Skip to main content
  • Research article
  • Open access
  • Published:

Dynamic transcription programs during ES cell differentiation towards mesoderm in serum versus serum-freeBMP4 culture

Abstract

Background

Expression profiling of embryonic stem (ES) cell differentiation in the presence of serum has been performed previously. It remains unclear if transcriptional activation is dependent on complex growth factor mixtures in serum or whether this process is intrinsic to ES cells once the stem cell program has been inactivated. The aims of this study were to determine the transcriptional programs associated with the stem cell state and to characterize mesoderm differentiation between serum and serum-free culture.

Results

ES cells were differentiated as embryoid bodies in 10% FBS or serum-free media containing BMP4 (2 ng/ml), and expression profiled using 47 K Illumina(R) Sentrix arrays. Statistical methods were employed to define gene sets characteristic of stem cell, epiblast and primitive streak programs. Although the initial differentiation profile was similar between the two culture conditions, cardiac gene expression was inhibited in serum whereas blood gene expression was enhanced. Also, expression of many members of the Kruppel-like factor (KLF) family of transcription factors changed dramatically during the first few days of differentiation. KLF2 and KLF4 co-localized with OCT4 in a sub-nuclear compartment of ES cells, dynamic changes in KLF-DNA binding activities occurred upon differentiation, and strong bio-informatic evidence for direct regulation of many stem cell genes by KLFs was found.

Conclusion

Down regulation of stem cell genes and activation of epiblast/primitive streak genes is similar in serum and defined media, but subsequent mesoderm differentiation is strongly influenced by the composition of the media. In addition, KLF family members are likely to be important regulators of many stem cell genes.

Background

Embryonic stem (ES) cells isolated from the inner cell mass of the early mammalian blastocyst-stage embryo retain pluripotency when cultured on mouse embryonic fibroblasts (MEFs) in the presence of leukemia inhibitory factor (LIF) [1]. In the absence of LIF and MEF attachment, ES cells spontaneously differentiate into multi-cellular aggregates termed embryoid bodies (EBs). Although the spatial complexity of organogenesis is not established during EB maturation, the dynamics of gene expression closely mimic those which characterize early stages of mouse development [2–5]. Thus, ES cell differentiation is an excellent model system for the discovery of genes involved in developmental processes.

Many studies have attempted to identify genes that define the stem cell state by mining for genes co-expressed in ES cells and various other stem cell populations such as hematopoietic and neural stem cells [6, 7], or ES cells and trophoblast stem cells [8]. Some of these studies have been criticized because they were unable to define a similar cohort of stem cell genes [9]. Also, this approach is likely to find co-expression of 'housekeeping' genes that are irrelevant to the stem cell state. More recently, dynamic expression profiling during ES cell differentiation has been undertaken using various EB culture techniques [10, 11] and the addition of exogenous growth factors [7]. Most of these studies followed differentiation for a short time period (up to 6–8 days) due to inherent limitations of liquid or adherent cultures cell differentiation systems.

Various platforms have been used to determine gene expression profiles, including differential display, SAGE [12] and slide micro-arrays, such as those provided by Affymetrix, Agilent and Compugen. In this study we employed Illumina® Sentrix Mouse-6 oligo bead arrays [13], which have ~47 K probes that are largely based on the MEEBO set of transcripts, plus ESTs discovered as part of the FANTOM2 transcriptome project [14], some alternatively employed exons, and a small subset of non-coding transcripts. This is a new, rich and sensitive platform for expression profiling [15].

ES cells were differentiated in methylcellulose cultures, which allowed consistent EB development over 16 days in either serum [2, 16] or defined serum-free media containing BMP4 (2 ng/ml), termed serum-freeB4L hereafter [17]. The dynamics of loss of stem cell gene expression and activation of epiblast and primitive streak gene expression programs were similar in serum and serum-freeB4L but QT clustering revealed a significant difference in mesoderm outcomes from day 6 onwards. In particular, cardiac gene expression was greater in serum-freeB4L whereas activation of the blood program was enhanced in serum.

High expression levels of Kruppel-like factor (KLF) family members: Klf2, Klf4, Klf5 and Klf9 was detected in undifferentiated ES cells, and confirmed by quantitative real time RT-PCR, indirect immuno-fluorescence and DNA-binding assays. Expression was down regulated rapidly upon differentiation and a different set of Klfs: Klf3, Klf16 and Klf1/Eklf, were activated upon ES cell differentiation. Also, KLF-binding sites were markedly over represented in the proximal promoters of most stem cell specific genes, suggesting possible direct regulation by KLFs. We suggest a model in which the balance between self-renewal and differentiation is regulated by competitive occupancy of the proximal promoters of key stem cell genes such as Zfp42/Rex-1, Nanog, Pou5f1(Oct4), Lefty1 and Lefty2, and others such as the Klf genes themselves.

Results

Differentiation of ES cells in serum and serum-freeB4L culture

The directed differentiation of embryonic stem cells into desirable cell types will depend upon the addition of specific growth factors to defined media and reproducible physical conditions of culture. To establish a baseline for such studies, we undertook a comparison of murine ES cell differentiation using an embryoid body (EB) methylcellulose culture system containing either 10% FBS [2] or a chemically defined BSA-based media containing 2 ng/ml of BMP4, termed serum-freeB4L [17]. Triplicate experiments were performed in which ES cells were harvested after feeder depletion, and set up in parallel EB cultures in serum versus serum-freeB4L. RNA was collected at regular intervals up to 16 days, quantitative RT-PCR was performed on a panel of genes representing defined stages of development to establish the robustness of the culture system (Figure 1), and expression profiling was performed using Illumina® Sentrix Mouse-6 bead arrays (see Methods and [13]).

Figure 1
figure 1

Dynamic gene expression during EB differentiation in serum and serum-free media containing BMP4 [2 ng/ml]. Expression of stem cell, primitive streak and late mesoderm genes were analysed by quantitative RT-PCR in undifferentiated ES cells (0, grey bar) and in EBs collected for up to 16 days in serum (white bars) or defined media (black bars) from the same starting ES cell populations. Bars represent the means of three (serum) or two (serum-freeB4L) biological replicates. The Y-axis represents a log scale normalized relative to the housekeeping gene HPRT. Error bars indicate standard deviation.

Changes in gene expression were extremely reproducible across ES differentiation experiments with overall correlation of global gene expression for biological replicates in the order or 0.95–0.96. Approximately 30% of the probes were differentially expressed at one or more stages of EB differentiation as determined by one-way Welch ANOVA with multiple testing corrections using the Benjamini and Hochberg False Discovery Rate algorithm (7,967 probes with a p-value of < 0.01 by Welch t-test) (Figure 2A).

