-3 (clone C-10; Santa Cruz Biotechnology), rabbit anti-SOX2 (Millipore, Billerica, MA, http://www.millipore.com), or goat anti-Nanog (R&D Systems Inc., Minneapolis, http://www.rndsystems.com). Then, samples were incubated with Alexa Fluor 488- or 594-conjugateed antibodies (Molecular Probes, Eugene, OR, http://probes.invitrogen.com), and staining was detected by indirect immunofluorescence microscopy.
For fluorescence-activated cell sorting (FACS) analysis, cells were dissociated with TrypLE (Invitrogen), fixed with 2% PFA, and reacted with TRA-1–60 or GCTM-2 antibody followed by Alexa Fluor 488-conjugated anti-mouse IgM antibodies. The stained cells were then analyzed using an LSRII flow cytometer (BD Biosciences).
For karyotyping, hESCs were treated with 100 ng/ml colcemid (KaryoMax; Invitrogen), dispersed, and fixed. Twenty to 50 cells at metaphase in each sample were analyzed for G-banding at the 300–500 band levels following trypsin digestion and Giemsa staining.
For differentiation assays, embryoid bodies (EBs) were formed for 2 weeks in low-attachment culture plates and then plated onto glass culture slides as described previously [26]. After the cells grew out from attached EBs, they were fixed with 4% PFA, incubated with anti-βIII-tubulin (clone TU-20; Chemicon, Temecula, CA, http://www.chemicon.com), anti-glial fibrillary acidic protein (anti-GFAP; clone 4A11; BD Biosciences), anti-α-smooth muscle actin (αSMA) (clone 1A4; DakoCytomation, Glostrup, Denmark, http://www.dakocytomation.com), anti-Desmin (clone Ab-1; Thermo Scientific), anti-SOX17 (clone 245013; R&D Systems), anti-SOX7 (goat polyclonal; R&D Systems), or anti-α-fetoprotein (AFP) antibody (clone C3; Sigma-Aldrich) as the primary antibodies and then detected using Alexa Fluor 488- or 594-conjugated secondary antibodies (Molecular Probes) and fluorescence microscopy.
For teratoma formation, hESCs were dissociated as clumps, and a 200-μl suspension containing approximately 107 cells was subcutaneously injected into SCID mice (Jackson Laboratory, Bar Harbor, ME, http://www.jax.org). After 2–3 months, the resulting teratomas were dissected out and fixed with Bouin's fixative solution. Samples were embedded in paraffin, sectioned at 5-μm thickness, and subsequently stained with hematoxylin and eosin. This study has been approved by the University of Southern California Institutional Animal Care and Use Committee (No. 11273).
Results
Small Molecule ID-8 Supports hESC Maintenance in Combination with Wnt-3a
The effects of the canonical Wnt ligand Wnt3a and several candidate small molecule Wnt signaling modulators were first evaluated briefly in a replating assay [26] (Fig. 1A). In the replating assay, colonies were dissociated into single cells and seeded on Matrigel-coated plates in MEF-CM [29]. We evaluated cell survival by colony number (replating efficiency: colony number/seeded cell number), proliferation by colony size, and the undifferentiated state by morphology and ALP positivity. When HES2 cells [2] were dissociated and seeded in control MEF-CM, the replating efficiency was ∼0.6%. Supplementation with Wnt3a enhanced colony formation slightly (∼1% at 100 ng/ml). The colonies were larger and consisted of more cells. All the colonies were still ALP positive. However, the cells in the Wnt-treated colonies appeared flatter than with the control non-Wnt conditions (Fig. 1B, 1C). These results suggested that Wnt3a supplementation enhances hESC survival and proliferation but also partially induces differentiation. As anticipated, treatment with the small molecule ICG-001, which directly modulates Wnt/β-catenin signaling by antagonizing the interaction of β-catenin with transcriptional cofactor CBP and enhancing its interaction with transcriptional cofactor p300 [21], strongly induced cell differentiation in the presence or absence of Wnt3a, and almost all colonies were ALP negative. The other modulator, IQ-1, which is sufficient to maintain mESC proliferation and pluripotency for extended periods of time in the absence of serum and LIF in conjunction with Wnt3a by enhancing the CBP/β-catenin association [22], did not significantly affect hESC survival and proliferation by itself even at the highest dose in the nontoxic range (10 μM), although it partially prevented Wnt-induced differentiation. By contrast, the third modulator, ID-8, which was identified in the same screen as IQ-1 [24], increased hESC survival (∼1.1% at 0.5 μM). The combination of Wnt3a and ID-8 further increased survival (∼1.7%) and completely prevented Wnt-induced differentiation morphologically without disrupting proliferation. Cells treated with this combination expressed pluripotency markers, including ALP, OCT4, NANOG, and SOX2 at essentially the same levels as control hESCs (Fig. 1D, 1F). Wnt-induced differentiation marker gene expression (i.e., GATA6, SOX17, T [Brachyury], GSC [goosecoid] and CDX2) was dramatically reduced by ID-8 at a dose that maintained the undifferentiated state in this assay (Fig. 1G, 1H). To examine the generality of this effect in hESCs, replating assays were performed with the additional hESC lines HES3 [2] and H9 [1] (Fig. 1E). In both, the combination of Wnt and ID-8 enhanced hESC replating efficiency, and colonies expressed ALP and displayed undifferentiated morphology.