Figure 2
figure 2

Gene expression profiles during 16 days of EB differentiation in serum and serum-free media containing BMP4 [2 ng/ml]. (A) GeneSpring representation of the subset of genes from the 48 K Illumina array that showed significant differences in gene expression at 2 or more time points. Each line represents the mean normalised expression of an individual Illumina probe. Colours represent relative expression (red 4× increased; green 0.25× reduced) compared to a gene mean of 1 (black) with expression in undifferentiated ES cells (day 0) as the decision point for colour coding. The expression profiles of T (brachyury), Hba (α-globin), and Igf2 are indicated for reference. (B) Clustering (tree) of the genes that showed significant (p < 0.01) changes in gene expression upon differentiation by One-way Welch ANOVA. Expression levels are heat map colour coded (inset) with high expression at Day 0 coloured red and low expression coloured green.

To mine for genes with similar patterns of expression across the time course, hierarchical clustering was performed on the 7,967 probes and represented graphically. A large percentage of these (~30%) displayed highest expression in undifferentiated ES cells and subsequent reduced expression over time (labeled stem in Figure 2B). A second major cluster showed marked induction after ~day 4–6 of differentiation (labeled mesoderm in Figure 2B).

Putative stem cell genes sets

Previous studies have identified genes important to the stem cell state by comparing expression profiling data from different stem cell populations such as ES cells, haematopoietic stem cells (HSCs) and neural stem cells [6, 18]. We used two alternative approaches to identify specific stem cell specific gene expression. Firstly, genes with dynamic expression patterns which correlated closely with Oct4, Sox2 or Nanog expression profiles (Pearson correlation > 0.9) were determined as previously described [6, 7]. Second, genes rapidly down regulated ≥3 fold by day 3 of differentiation, a time point where ES cell clonogenicity decreases dramatically, were determined. Not surprisingly there was very little overlap between these gene lists, primarily because Oct4 and Nanog expression persists during EB differentiation longer than many other stem cell genes (Figure 1). Full lists of genes ranked according to similarity of expression (Pearson correlation >0.9) with expression patterns for Oct4, Nanog or Sox2 are provided in Additional files 1, 2, 3, respectively.

Only fifty-nine genes were down regulated ≥3 fold by day 3 of EB differentiation (Table 1). Many of these have been identified as stem cell markers by other groups using alternative strategies [6, 7, 19]. Information was compiled on their known expression patterns and/or gene knockout phenotypes via literature searches as referenced in Table 1. Insufficient space exists to discuss every gene and most have been previously identified as ES cell enriched transcripts. Osteopontin is secreted by osteoblasts in the bone marrow niche where it is thought to play an important role in the maintenance of HSC quiescence and maintenance of 'stemness' [20]. It has also been discovered by other groups as a strong marker of ES cells and is regulated directly by the key stem cell transcription factors, Oct4 and Sox2 [21]. Thus, osteopontin is likely to be a useful marker of ES cell pluripotency and may play autocrine or paracrine roles in maintenance of stemness. The second most rapidly down regulated gene was estrogen related receptor β, Esrrb, which had previously been identified as a stem cell specific gene and shown to be critical for maintenance of the stem cell state [19]. F-box 15 (Fbxo15), a member of the large F-box family of genes [22] was also rapidly down regulated. Fbxo15 is known to be highly expressed in ES cells, is regulated by Oct4 and Sox2, but is dispensable for ES cell self-renewal [23]. Recently, the stem cell specificity of this locus was used to screen for cDNAs capable of transforming mouse fibroblasts into ES cell populations [24]. Other notable rapidly down regulated genes included the nodal inhibitors Lefty-1 and Lefty-2, and Zfp42/Rex-1, which are also well established as stem cell markers [19].

Table 1 Genes rapidly down regulated (>3 fold in 3 days) upon ES cell differentiation

Oct4, Nanog and Sox2 are not included in Table 1, even though each is required for the maintenance of ES cell pluripotency [25–27] due to their persistent expression during the first 3 days of EB differentiation (Figure 1 and 5A, and Additional file 1). Indeed, Oct4 expression is known to persist beyond the inner cell mass stage of development in vivo, with transcripts detected in the epiblast and later in the primitive streak, before it becomes further restricted to primordial germ cells [10, 28]. 779 genes with a similar pattern of expression to Oct4 (Pearson correlation of > 0.9) were identified (Additional file 1). As expected this list also includes many well known stem cell genes such as Sal-like 1, nodal, chromobox homolog 7 (Cbx7), Dnmt3b and Sprouty homolog 4 [19]. We also mined for stem cell genes by looking for similarity (Pearson correlation of > 0.9) with Nanog and Sox2 gene expression profiles. These provided overlapping but not identical sets of putative stem cell specific genes (Additional files 2, 3). Full data for the 16 days of differentiation can be interrogated and mined for specific differentiation outcomes via Signet (Login_Bruce) (see Methods).

Tightly co-ordinated waves of gene expression suggest sequential activation of epiblast, primitive streak and mesoderm differentiation programs

Many alternative differentiation programs occur simultaneously during ES cell differentiation. Therefore, it is difficult to find gene syn-expression patterns that identify specific programs using standard clustering methods. An alternative approach is to search for expression patterns that closely resemble genes of known function. For example, the T box gene, brachyury, is a well characterized specific marker of the primitive streak [29]. Brachyury expression is transient in vivo, a feature recapitulated during ES cell differentiation [2]. Using qRT-PCR, brachyury was induced ~1000 fold from D0 to day 4 and then rapidly silenced to baseline levels by D6 (Figure 1). The dynamics of brachyury expression were similar between serum and serum-freeB4L media (Figure 1 and 3A), but serum-free media was unable to support brachyury induction in the absence of BMP-4 (data not shown). Thus, BMP4 (or related factors in serum such as activin) is essential for mesoderm generation. Thirty genes were identified with a similar expression pattern to brachyury (Pearson correlation >0.9) (Table 2) including many genes with established expression or critical functions during primitive streak formation, such as Mix1 [30], Lim1 [31], Sp5 [32] and eomesodermin [33]. Transient expression of many of these 'streak-specific' genes was validated by qRT-PCR (Figure 3B and data not shown). In all cases there was a close correlation between the qRT-PCR results and the profiling data (Figure 3A).

Figure 3
figure 3

Mining for genes expressed in patterns consistent with primitive streak, and epiblast differentiation programs. (A) GeneSpring representation of epiblast and streak gene expression profiles during EB differentiation in 10% serum or serum-freeB4L culture. The up-regulation of genes between days 2–4 were similar under both conditions. (B) qRT-PCR analysis of Mixl, Lim1, Cdx4 and Riken clone 8430415E04Rik. The Y-axis represents expression relative to the housekeeping gene HPRT. Error bars indicate ± SD from three biological replicates. (C) 102 genes transiently up regulated during the first 4 days of ES cell differentiation were clustered using a tree algorithm and Pearson correlation of > 0.9. The 'Brief' group represents gene with very transient expression at day 3. (D) Coding region for 8430415E04Rik. The shaded areas represent the three Heat domains.