Figure 1.
A combination of ID-8 and Wnt ligand enhances human embryonic stem cell (hESC) survival and proliferation. (A): Screening of small molecule Wnt signaling modulators in replating assays with HES2 cells in mouse embryonic fibroblast conditioned medium (MEF-CM). Colony numbers per seeded cell numbers indicate replating efficiency. Statistical significance (p value) was calculated by Student's t test. Shown are cultured plates stained with ALP (B) and morphologies of typical colonies at day 7 (C). (D): ALP staining and OCT4 immunostaining of HES2 cells cultured in MEF-CM with 0.5 μM ID-8 and 100 ng/ml Wnt3a. (E): Replating efficiency of HES2, HES3, and H9 cells cultured in MEF-CM with or without ID-8 and Wnt3a. Statistical significance (p value) was calculated by Student's t test. Graphs show quantitative polymerase chain reaction analysis of pluripotent markers (F) and differentiation markers (G, H) in HES2 cells cultured in MEF-CM with or without ID-8 and Wnt3a. Scale bars = 200 μm; error bars indicate SD. Abbreviation: ALP, alkaline phosphatase.
ID-8 Targets DYRK in hESCs
We next sought to investigate the molecular targets and mechanism(s) of action of ID-8 in Wnt/β-catenin signaling. To identify the molecular target(s) of ID-8 (Fig. 2A), we used affinity chromatography (Fig. 2B). We identified DYRK2 as a binding partner of ID-8 in cell lysates; its binding to the ID-8 affinity column was competed away by free ID-8. Immunoblotting of the bound proteins identified not only DYRK2 but also DYRK4 (Fig. 2C). These data suggested that these or other members of the DYRK family are critical molecular targets of ID-8 in hESCs. To confirm this, we designed miRNAs to target the DYRK family and evaluated their effects on protein expression after transfection by immunoblotting (Fig. 2D, 2E). The DYRK miRNA-transfected hESCs maintained their undifferentiated state, as did the untransfected or scrambled control miRNA-transfected cells in MEF-CM or on feeders (supplemental information Fig. 1), showing that there was no adverse effect of the miRNA under conditions that support hESC renewal. Next, we performed replating assays with the stably transfected hESCs. Survival and self-renewal were not different between DYRK-knockdown cells and control cells in the absence of Wnt. However, both parameters were enhanced in the DYRK-knockdown cells in the presence of Wnt ligand (Fig. 2F). The effect of DYRK-knockdown was thus very similar to ID-8 treatment in the replating assay. Taken together, these data indicate that members of the DYRK family are direct targets of ID-8 and that ID-8 enhances Wnt-mediated hESC survival and proliferation via inhibition of DYRKs.
Figure 2.
Identification of ID-8 target molecule. (A): Molecular structure of ID-8. (B): Silver staining of proteins pulled down with biotin-conjugated ID-8 and streptavidin-Sepharose in SDS-polyacrylamide gel electrophoresis. Arrow indicates a specific band (60 kDa) decreased by competition with nonbiotin-conjugated ID-8. (C): Western blotting with DYRK family-specific antibodies. Samples were pulled down with resin only, biotin-conjugated ID-8, or biotin-conjugated ID-8 plus nonconjugated ID-8. (D): Western blotting with DYRK antibodies after transient transfection of DYRK miRNA or control scrambled miRNA expression plasmid. (E): Western blotting of stable transfectants. (F): Replating efficiency of H9 human embryonic stem cells (hESCs) stably transfected with miRNA. Error bars indicate SD. Statistical significance (p value) was calculated by Student's t test. (G): Western blotting image for coimmunoprecipitation of nuclear β-catenin from ID-8, ICG-001, or nontreated cells with anti-CBP, anti-p300 negative control IgG. (H): Band intensity analysis of CBP or p300-associated β-catenin in ID-8- or ICG-001-treated cells relative to nontreated control cells. (I): Western blotting image for coimmunoprecipitation of nuclear β-catenin from DYRK miRNA or control scrambled miRNA-transfected cells. (J): Band intensity analysis of CBP or p300-associated β-catenin in DYRK miRNA-transfected cells relative to scrambled miRNA-transfected control cells. Abbreviations: Cont, control; DYRK, dual-specificity tyrosine phosphorylation-regulated kinase; IP, immunoprecipitation.
Based upon our model and previous investigations in mESCs [22], we anticipated that ID-8 would modulate Wnt/β-catenin signaling through the enhancement of CBP/β-catenin association at the expense of the p300/β-catenin association in hESCs. To test this hypothesis, CBP and p300 were immunoprecipitated from purified nuclear fractions and then coprecipitated β-catenin amount was examined by immunoblotting (Fig. 2G, 2H). As expected, levels of the CBP/β-catenin complex were significantly enhanced in ID-8-treated cells compared with control and in particular to ICG-001-treated cells. A similar result was obtained in DYRK-knockdown cells (Fig. 2I, 2J). We conclude that ID-8, through inhibition of DYRK family members, supports maintenance of the undifferentiated state in the presence of Wnt by modulating Wnt/β-catenin signaling and enhancing the CBP/β-catenin association in hESCs, similar to the mechanism observed in mESCs treated with IQ-1 and Wnt.
ID-8 and Wnt-Based Xeno- and Feeder-Free Culture System for hESCs and Human iPSCs
Because Wnt and IQ-1 could maintain mESCs without serum and LIF [22], we next investigated the development of a xeno-free, feeder-free hESC culture system using ID-8 with Wnt activation. We first examined the maintenance of hESCs in conventional hESC medium, supplemented with Wnt3a and ID-8, and stepwise elimination of bFGF and KSR components, such as albumin, ITS, and ascorbic acid. After extensive investigation, we determined that the minimal medium required to maintain hESC proliferation and pluripotency consisted of DMEM/F-12 with Wnt3a (25 ng/ml), ID-8 (0.5 μM), and ITS (supplemental information Fig. 2A). In the absence of ID-8, Wnt3a, or ITS, no colonies were obtained. l-Ascorbic acid did not affect the maintenance of hESCs in this assay, apart from causing growth inhibition at doses greater than 25 μg/ml. In the minimal effective condition, hESCs maintained their undifferentiated morphology in the absence of bFGF; however, proliferation was significantly enhanced as in MEF-CM by supplementation of 4 ng/ml bFGF (supplemental information Fig. 2A). This concentration of bFGF is much lower than that reported previously for xeno- and feeder-free medium supplemented with bFGF (100 ng/ml) [10].
Initially, we maintained cells on Matrigel, a complex animal-derived extracellular matrix (ECM). However, we investigated various ECM combinations and found that a combination of fibronectin, laminin, and vitronectin (FLV) was best for supporting hESC attachment (supplemental information Fig. 2B) and self-renewal and for maintenance of undifferentiated colony morphology in the medium. Using this ECM combination, the majority of the colonies remained intact up to 7 days in this culture condition, although some colonies tended to detach after 5 days. Therefore, it proved optimal to passage at around 5 days after seeding.
Because of the extremely low level of protein in these cultures (i.e., no albumin or Matrigel), the hESCs are extremely sensitive to damage caused by the dissociation enzymes normally used for passaging, such as collagenase IV, dispase, or trypsin/EDTA. ROCK inhibitor, which has been reported to enhance hESC survival [30], was not very effective under these conditions and also induced differentiation, even with manual mechanical passaging. To develop a passaging process compatible with the very low protein content of our defined medium, we investigated several integrin antagonists. We found that both the synthetic peptide disintegrin GRGDTP and echistatin effectively induced colony detachment and allowed for the passage of hESCs in our medium.