Table 2 Brachyury-like (Primitive Streak) gene list (Pearson correlation >0.9 with brachyury)

Interestingly, four novel transcripts were identified in this list (Table 2). One of these, 8430415E04Rik, was detected by two independent probes, strongly supporting its 'streak-specific' expression; by qRT-PCR there was ~50 fold induction from day 2 to day 3 of differentiation and a decrease to near baseline levels by day 6 (Figure 3B). The gene contains 24 exons, and the full length cDNA encodes for a protein of 868 amino acids (Figure 3D) which is highly conserved in vertebrates (Additional file 4). It has little homology to other proteins apart from three predicted HEAT domains or armadillo domains (grey shading in Figure 3D). Drosophila armadillo interacts with β-catenin via its HEAT/armadillo repeat [34], which suggests the protein encoded by 8430415E04Rik may also be involved in protein-protein interactions.

The list of primitive streak genes in table 2 is smaller than that previously published [10] due to the very stringent criteria used to determine brachyury-like patterns of expression. Thus, to find further genes characteristic of epiblast, early and late primitive streak programs, probes were identified which were transiently up and down regulated at least two fold with peak expression at day 2, 3 or 4 of EB differentiation. Gene lists were combined (102 Illumina probes) and clustered into epiblast, early, mid and late streak groups plus a unique group in which expression was very transient at day 3 of differentiation (designated 'brief' at the bottom of Figure 3C). Also, lists were generated by searching for similarity of dynamic expression profiles to known genes which are accepted specific markers of specific stages of mouse development. For example, Fgf5 is specifically expressed in the epiblast [35], and Wnt5a is expressed in the late primitive streak [36]. The Fgf5-like (epiblast) list includes the methyltransferase, Dnmt3b, which was massively up-regulated in the first 2 days of EB differentiation (Additional file 5) and is known to be essential for mesoderm differentiation [37]. This list also includes Pim2, Zic2, Wnt8a, the SP1 (and possibly SP5) cofactor Crsp2, and Irf1. The Wnt5a-like (late primitive streak) list contains 76 genes including many homeobox genes (Hoxb2, Hoxd1, Hoxb6 and Hox8/Msx2), Cdx2, Hand1, Tbx3, Bmp4 and the VEGF receptor Flt1 (Additional file 6). Many are expressed at E10 of mouse development in the remnants of the primitive streak [36], suggesting the Wnt5a-like list includes many genes known to pattern the embryo following formation of the germ layers. There is a significant overlap between the genes we identified as being epiblast-, early and late streak-specific and those recently identified using a similar EB differentiation system and Affymetrix arrays [10], and by other groups [2, 38]. In addition, there are addition novel RIKEN cDNAs in these lists, which are likely to encode for interesting proteins worthy of further study (Additional files 5, 6).

Following the primitive streak wave of gene expression there was a dramatic activation of many genes (Figure 2A). Many of the most highly induced genes, such as globin genes, heme synthesis enzymes and haematopoietic transcription factors, are known to be expressed in the blood. qRT-PCR for globin genes confirmed > 10,000 fold induction between days 3 and 8 with a steady decline thereafter (Figure 1). Other mesoderm and endoderm programs are also activated at this stage, so it is difficult to mine for these different outcomes.

Enhanced cardiac gene expression and reduced erythropoiesis in serum-freeB4L EB culture

Overall there were very strong similarities between ES cell differentiation in serum and serum-free cultures supplemented with 2 ng/ml BMP4. For example, all 29 of the primitive streak gene set, as defined from serum-containing cultures, were also expressed transiently and to a similar level in serum-freeB4L media (Figure 3A). Genes differentially expressed at each EB time point (Day 1–16) were pooled and subjected to QT clustering to find sets differentially expressed between serum and serum-freeB4L EB culture. Using a minimal cut-off of five genes per group, nine gene sets were identified; seven showed higher expression in serum and two showed higher expression in serum-freeB4L (Table 3 and Figure 4).

Table 3 Genes differentially expressed during ES cell differentiation in serum versus serum-free defined media (BMP4 2 ng/ml)
Figure 4
figure 4

QT clustering revealed sets of genes differentially expressed between serum and serum-freeB4L EB culture. QT clustering revealed nine sets of genes with similar dynamic expression profiles (Pearson Correlation coefficient of > 0.9). In sets 1–7, expression is higher in serum (black sets). In sets 8–9, expression is greater in serum-freeB4L EB culture (red sets). Some sets have been named based on the similarity of overall gene function during differentiation pathways (see Table 3).

The genes in set 9 were expressed more highly in serum-freeB4L media from day 6, and genes in set 8 showed up-regulation at day 12 and 16 in the absence of serum (Figure 4). Both sets are comprised almost exclusively of cardiac-specific genes such as myosin heavy chain 7 (Myh7), myosin light chain 7 (Myl7), the cardiac isoform of α-actin (Actc1), troponin (Tnnc2), tropomysin (Tpm2), the nicotinic cholinergic receptor (Chrng), and cardiac muscle fast twitch 1 specific ATPase (Atp2a1) (Table 3). Further interrogation of the array data revealed a cohort of cardiac transcription factors, including ISL1, MEF2C, Bop/Smyd1 and Hand2 [39], which were transiently up regulated 1.4 to 3.5 fold in serum-freeB4L media compared to serum at day 6 (Signet Login_Bruce). Importantly, this elevated cardiac gene expression program correlated with increased numbers of spontaneously beating EBs in serum-freeB4L media (data not shown). Enhanced expression (~100 fold) of Actc1, Myl4 and Myl7 in serum-freeB4L media compared with serum-containing media was confirmed by qRT-PCR (Figure 1). Taken together, this data strongly suggests cardiac muscle cell differentiation is either inhibited in serum or enhanced by the addition of BMP4 to serum-free culture.

The genes in set 2 were expressed in both serum and serum-freeB4L conditions immediately following primitive streak formation, but expressed to higher levels and slightly earlier in serum (Table 3 and Figure 4). Many of these genes are globins (both embryonic and definitive) suggesting erythropoiesis is activated more robustly in the presence of serum (See Discussion). Alas2, a gene encoding the erythroid specific isoform of the first and rate limiting enzyme in the heme biosynthesis pathway, and the erythroid specific transcription factor, p45-NF-E2 are also in set 2 [40]. qRT-PCR for α- and βh1-globin confirmed more rapid expression in serum versus SFB4L media (Figure 1), suggesting factors in addition to BMP4 enhance activation of the blood program from a similar primitive streak platform.

The genes in set 3 were expressed at a similar time in serum but expressed at very low levels or not at all in serum-freeB4L media. Again, this set is highly enriched for erythroid specific genes such as alpha hemoglobin stabilising protein (Ahsp) [41], Glycophorin A, erythroid Kruppel-like factor (EKLF/Klf1) [42] and mitochondrial solute carrier protein (mitoferrin) [43] (Figure 4 and Table 3). Genes in sets 1 and 5 are activated more robustly at late time points in serum cultures. Most of these are specifically expressed in macrophages. Together, these results suggest delayed and less robust primitive haematopoiesis occurs in SFB4L culture compared with serum, an observation confirmed by less robust erythroid cell generation and 'redness' of the EBs from day 6–10 of differentiation (data not shown).