Combining these discoveries, a simple xeno-free and feeder-free culture system was developed incorporating ITS/Wnt3a/ID-8/DMEM/F-12 media, FLV-coated dishes, and disintegrin passaging. Ascorbic acid could be omitted in short-term initial screening; however, we decided to use 1 μg/ml ascorbic acid for additional long-term experiments because it was reported that ascorbate supports hESCs epigenetic status [31]. hESCs transferred into this culture system from normal serum replacement-containing medium were stable and required no culture adaptation. All three hESC lines examined (HES2, HES3, and H9) maintained undifferentiated morphologies for >24 passages (Fig. 3A, passage 17) in this system without bFGF supplementation. The cell proliferation and population doubling rate in these culture conditions without bFGF was slightly lower than that with bFGF (data not shown) or the control MEF-CM (supplemental information Fig. 3A, 3C). All pluripotent stem cells markers examined, including OCT4, SOX2, NANOG, SSEA3, SSEA4, TRA-1–60, TRA-1–81, GCTM-2, and ALP, were expressed in the cells (Fig. 3B). FACS analysis showed that the populations of TRA-1–60- or GCTM-2-positive cells were similar to those maintained under MEF-CM culture conditions (Fig. 3C). Gene expression levels of pluripotent stem cell markers, including OCT4, NANOG, SOX2, and DNMT3B, were not significantly different from those of cells maintained on feeder layers and/or in MEF-CM (Fig. 3D). Normal karyotypes were also maintained in these cells after 22 passages (Fig. 3E). After 17–24 passages, EB-mediated in vitro differentiation and immunostaining for the three germ layer cell markers were performed to evaluate the differentiation capacity of the cultured hESCs (Fig. 4A). These cells readily differentiated into derivatives expressing markers characteristic of all three germ layers, with ectodermal cells expressing βIII-tubulin and GFAP, mesodermal cells expressing αSMA and Desmin, endodermal cells expressing AFP, definitive endodermal cells expressing SOX17 but not expressing SOX7, and extraembryonic endodermal cells expressing SOX7 strongly and SOX17 weakly. In addition, after 22 passages, the cells formed teratomas containing derivatives of all three germ layers when injected subcutaneously into immunodeficient mice (Fig. 4B). In addition to the female hESC cell lines used in the experiments above, a male hESC line H1 [1] and a human dermal fibroblast-derived iPSC line [25] were examined in our culture system. Both cell line cells were maintained in the undifferentiated state for at least 15 passages (Fig. 3A; supplemental information Fig. 3A, 3C). These data suggest that our xeno-free, feeder-free culture system can be used to maintain diploid human pluripotent stem cells long-term.
Figure 3.
Characterization of human pluripotent cells maintained in xeno-free and feeder-free culture system. (A): Typical colony morphologies at passage 7. (B): Immunostaining of undifferentiated cell markers in HES3 cells at passage 24. Similar results were observed in H9 and HES2 cells (data not shown). (C): Flow cytometry analysis of undifferentiated cell markers in H9 cells at passage 26. White histograms indicate cells stained with secondary antibody; black histograms indicate cells stained with primary and secondary antibodies. Percentages of positive cells are listed on the bars. Similar results were observed in HES2 and HES3 cells (data not shown). (D): Quantitative polymerase chain reaction analysis of pluripotent stem cell markers in HES2 cells at passage 24. Error bars indicate SD. Similar results were observed in H9 and HES3 cells (data not shown). (E): G-banding karyotype at passage 22. Abbreviations: iPSC, induced pluripotent stem cell; MEF-CM, mouse embryonic fibroblast conditioned medium.
Figure 4.
Differentiation capacity of human pluripotent stem cells maintained in xeno-free and feeder-free culture system. (A): Immunostaining of ectoderm (βIII-tubulin and GFAP), mesoderm (αSMA, Desmin), endoderm (AFP), definitive endoderm (SOX17-positive and SOX7-negative) and extraembryonic endoderm (SOX7-positive and SOX17-weakly positive)-derived cell markers from in vitro differentiation assay with HES3 cells after passages 17–24. Similar results were observed in the differentiation assay with H9 and HES2 cells (data not shown). (B): Ectodermal (neuroepithelium and pigmented cell), mesodermal (muscle and cartilage), and endodermal (gut-like and lung-like) tissue in teratoma formed with H9 cells after passage 22 in the culture system. Similar results were observed in teratomas from HES2 and HES3 cells (data not shown). Scale bars = 100 μm. Abbreviations: AFP, α-fetoprotein; GFAP, glial fibrillary acidic protein; SMA, smooth muscle actin.