Rapid changes in Kruppel-like factor gene expression upon ES cell differentiation

Three Kruppel-like factor genes, Klf2, Klf4 and Klf5, were rapidly down regulated during the first few days of EB development (bold type in Table 1). All are members of the KLF family of transcription factors which are characterized by a similar C-terminal domain of three C2H2 zinc fingers which confers binding to CACC box elements in promoters and more distant regulatory elements [44, 45]. Klf2 and Klf4 have N-terminal transcriptional action domains and act primarily as transcriptional activators, whereas Klf5 is best known as a transcriptional repressor [46]. Until recently, Klf2 and Klf4 were considered to have restricted expression patterns [47, 48] with gene knockout animals showing late developmental defects in vasculogenesis and skin differentiation, respectively [49, 50]. Recently however, Klf4 expression was noted in ES cells [19, 51], and forced expression was shown to enhance Oct4 expression and promote self-renewal [51].

qRT-PCR confirmed the rapid down regulation of Klf 2, Klf4 and Klf5 in both serum and serum-freeB4L differentiation conditions, although Klf5 expression was down regulated more slowly (Figure 5A and 5B, and Table 4). Klf2, Klf4 and Klf5 were all re-induced from day 3–5 of differentiation as mesoderm and endoderm developmental pathways are activated (Figure 5A and 5B). Sixteen of the 17 known KLFs were examined in detail throughout the entire differentiation time course (Table 4). Klf9 was also steadily down regulated (>10 fold) between ES cells and day 5 EBs in serum and serum-freeB4L media (Figure 5B). Klf3, a well characterized transcriptional repressor [52] via its ability to recruit the co-repressor, CtBP [53], displayed an interesting biphasic expression pattern with initial rapid down regulation then re-induction from days 5–6 (Figure 5B). In contrast, Klf16 displayed little change in expression levels over the 16 days of EB maturation (Figure 5B).

Table 4 Kruppel-like factor gene expression during ES cell differentiation
Figure 5
figure 5

Kruppel like factors (Klfs) are dynamically expressed during the first few days of ES cell differentiation. (A) GeneSpring plot of normalized gene expression for all KLFs detected during ES cell differentiation in serum or serum-freeB4L culture. Most of the genes are listed on the y-axis in order from their highest relative expression in ES cells. There is dramatic up regulation of Klf1 (Eklf) only in serum following day 6 of differentiation. Plots representing Nanog (red), Sox2 (red) and Pou5f1(Oct4) (green) are shown for comparison. Pou5f1 and Nanog gene expression persists at high levels for 2–3 days after Klf2, Klf4 and Klf5 are down regulated. (B) Validation of changes in gene expression of six members of the KLF family by quantitative real time RT-PCR. Scheme as described for Figure 2B.

Klf2 and Klf4 co-localize with OCT4 in the nuclei of ES cells (Figure 6A). Interestingly, all three proteins were found to preferentially reside in a nuclear sub-compartment (possibly nucleoli for transcription factories), suggesting possible co-involvement in a protein complex or network. Also, endogenous KLF2 was detected by specific CACC box DNA-binding activity in undifferentiated ES cells, which was lost upon EB differentiation (Figure 6B). In contrast, endogenous ES cell KLF4 DNA binding activity was not detected using the p18INK4c promoter CACC box sequence, although recombinant KLF4 was shown to bind to this probe (data not shown). SP1 and SP3 are ubiquitously expressed CACC-box binding proteins, which are dominant in ES cell nuclear extracts (Figure 6B). We hypothesize KLF2, KLF3, KLF4, KLF5 and KLF6 compete with SP1 family members for DNA binding to key cis elements in various stem cell genes to regulate expression in positive and negative fashions (see Discussion).

Figure 6
figure 6

Kruppel like factor expression and DNA binding activity during ES cell differentiation. (A) Co-expression of Oct4 with KLF2 or KLF4 in ES cells. Indirect immuno-fluorescence shows co-localization of KLF2 and KLF4 with Oct4 in sub-nuclear compartments (possibly nucleoli). Individual confocal images for OCT4, KLF2, KLF4, and DAPI are shown with the corresponding composite image. Scale bar 40 μm. (B) Electro-mobility gel shift assay showing changes in DNA binding activities at a conserved CACC box site in the p18-INK4c gene promoter. Nuclear extracts were generated from ES cells or EBs differentiated for four days in serum. Super-shifts were performed with specific antisera for SP1, SP3, KLF2, KLF3, and KLF4 (See Methods). There is strong binding of endogenous Sp1 to the CACC element in ES cells and EB cells. KLF2 DNA-binding activity is present in ES cells as determined by a specific inhibition of binding of the indicated DNA complex with a KLF2 antibody. This activity is lost upon differentiation into EBs. The identity of the CACC box binding activity in EBs denoted CAC-X, and the binding activity in ES cells denoted CAC-Y, was not definitively identified using this panel of antibodies.

Enrichment of KLF-binding cis elements in the proximal promoters of stem cell specific genes

To investigate whether Kruppel-like factors might directly regulate stem cell gene expression, and therefore play a role in the maintenance or loss of 'stemness', the presence of KLF transcription factor binding sites (TFBS) within the proximal promoters of all genes in Table 1 was determined. Although a position weighted matrix (PWM) for KLF4 binding site specificity has been published [54], this site does not resemble the CACC box sequences known to bind other KLFs. Since KLF4 has identical DNA-binding residues in each of its three C-terminal C2H2 zinc fingers to other members of the Kruppel-like factor family [45], it should bind to similar sequences. Based on the crystal structure of the zinc fingers of Zif268 and SP1 bound to DNA [55, 56], and testing of specific binding of CACC box variants to recombinant KLF1, KLF3 and KLF17 [45, 52, 57, 58], a generic KLF TFBS has been proposed. Although a SELEX experiment for KLF1 binding has not been published, experiments from our group suggest a C at position 1, an A or C at position 3, an A at position 5, and an A, T or C at position 9 of the 9 bp consensus provides enhanced affinity for KLF1 binding. Taken together, these studies have enabled the development of a PWM (called here KLF-A) that should predict KLF4, KLF2 and other KLF TFBSs (see Methods and Additional file 7).

Clover [59] was used to determine if defined TFBSs were statistically over-represented in the stem cell lists. We searched for PWMs for KLF-A, KLF4 [54], octamer, extended octamer, nanog, and Oct-Sox binary sites as well as negative control sites (gata-1 and E-box) in sequences 2 kb upstream of the TSSs of stem cell genes (Table 1), versus 2 kb of sequences upstream of the entire murine transcriptome (See Additional file 7 for details of PWMs). There was significant over-representation of KLF-A TFBS within the first 2 kb of promoter sequences from the stem list gene list (p value of < 0.01) (Figure 7A), as well as over-representation of octamer sites and extended Oct4-binding sites as defined from ChIP-PET data [60]. No statistically significant over-representation of KLF4 [54], NANOG, GATA1 or E-box TFBSs were identified. All genes in the rapidly down regulated gene list have at least one KLF-A type CACC site, or octamer or Oct4 [60] TFBS. A full list of the promoters with highlighted sites is available on request. Together, this data strongly suggests direct transcriptional regulation of many stem cell genes by KLFs.

Figure 7
figure 7

KLF and octamer binding sites are highly enriched in stem cell gene promoters. (A) A Clover analysis was used to identify over-represented transcription factor binding sites within the promoter sequences of all stem cell genes identified in Table 1. Representative gene promoters are shown, indicating KLF-A binding sites (pink), octamer sites (ATGCWAAT) (green) and extended Oct4 binding sites (Oct4-Loh)(cyan) [60]. (B) Clover output for 2 kb of promoter sequence of murine Zfp42/Rex1. The positions and sequences corresponding to PWMs for KLF-A, octamer and Oct4 occupancy sites are indicated in the table and colour coded in the sequence. The positions are relative to the transcriptional start site. (C) ECR Browser output of conserved sequence identity between mouse and rat in the promoter and part of the first intron of the Zfp42 gene. The blue arrows indicate the direction of transcription. Yellow indicates the extent of the first (non-coding) exon, pink indicates regions of sequence conservation in the first intron and red indicates regions of sequence conservation in the 5' upstream region. rVISTA was used to find all of the KLF-A and octamer sites in the murine gene (Murine only) and conserved sites between mouse and rat (Conserved mouse, rat).

The Zfp42/Rex1 gene promoter is presented as an example of output from the Clover program (Figure 7B). It contains five KLF-A, one octamer and three OCT4 TFBS. The two CACC box elements within the first 250 base pairs of the TSS represent classical extended KLF-binding sites [42, 61]. These regions are the same as previously reported [62] although their functional importance has not been determined. Interestingly, two of the three KLF-A TFBS were evolutionarily conserved between the mouse and rat gene promoters (Figure 7C). Also, two further conserved CACC sites at -1.4 and -1.5 kb were identified within 500 bp of extended evolutionary conservation (>70% identity) (Figure 7C). This might act as a KLF dependent enhancer. Most of the stem cell genes in list 1 have conserved CACC box elements in their promoters [63–66]. In some cases these have been reported to bind the ubiquitous SP1, but our Clover analysis suggests they are also likely to bind KLFs (see Discussion).

Discussion

Expression profiling of murine ES cell differentiation was undertaken over a 16-day time course. We compared gene expression in methylcellulose cultures containing serum versus chemically defined media containing LIF (1 U/ml) and low concentrations of BMP4 (2 ng/ml) [67]. The Illumina® Sentrix Mouse 6 bead array provided a sensitive and detailed platform for analysis of dynamic gene expression. Using various data mining approaches, lists of stem cell-enriched genes and genes that are induced during the in vitro equivalent of epiblast and primitive streak stages of differentiation were generated. In combination with other ES cell profiling studies [7], our detailed expression data provides a useful resource for future reverse genetic approaches (i.e. siRNA knockdown) to study the function of these genes during ES cell differentiation and in vivo development. We also found a number of previously uncharacterized cDNAs (RIKEN clones) which could play important roles during development.

Importantly, the loss of pluripotency, measured by Oct4 gene expression, was comparable between serum and serum-freeB4L EB culture, following a predicted decrease over the first 6 days. Surprisingly, Oct4 gene expression gradually increased following day 6, an observation independent of the cell lines used (data not shown). It remains undetermined if expansion of Oct4 positive ES cells persists as undifferentiated populations within our EB culture system. Previous studies have identified the development of Oct4 positive primordial germ cells (PGC) following 12 days of ES cell differentiation [68–70], suggesting the Oct4 profile may alternatively represent the expansion of non-ES cell populations. Although the expansion of some undifferentiated ES cells is possible, the array profile does not suggest global persistence or up-regulation of ES gene expression late in the EB program. Also, markers of mesoderm induction such as brachyury and Mixl, do not show persistent up-regulation following peak expression at day 4, suggesting EBs are unlikely to harbour cells which are delayed or arrested from entering the differentiation program.

We found significant differences in cardiac gene expression during EB differentiation in serum and serum-freeB4L culture. Our current understanding of cardiac development provides possible insights into this observation. Briefly, in vivo studies have revealed BMPs secreted from the anterior lateral plate, and ill-defined signals from anterior primitive endoderm, are key inducers of cardiac development [71–73]. The administration of recombinant BMP2 or BMP4 to chick explant cultures induces cardiac differentiation in non-cardiogenic mesoderm [71] and Bmp2 knockout animals develop cardiac abnormalities [74]. Conversely, Wnts secreted by the neural tube are strong suppressors of cardiogenesis. Together, these opposing signals act to establish the borders of the heart field [71, 75]. By mimicking the environment that establishes cardiogenesis in vivo, assays capable of directing cardiomyocyte production from ES cells have been established. BMPs can efficiently enhance the cardiac program when added to EB culture [76, 77], whereas BMP inhibition drastically suppresses this outcome. In our hands cultures supplemented with BMP4 (2 ng/ml) supported cardiomyocyte maturation (as determined by expression of cardiac specific genes) with greatest expression detected at Day 8–10. This correlated with increased numbers of spontaneously beating EBs, and the timing of initial spontaneous contractions during murine embryogenesis [78]. The cardiac program was significantly reduced in culture containing 10% serum, an observation supported by a number of other studies [79, 80]. This suggests the constituents of serum are inhibitory to cardiomyocyte development. Although a detailed assessment of cardiomyocyte differentiation was not the focus of this analysis, the array output, qRT-PCR profiles and morphological observations described supports the usefulness of this assay in future investigations. In addition, the defined constituents of serum-freeB4L media provides an excellent opportunity to identify additional recombinant factors required to further expand cardiac progenitor cell production from EBs.

In contrast, the hematopoietic program was more pronounced within EBs grown in 10% serum than BMP4 alone (Figure 1 and 4). In particular, EKLF (Klf1) expression was significantly reduced in serum-free media supplemented with BMP4 (2 ng/ml) compared with serum (Figure 5A and Table 4). EKLF is essential for regulation of a large cohort of erythroid specific genes [81, 82], many of which were identified in sets 2 and 3 (Figure 4). Thus, it is likely that reduced EKLF expression in SFB4L media directly results in inhibition of a cascade of erythroid gene expression. Previous work showed the addition of BMP (at 5 ng/ml) to serum-free ES cell culture induces EKLF expression and restores hematopoietic cell differentiation [83]. Although a weak hematopoietic program was observed in SFB4L culture, the concentration of BMP4 used was less than that used by Adelman et al. Thus, we suggest a robust cardiac (anterior-ventral mesoderm) gene expression program is induced by low conentrations of BMP4 (or other BMPs), whereas a robust blood gene expression program (posterior-ventral mesoderm) requires >2 ng/ml of BMP4 or additional growth factors. Recently, mesoderm derived progenitor cell populations for hematopoietic and cardiac lineages were studied during EB development using ES cells in which GFP is targeted to the brachyury locus [80]. Within day 3.25 EBs, GFP+Flk1+ cells were shown to represent hematopoietic precursors, whereas GFP+Flk1- cells were significantly enriched for cardiac progenitor populations. It would be interesting to determine if the ratio of Flk1+/Flk1- within the brachyury positive population is altered between our two culture conditions.