Discussion
Historically, hESCs or iPSCs have been maintained on MEF feeder layers and in media containing serum or serum replacement [1–4]. These conditions include animal components, and cultures so maintained may be contaminated with pathogens. Therefore, the development of a xeno-free hESC and iPSC culture system is highly desirable for regenerative medicine applications of human pluripotent cells. Several reports have described xeno-free culture systems using human feeders and human serum components [32, 33]. A few additional reports demonstrated xeno-free and feeder-free culture systems for the maintenance of the undifferentiated state of hESCs [5–7]. These culture systems are quite complex and require a large number of human protein components and/or GMP-certified animal components. On the basis of these reports, a few xeno-free and feeder-free culture systems have become commercially available. However, the complexity incurred by the requirement of multiple recombinant proteins, human and/or certified animal components likely will result in cost-prohibitive protocols and more complexities in quality control. A very recent report described a simple xeno-free and feeder-free E8 medium consisting of eight components [10]. Six of the eight components, DMEM/F-12, insulin, transferrin, selenium, ascorbic acid, and NaHCO3 (present in DMEM/F-12), are also included in our medium, but the remaining two, bFGF and TGFβ/Nodal, are not required for our medium, which instead contains Wnt3a and ID-8. Replacement of all recombinant growth factors with chemical compounds, however, would be highly beneficial. However, to date, no previous defined formulation has eliminated the use of bFGF or TGFβ. In addition, several reports have appeared describing the screening of large chemical libraries for small molecules that support hESC renewal [34, 35]; however, to this point, these efforts have not led to a vastly simplified xeno-free culture medium. Because there are several compounds reported to act as activators of the Wnt/β-catenin signaling pathway, we anticipate that these could be incorporated into the system we describe to further eliminate protein growth factor requirements.
Several reports have demonstrated a role for Wnt/β-catenin signaling in mESC self-renewal together with FGF/MEK pathway inhibitors in the absence of LIF and serum. Under these conditions, Wnt appears to function via the interaction of β-catenin and the Tcf3 transcriptional repressor, with abrogation of Tcf3 repressor function and recruitment of Tcf1 as a transcriptional activator to pluripotent gene promoters. However, direct Wnt/β-catenin activation per se is dispensable in mESCs in LIF-dependent culture [14–16]. Additionally, it was recently shown that β-catenin, through a Tcf-independent mechanism, reinforces Oct4 activity to maintain pluripotency [36], and Wnt in combination with LIF prevents mESC differentiation to epiblast stem cell (EpiSC) [37]. To date, our understanding of Wnt/β-catenin signaling in human pluripotent stem cells has remained highly controversial because of the unresolved basis for the dual function of Wnt/β-catenin signaling in pluripotency and differentiation. Sato et al. described positive if short-term effects of BIO, a known small molecule inhibitor of GSK-3, on both mESC and hESC maintenance, demonstrating a normal differentiation program capacity after withdrawal from these conditions [12], although there has been significant controversy regarding these findings [13, 26].
Our previous investigations demonstrated that a small molecule Wnt/β-catenin signaling modulator could maintain mESC proliferation and pluripotency for extended periods of time in LIF- and serum-independent culture by modulating β-catenin transcriptional coactivator usage from p300 to CBP [22, 24]. The small molecules we studied, IQ-1 and ID-8, were originally identified through a screen for maintenance of mESC pluripotency. Here we show that ID-8 binds DYRK family members. The target of ID-8 is completely different from the target of IQ-1, the PR72/130 subunit of PP2A; however, ID-8 also enhances the CBP/β-catenin interaction indirectly in hESCs, and in that regard, its effects are mechanistically similar to the effects of IQ-1 in mESCs. The mechanism of DYRK inhibition in hESCs by ID-8 was confirmed by the observation of a similar phenotype in DYRK knockdown and ID-8-treated hESCs. Therefore, the different effects of IQ-1 and ID-8 in hESCs may be dependent on the differences in the function or expression of PR72/130 and DYRKs in mESCs and hESCs. The DYRK family is known to facilitate differentiation in many tissues and to interact with many proteins involved in stem cell proliferation and survival, including p53, STAT3, cyclin-D1, and caspase-9 [38–40]. However, their role in hESC self-renewal, differentiation, or Wnt/β-catenin signaling has not been previously reported. Further identification of DYRK substrates and the investigation of interaction between TCFs, CBP/p300, and DYRK in hESCs will be required to achieve a detailed understanding of the molecular mechanisms and of Wnt/β-catenin/TCF signaling in hESC self-renewal.