A number of the Kruppel-like factor family of transcription factors were dynamically expressed in the first few days of ES cell differentiation. It was initially surprising to find highly enriched Klf2 and Klf4 expression in ES cells, since both are considered markers of terminally differentiated cell types such as skin [50], gut [84], vascular smooth muscle [49, 85] and lymphocytes [86]. However, recent evidence suggests KLFs may regulate stem cell function, since Klf4 is enriched in ES cells [19] and forced over-expression within these cells can maintain pluripotency in the absence of LIF [51]. Furthermore, Klf4 can bind the Lefty1 gene core promoter co-operatively with Oct4 and Sox2 [65]. Although Lefty 1 and 2 are best characterized as repressors of nodal, acting to regulate left-right patterning [87], our results and those of others, suggest a possible redundant role for lefty proteins during the maintenance of ES cell pluripotency [65]. Similarly to the Oct4 expression profile, many of the KLF family members also increased in expression late in the differentiation program. As mentioned, the KLFs are expressed in diverse tissues during development. It is therefore expected that the profiles obtained reflect the generation and early specification of mesoderm and endoderm cells following primitive streak gene activation at day 4.

Based on the likely identical binding specificity of the KLF family, established transcriptional activation and repression roles of certain family members [44], and bioinformatic evidence of a high prevalence of KLF TFBSs in many stem cell genes, we propose KLF competition for occupancy of these CACC box elements might determine self-renewal versus differentiation of ES cells. According to this model, high levels of KLF2 and KLF4 expression in undifferentiated ES cells would lead to occupancy of CACC box elements in promoters of stem cell genes such as Pou5f1/Oct4, Nanog, Esrrb, Zfp42/Rex1, Lefty1 and Lefty2. Furthermore, the Klf2 and Klf4 genes themselves have CACC box elements in their proximal promoters (Figure 7A), suggesting a positive feedback loop within ES cells is likely. Interestingly, gene knockout and lentiviral shRNA gene knockdown of Klf2 or Klf4, does not lead to an obvious stem cell defect [19, 49, 50, 85], suggesting these two KLFs may have functionally redundant roles during the maintenance of pluripotency. Future analysis of KLF2/KLF4 double knockout ES lines or the knockdown of both proteins using RNAi technologies will be necessary to validate this hypothesis. Upon ES cell differentiation, down regulation of Klf2 and Klf4 was very rapid, whereas down regulation of Oct4 and Nanog was delayed for one to two days. We suggest loss of KLF2 and KLF4 binding to Pouf1/Oct4, Nanog, Esrrb and perhaps other stem cell promoters, could be directly responsible for their down regulation. Moreover, Klf5 was down regulated more slowly, and Klf3 was up-regulated in the first two days of differentiation, suggesting these KLFs may function primarily as transcriptional repressors at the Pouf1/Oct4, nanog, Zfp42 and Esrrb gene promoters. Once this differentiation driving transcription network is activated we suggest KLF3, KLF5 and KLF9 can accelerate loss of the stem cell state by directly repressing expression of the Klf2 and Klf4 genes themselves. Therefore, a combination of cross regulation of transcriptional outputs, and competition for occupancy of key stem cell promoters by KLF proteins with differing biochemical properties but identical DNA-binding capacities, may determine self-renewal versus differentiation outcomes. Again validation of this model requires functional analysis. It is important to note we have not proven occupancy of stem cell gene promoters by the KLFs in vivo. Chromatin immuno-precipitation (ChIP) experiments in ES cells and differentiated populations are essential to validate this model, however ChiP grade antibodies are not currently available.

This scenario is not necessarily limited to ES cell differentiation. A similar program may take place in many adult stem and progenitor cells populations. Certainly competition between Klf4 and Klf5 has been suggested to regulate cell growth [88], and Klf1 and Klf3 compete for binding and have opposing effects in blood cells [89]. Furthermore, an imbalance between occupancy of key stem cell or proliferation gene promoters by KLFs, with differing activating and repressing functions, could be responsible for the development or progression of many common forms of cancer. For example, loss of hetero-zygosity for Klf4 and Klf5 has been found in colon, stomach, breast, prostate and liver cancers [90, 91].

In addition to the proposed complex transcriptional interplay between KLFs and stem cell gene promoters, direct protein-protein interactions and protein networks involving KLFs are possible. We showed sub-nuclear localization of KLF2 and KLF4 with OCT4 in ES cells (Figure 6A), and KLF4 can co-operate with OCT4 and SOX2 to drive expression of the Lefty1 core promoter [65]. Also, very recent biochemical purification of NANOG-interacting proteins discovered KLF4 as part of a NANOG-OCT4 network in ES cells (Stuart Orkin, personal communication). This is also consistent with the ability of KLF4 to co-operate with OCT4, SOX2 and c-MYC to drive de-differentiation of fibroblasts into ES-like cells [24].

Conclusion

Defining the genetic regulation of ES cell self renewal and differentiation will be instrumental for the development of future cell based therapies. Using 47 K Illumina® Sentrix bead arrays, the differential expression of genes during 16 days of ES cell differentiation was determined. Hierarchical gene clustering and correlation statistical analyses lead to the identification of a small cohort of genes which define the stem cell state. Historically, ES cell differentiation is achieved in culture media containing 10–15% FBS. A direct comparison between ES cell differentiation in serum and a serum-free media was undertaken. Surprisingly, global gene expression profiles were comparable between culture conditions, with the exception of mesoderm derived cardiac and haematopoietic transcripts. These results support the use of defined serum-free approaches for the directed differentiation of ES cell commitment. Expression of many KLF family members were enriched in ES cells and rapidly down regulated upon differentiation. KLF2 and KLF4 were co-localisation with OCT4 in ES cell nuclei and KLF transcription factor binding sites were over-represented within the promoters of the putative ES cell gene list (p < 0.01). Taken together, this data strongly suggests KLF family members regulate the maintenance of ES cell pluripotency.

Methods

ES cell culture, embryoid body formation and immunohistochemistry

W9.5 ES cell differentiation was performed as described (Bruce et al., Differentiation, in press). For immuno-fluorescence, feeder depleted W9.5 ES cells were seeded onto sterile gelatin-coated glass cover slips in ES cell media, fixed in 4% PFA for 10 min then washed in PBS. Cover slips were boiled in 10 mM citric acid (pH 6.4) for 10 min, washed in PBS and permeabilized in 0.18% triton X-100 in PBS for 10 mins. For OCT4/KLF4 detection, the cover slips were blocked in 1% BSA then incubated with rabbit polyclonal antibody raised against OCT4 (Abcam #ab19857) and goat Anti-mouse KLF4 (R&D Systems #AF3158) overnight at 4°C, then incubated in goat anti-rabbit Alexa-488 (1:400) (Molecular Probes #A11008) and donkey anti-goat Alexa-647 (1:400) for 1 hour. For co-detection of OCT4/KLF2, the Zenon Rabbit IgG labelling kit was used (Molecular Probes #Z-25308). 1 μg of KLF2 rabbit polyclonal antibody (Santa Cruz SC-28675) was labeled with Alexa647 Fab fragment following the manufacturer's instructions. Cover slips already stained for OCT4 (Alexa488) were incubated with labeled KLF2 for 1 hr then washed and post-fixed in 4% PFA to ensure strong signal detection. DAPI 1:5000 (Molecular Probes #D3571) was used to detect nuclear localization. Cover slips were mounted on SuperFrost® Plus slides (Menzel-Glaser) with VectaShield mounting media (Vector Laboratories H-1000). Confocal microscopy was performed on LSM 510 META Carl Zeiss microscope system.