In the present study, we demonstrated long-term maintenance of pluripotency of hESCs in a Wnt/β-catenin/CBP-dependent fashion. Thus, a common plausible molecular mechanism for Wnt/β-catenin signaling maintaining in both mESC and hESC self-renewal is emerging, in which β-catenin by specifically interacting with the coactivator CBP maintains pluripotency [41]. The role of Tcf1/Tcf3, either interacting via β-catenin and CBP/p300 or independently, has not yet been determined and is the subject of ongoing investigations. In this report, we demonstrated that Wnt/β-catenin signaling together with a small molecule DYRK inhibitor is sufficient to maintain hESCs in the undifferentiated state. Furthermore, we show that β-catenin coactivator usage is shifted from p300 to CBP, similar to our previous report concerning mESCs using the small molecule IQ-1, which although interacting with a completely different molecular target, also biases CBP/β-catenin transcription to maintain pluripotency. In our culture system, bFGF signaling is dispensable and only enhances proliferation, and the dosage of bFGF required to achieve this effect is much lower than that used in other xeno- and feeder-free culture systems [10]. This is the first demonstration to our knowledge that hESC self-renewal can be maintained independently of exogenous activation of FGF or TGFβ signaling. Our findings suggest that the controversy surrounding the role of Wnt/β-catenin signaling in embryonic stem cells in previous reports may be due to initial biasing of CBP/β-catenin transcription, which also drives a negative feedback loop, thereby increasing the p300/β-catenin interaction. Furthermore, mixed results from concurrent activation of other signaling pathways, such as bFGF and/or TGFβ, may have also complicated previous interpretations. This situation is similar to the requirements for Wnt/β-catenin signaling in mESCs in LIF-dependent versus LIF-independent culture systems. The culture system we describe provides a powerful platform to explore these questions, as it relies on the activation of a relatively limited and very different set of signaling pathways compared with other systems described to date.
Conclusion
In conclusion, we have developed a simple human pluripotent stem cell culture system containing minimal growth factors Wnt and the small molecule DYRK inhibitor ID-8 that provides for stem cell renewal in the absence of bFGF and TGFβ/Nodal/Activin signaling. Additionally, we have developed a gentle subculture methodology that uses a synthetic disintegrin peptide. Our system still requires purified human ECM molecules, which can be certified for GMP; however, this need could be obviated with the development of chemically defined substrates. GMP manufacturing of hESCs or iPSCs using this culture system could be an important step toward future regenerative medical therapies. These findings also provide novel insights into the biochemical regulation of hESC self-renewal and new tools to probe the signaling pathways that control it.
Acknowledgments
We thank the Stem Cell Core facilities at Children's Hospital Los Angeles and at the University of Southern California for providing hESCs. Work at Kyoto University was supported by New Energy and Industrial Technology Development Organization (NEDO) Japan, Grant #P10027 Fundamental Technology Development for Promoting Industrial Application of Human Stem Cells. Work at the University of Southern California was supported by California Institute for Regenerative Medicine, New Cell Lines Grant #RL1-00667-1, New Technology for the Derivation of Human Pluripotent Stem Cell Lines for Clinical Use.
Author Contributions
K.H.: conception, design, experimentation, data analysis and interpretation, writing, final approval of manuscript; J.-L.T.: conception, design, experimentation, data analysis and interpretation, writing; S.Y. and C.N.: experimentation, data analysis; C.-L.H.: data analysis and interpretation; H.S., M.M., T.M., M.Y., and C.L.: provided research materials; N.N.: data analysis and interpretation, financial support; M.F.P.: data analysis and interpretation, writing, financial support; M.K.: conception, design, data analysis and interpretation, writing, financial support, final approval of manuscript.
Disclosure of Potential Conflicts of Interest
Kouichi Hasegawa and Michael Kahn both have equity positions in Cell Guidance Systems, Inc. Cell Guidance Systems, Inc., has licensed technology from University of Southern California related to this work.