RNA extraction, cDNA synthesis and real-time RT-PCR data analysis

Total RNA was made using TRIzol (Invitrogen), and cDNA was synthesized from 2 μg of DNase1-treated total RNA using Superscript III (Invitrogen) and oligo-DT12–15 (Promega) according to the manufacturer's instructions. Quantitative RT-PCR (qRT-PCR) was carried out using the Applied Biosystems SYBR-green dye system and 7500 Real Time Cycler in 96-well plates. Cycling variables were as follows: 50°C for 2 minutes, 95°C for 10 minutes, then 40 cycles of 15-second denaturation at 95°C and 1-minute at appropriate annealing temperatures, optimized for each set of primers based on dissociation curves (Additional file 8). Expression levels were normalized to HPRT as determined from the ratio of delta CT values. Mean of relative expression ± SD was determined from three biological replicates. Sequencing of gel-purified amplicons was performed to ensure correct product amplification.

Micro-array hybridization and analysis

Expression profiling was performed using Sentrix Mouse-6 Expression BeadChip arrays from Illumina®. RNA was assessed for integrity using the Agilent Bioanalyzer 2100 and RNA integrity (RIN) scores above 9.5 were present in all samples. Amplification was performed with 500 ng of total RNA using the Illumina TotalPrep RNA Amplification kit (Ambion) with a 12 hour in vitro transcription reaction period. The quantity and quality of biotin-UTP incorporated cRNA was also assessed on the Agilent Bioanalyzer 2100. Amplified cRNA (1500 ng per array) was hybridized to Mouse-6.v1 BeadChip arrays according to the manufacturer guidelines and detected with Fluorolink Streptavidin-Cy3 (Amersham Biosciences). Arrays were scanned using the Illumina BeadStation Scanner. The raw intensity values obtained for the scanned array images were compiled using the proprietary BeadStudio v1.5.1.3 software and imported into GeneSpring GX v7.3.1 (Agilent). A mouse Illumina probe set was defined in the GeneSpring Workgroup using the Illumina targetIDs as the unique identifiers and annotated according to array content files supplied by illumina®. Data normalization was performed by first setting all measurements less than 0.01 to 0.01, then applying per chip normalization to the 50th percentile, and per gene normalization to the median.

From an interpretation which included the three FBS experiments, a non-parametric Welch ANOVA (where variances were not assumed equal) was performed on all 46,120 probes to find a subset of genes whose expression varied significantly throughout the differentiation time course. A Benjamini and Hochberg False Discovery Rate multiple testing correction was applied to reduce the number of false positives. This yielded a set of 7,967 probes that showed statistically significant (p < 0.05) differences in expression by Welch t-test. This approach simplified running the data mining algorithms to find syn-expression patterns by removing genes such as housekeeping genes which did not display differential expression, or genes which did not reach a threshold of expression at any time points.

Genome interpretations were generated in GeneSpring GXv7.3.1 (Agilent) in which the mean relative expression level for each gene was represented at each time point relative to the average of all time points. A number of statistical approaches were used to find syn-expression patterns. A Pearson correlation (between 0.9 and 1.0) was performed to find genes with similar expression profiles to Pouf1/Oct4, nanog, sox2, fgf5, wnt5 and brachyury. Hierarchical clustering was performed using the gene by gene and group by group 'tree' algorithm within the GeneSpring (Agilent) program. This compares each probe with every other in the lists to generate similarity statistics and a tree representation of similarity based on a Pearson correlation. Also, quality threshold (QT) clustering [92] was used to define distinct subsets of co-expressed genes. The entire data set for the experiments is available via GEO as well as via our instance of Genespring (contact corresponding author).

Electro-mobility gel shift assays (EMSA)

Nuclear extracts were made from undifferentiated ES cells and embryoid bodies grown for four days in 10% serum. Electromobility gel shift assays (EMSA) were performed as previously described [61] using double stranded oligonucleotide probes corresponding to an extended CACC site in the p18INK4c promoter (sense strand 5'-gttgggcggggcgtgggcggggcc-3') (Tallack, et al., submitted). Supershifts were performed with specific antibodies raised against SP1 (Santa Cruz SC-059x), SP3 (Santa Cruz SC-644x), KLF2 (Santa Cruz SC-28675), KLF3 [52] and KLF4 (Santa Cruz SC-12538x).

Bioinformatic searches for CACC sites and other over represented elements in promoters of stem cell genes

The data-mining tool Biomart (accessed via Ensembl release 36 of the mouse genome), was used to collate 2 kb of sequences upstream of the transcriptional start sites (TSSs) of all genes listed in Table 1. The Clover program, run via the MotifViz web interface [59], was used to search for over-representation of position weighted matrices (PWMs) for octamer [93], Oct4 [60], Oct-Sox [60], KLF4 [54], E-box, GATA-1 [94] and CACC (KLF-A) elements in these promoter sets. The PWMs are listed in Additional file 7. The ECR Browser [95] was used to determine phylogenetic conservation of cis elements between mouse, rat and human genomes.

Abbreviations

BSA:

bovine serum albumin

EB:

embryoid body

EMSA:

electromobility gel shift assay

ES:

embryonic stem

FBS:

fetal bovine serum

KL:

Fkruppel-like factor

LIF:

leukemia inhibitory factor

MEF:

mouse embryonic fibroblasts

PFA:

paraformaldehyde

PWM:

position weighted matrix

TFBS:

transcription factor binding sites.

References

  1. Evans MJ, Kaufman MH: Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981, 292 (5819): 154-156. 10.1038/292154a0.

    CAS  PubMed  Google Scholar 

  2. Keller G, Kennedy M, Papayannopoulou T, Wiles MV: Hematopoietic commitment during embryonic stem cell differentiation in culture. Mol Cell Biol. 1993, 13 (1): 473-486.

    CAS  PubMed Central  PubMed  Google Scholar 

  3. Vittet D, Prandini MH, Berthier R, Schweitzer A, Martin-Sisteron H, Uzan G, Dejana E: Embryonic stem cells differentiate in vitro to endothelial cells through successive maturation steps. Blood. 1996, 88 (9): 3424-3431.

    CAS  PubMed  Google Scholar 

  4. Bain G, Kitchens D, Yao M, Huettner JE, Gottlieb DI: Embryonic stem cells express neuronal properties in vitro. Dev Biol. 1995, 168 (2): 342-357. 10.1006/dbio.1995.1085.