References
- 1.Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–1147. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
- 2.Reubinoff BE, Pera MF, Fong CY, et al. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol. 2000;18:399–404. doi: 10.1038/74447. [DOI] [PubMed] [Google Scholar]
- 3.Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–1920. doi: 10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
- 4.Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–872. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
- 5.Ludwig TE, Levenstein ME, Jones JM, et al. Derivation of human embryonic stem cells in defined conditions. Nat Biotechnol. 2006;24:185–187. doi: 10.1038/nbt1177. [DOI] [PubMed] [Google Scholar]
- 6.Lu J, Hou R, Booth CJ, et al. Defined culture conditions of human embryonic stem cells. Proc Natl Acad Sci U S A. 2006;103:5688–5693. doi: 10.1073/pnas.0601383103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Yao S, Chen S, Clark J, et al. Long-term self-renewal and directed differentiation of human embryonic stem cells in chemically defined conditions. Proc Natl Acad Sci U S A. 2006;103:6907–6912. doi: 10.1073/pnas.0602280103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pera MF, Tam PP. Extrinsic regulation of pluripotent stem cells. Nature. 2010;465:713–720. doi: 10.1038/nature09228. [DOI] [PubMed] [Google Scholar]
- 9.Hasegawa K, Pomeroy JE, Pera MF. Current technology for the derivation of pluripotent stem cell lines from human embryos. Cell Stem Cell. 2010;6:521–531. doi: 10.1016/j.stem.2010.05.010. [DOI] [PubMed] [Google Scholar]
- 10.Chen G, Gulbranson DR, Hou Z, et al. Chemically defined conditions for human iPSC derivation and culture. Nat Methods. 2011;8:424–429. doi: 10.1038/nmeth.1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis. 2008;4:68–75. doi: 10.4161/org.4.2.5851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sato N, Meijer L, Skaltsounis L, et al. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat Med. 2004;10:55–63. doi: 10.1038/nm979. [DOI] [PubMed] [Google Scholar]
- 13.Dravid G, Ye Z, Hammond H, et al. Defining the role of Wnt/β-catenin signaling in the survival, proliferation, and self-renewal of human embryonic stem cells. Stem Cells. 2005;23:1489–1501. doi: 10.1634/stemcells.2005-0034. [DOI] [PubMed] [Google Scholar]
- 14.Lyashenko N, Winter M, Migliorini D, et al. Differential requirement for the dual functions of beta-catenin in embryonic stem cell self-renewal and germ layer formation. Nat Cell Biol. 2011;13:753–761. doi: 10.1038/ncb2260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wray J, Kalkan T, Gomez-Lopez S, et al. Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat Cell Biol. 2011;13:838–845. doi: 10.1038/ncb2267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Yi F, Pereira L, Hoffman JA, et al. Opposing effects of Tcf3 and Tcf1 control Wnt stimulation of embryonic stem cell self-renewal. Nat Cell Biol. 2011;13:762–770. doi: 10.1038/ncb2283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cai L, Ye Z, Zhou BY, et al. Promoting human embryonic stem cell renewal or differentiation by modulating Wnt signal and culture conditions. Cell Res. 2007;17:62–72. doi: 10.1038/sj.cr.7310138. [DOI] [PubMed] [Google Scholar]
- 18.Melchior K, Weiß J, Zaehres H, et al. The WNT receptor FZD7 contributes to self-renewal signaling of human embryonic stem cells. Biol Chem. 2008;389:897–903. doi: 10.1515/BC.2008.108. [DOI] [PubMed] [Google Scholar]
- 19.Sumi T, Tsuneyoshi N, Nakatsuji N, et al. Defining early lineage specification of human embryonic stem cells by the orchestrated balance of canonical Wnt/β-catenin, Activin/Nodal and BMP signaling. Development. 2008;135:2969–2979. doi: 10.1242/dev.021121. [DOI] [PubMed] [Google Scholar]
- 20.Otero JJ, Fu W, Kan L, et al. β-Catenin signaling is required for neural differentiation of embryonic stem cells. Development. 2004;131:3545–3557. doi: 10.1242/dev.01218. [DOI] [PubMed] [Google Scholar]
- 21.Emami KH, Nguyen C, Ma H, et al. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription [corrected] Proc Natl Acad Sci U S A. 2004;101:12682–12687. doi: 10.1073/pnas.0404875101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Miyabayashi T, Teo J-L, Yamamoto M, et al. Wnt/β-catenin/CBP signaling maintains long-term murine embryonic stem cell pluripotency. Proc Natl Acad Sci U S A. 2007;104:5668–5673. doi: 10.1073/pnas.0701331104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lukaszewicz AI, McMillan MK, Kahn M. Small molecules and stem cells. Potency and lineage commitment: The new quest for the fountain of youth. J Med Chem. 2010;53:3439–3453. doi: 10.1021/jm901361d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Miyabayashi T, Yamamoto M, Sato A, et al. Indole derivatives sustain embryonic stem cell self-renewal in long-term culture. Biosci Biotechnol Biochem. 2008;72:1242–1248. doi: 10.1271/bbb.70717. [DOI] [PubMed] [Google Scholar]
- 25.Hasegawa K, Zhang P, Wei Z, et al. Comparison of reprogramming efficiency between transduction of reprogramming factors, cell-cell fusion, and cytoplast fusion. Stem Cells. 2010;28:1338–1348. doi: 10.1002/stem.466. [DOI] [PubMed] [Google Scholar]
- 26.Hasegawa K, Fujioka T, Nakamura Y, et al. A method for the selection of human embryonic stem cell sublines with high replating efficiency after single-cell dissociation. Stem Cells. 2006;24:2649–2660. doi: 10.1634/stemcells.2005-0657. [DOI] [PubMed] [Google Scholar]
- 27.Willert K, Brown JD, Danenberg E, et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature. 2003;423:448–452. doi: 10.1038/nature01611. [DOI] [PubMed] [Google Scholar]
- 28.Veeman MT, Slusarski DC, Kaykas A, et al. Zebrafish prickle, a modulator of noncanonical Wnt/Fz signaling, regulates gastrulation movements. Curr Biol. 2003;13:680–685. doi: 10.1016/s0960-9822(03)00240-9. [DOI] [PubMed] [Google Scholar]
- 29.Xu C, Inokuma MS, Denham J, et al. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol. 2001;19:971–974. doi: 10.1038/nbt1001-971. [DOI] [PubMed] [Google Scholar]
- 30.Watanabe K, Ueno M, Kamiya D, et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 2007;25:681–686. doi: 10.1038/nbt1310. [DOI] [PubMed] [Google Scholar]
- 31.Chung TL, Turner JP, Thaker NY, et al. Ascorbate promotes epigenetic activation of CD30 in human embryonic stem cells. Stem Cells. 2010;28:1782–1793. doi: 10.1002/stem.500. [DOI] [PubMed] [Google Scholar]
- 32.Ellerström C, Strehl R, Moya K, et al. Derivation of a xeno-free human embryonic stem cell line. Stem Cells. 2006;24:2170–2176. doi: 10.1634/stemcells.2006-0130. [DOI] [PubMed] [Google Scholar]
- 33.Crook JM, Peura TT, Kravets L, et al. The generation of six clinical-grade human embryonic stem cell lines. Cell Stem Cell. 2007;1:490–494. doi: 10.1016/j.stem.2007.10.004. [DOI] [PubMed] [Google Scholar]
- 34.Desbordes SC, Placantonakis DG, Ciro A, et al. High-throughput screening assay for the identification of compounds regulating self-renewal and differentiation in human embryonic stem cells. Cell Stem Cell. 2008;2:602–612. doi: 10.1016/j.stem.2008.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Damoiseaux R, Sherman SP, Alva JA, et al. Integrated chemical genomics reveals modifiers of survival in human embryonic stem cells. Stem Cells. 2009;27:533–542. doi: 10.1634/stemcells.2008-0596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kelly KF, Ng DY, Jayakumaran G, et al. β-Catenin enhances Oct-4 activity and reinforces pluripotency through a TCF-independent mechanism. Cell Stem Cell. 2011;8:214–227. doi: 10.1016/j.stem.2010.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Berge DT, Kurek D, Blauwkamp T, et al. Embryonic stem cells require Wnt proteins to prevent differentiation to epiblast stem cells. Nat Cell Biol. 2011;13:1070–1075. doi: 10.1038/ncb2314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mercer SE, Friedman E. Mirk/Dyrk1B: a multifunctional dual-specificity kinase involved in growth arrest, differentiation, and cell survival. Cell Biochem Biophys. 2006;45:303–315. doi: 10.1385/CBB:45:3:303. [DOI] [PubMed] [Google Scholar]
- 39.Takahashi-Yanaga F, Sasaguri T. GSK-3beta regulates cyclin D1 expression: a new target for chemotherapy. Cell Signal. 2008;20:581–589. doi: 10.1016/j.cellsig.2007.10.018. [DOI] [PubMed] [Google Scholar]
- 40.Park J, Song W-J, Chung KC. Function and regulation of Dyrk1A: towards understanding Down syndrome. Cell Mol Life Sci. 2009;66:3235–3240. doi: 10.1007/s00018-009-0123-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Marson A, Foreman R, Chevalier B, et al. Wnt signaling promotes reprogramming of somatic cells to pluripotency. Cell Stem Cell. 2008;3:132–135. doi: 10.1016/j.stem.2008.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]