    CAS  PubMed  Google Scholar 

  5. Rohwedel J, Maltsev V, Bober E, Arnold HH, Hescheler J, Wobus AM: Muscle cell differentiation of embryonic stem cells reflects myogenesis in vivo: developmentally regulated expression of myogenic determination genes and functional expression of ionic currents. Dev Biol. 1994, 164 (1): 87-101. 10.1006/dbio.1994.1182.

    CAS  PubMed  Google Scholar 

  6. Ramalho-Santos M, Yoon S, Matsuzaki Y, Mulligan RC, Melton DA: "Stemness": transcriptional profiling of embryonic and adult stem cells. Science. 2002, 298 (5593): 597-600. 10.1126/science.1072530.

    CAS  PubMed  Google Scholar 

  7. Ivanova NB, Dimos JT, Schaniel C, Hackney JA, Moore KA, Lemischka IR: A stem cell molecular signature. Science. 2002, 298 (5593): 601-604. 10.1126/science.1073823.

    CAS  PubMed  Google Scholar 

  8. Tanaka TS, Kunath T, Kimber WL, Jaradat SA, Stagg CA, Usuda M, Yokota T, Niwa H, Rossant J, Ko MS: Gene expression profiling of embryo-derived stem cells reveals candidate genes associated with pluripotency and lineage specificity. Genome Res. 2002, 12 (12): 1921-1928. 10.1101/gr.670002.

    CAS  PubMed Central  PubMed  Google Scholar 

  9. Vogel G: Stem cells. 'Stemness' genes still elusive. Science. 2003, 302 (5644): 371-10.1126/science.302.5644.371a.

    CAS  PubMed  Google Scholar 

  10. Hirst CE, Ng ES, Azzola L, Voss AK, Thomas T, Stanley EG, Elefanty AG: Transcriptional profiling of mouse and human ES cells identifies SLAIN1, a novel stem cell gene. Dev Biol. 2006, 293 (1): 90-103. 10.1016/j.ydbio.2006.01.023.

    CAS  PubMed  Google Scholar 

  11. Bouhon IA, Kato H, Chandran S, Allen ND: Neural differentiation of mouse embryonic stem cells in chemically defined medium. Brain Res Bull. 2005, 68 (1-2): 62-75. 10.1016/j.brainresbull.2005.08.022.

    CAS  PubMed  Google Scholar 

  12. Richards M, Tan SP, Tan JH, Chan WK, Bongso A: The transcriptome profile of human embryonic stem cells as defined by SAGE. Stem Cells. 2004, 22 (1): 51-64. 10.1634/stemcells.22-1-51.

    CAS  PubMed  Google Scholar 

  13. Kuhn K, Baker SC, Chudin E, Lieu MH, Oeser S, Bennett H, Rigault P, Barker D, McDaniel TK, Chee MS: A novel, high-performance random array platform for quantitative gene expression profiling. Genome Res. 2004, 14 (11): 2347-2356. 10.1101/gr.2739104.

    CAS  PubMed Central  PubMed  Google Scholar 

  14. Okazaki Y, Furuno M, Kasukawa T, Adachi J, Bono H, Kondo S, Nikaido I, Osato N, Saito R, Suzuki H, Yamanaka I, Kiyosawa H, Yagi K, Tomaru Y, Hasegawa Y, Nogami A, Schonbach C, Gojobori T, Baldarelli R, Hill DP, Bult C, Hume DA, Quackenbush J, Schriml LM, Kanapin A, Matsuda H, Batalov S, Beisel KW, Blake JA, Bradt D, Brusic V, Chothia C, Corbani LE, Cousins S, Dalla E, Dragani TA, Fletcher CF, Forrest A, Frazer KS, Gaasterland T, Gariboldi M, Gissi C, Godzik A, Gough J, Grimmond S, Gustincich S, Hirokawa N, Jackson IJ, Jarvis ED, Kanai A, Kawaji H, Kawasawa Y, Kedzierski RM, King BL, Konagaya A, Kurochkin IV, Lee Y, Lenhard B, Lyons PA, Maglott DR, Maltais L, Marchionni L, McKenzie L, Miki H, Nagashima T, Numata K, Okido T, Pavan WJ, Pertea G, Pesole G, Petrovsky N, Pillai R, Pontius JU, Qi D, Ramachandran S, Ravasi T, Reed JC, Reed DJ, Reid J, Ring BZ, Ringwald M, Sandelin A, Schneider C, Semple CA, Setou M, Shimada K, Sultana R, Takenaka Y, Taylor MS, Teasdale RD, Tomita M, Verardo R, Wagner L, Wahlestedt C, Wang Y, Watanabe Y, Wells C, Wilming LG, Wynshaw-Boris A, Yanagisawa M, Yang I, Yang L, Yuan Z, Zavolan M, Zhu Y, Zimmer A, Carninci P, Hayatsu N, Hirozane-Kishikawa T, Konno H, Nakamura M, Sakazume N, Sato K, Shiraki T, Waki K, Kawai J, Aizawa K, Arakawa T, Fukuda S, Hara A, Hashizume W, Imotani K, Ishii Y, Itoh M, Kagawa I, Miyazaki A, Sakai K, Sasaki D, Shibata K, Shinagawa A, Yasunishi A, Yoshino M, Waterston R, Lander ES, Rogers J, Birney E, Hayashizaki Y: Analysis of the mouse transcriptome based on functional annotation of 60,770 full-length cDNAs. Nature. 2002, 420 (6915): 563-573. 10.1038/nature01266.

    PubMed  Google Scholar 

  15. Verdugo RA, Medrano JF: Comparison of gene coverage of mouse oligonucleotide microarray platforms. BMC Genomics. 2006, 7: 58-10.1186/1471-2164-7-58.

    PubMed Central  PubMed  Google Scholar 

  16. Perkins AC: Enrichment of blood from embryonic stem cells in vitro. Reprod Fertil Dev. 1998, 10 (7-8): 563-572. 10.1071/RD98104.

    CAS  PubMed  Google Scholar 

  17. Wiles MV, Johansson BM: Embryonic stem cell development in a chemically defined medium. Exp Cell Res. 1999, 247 (1): 241-248. 10.1006/excr.1998.4353.

    CAS  PubMed  Google Scholar 

  18. Bhattacharya B, Miura T, Brandenberger R, Mejido J, Luo Y, Yang AX, Joshi BH, Ginis I, Thies RS, Amit M, Lyons I, Condie BG, Itskovitz-Eldor J, Rao MS, Puri RK: Gene expression in human embryonic stem cell lines: unique molecular signature. Blood. 2004, 103 (8): 2956-2964. 10.1182/blood-2003-09-3314.

    CAS  PubMed  Google Scholar 

  19. Ivanova N, Dobrin R, Lu R, Kotenko I, Levorse J, DeCoste C, Schafer X, Lun Y, Lemischka IR: Dissecting self-renewal in stem cells with RNA interference. Nature. 2006, 442 (7102): 533-538. 10.1038/nature04915.

    CAS  PubMed  Google Scholar 

  20. Nilsson SK, Johnston HM, Whitty GA, Williams B, Webb RJ, Denhardt DT, Bertoncello I, Bendall LJ, Simmons PJ, Haylock DN: Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. 2005, 106 (4): 1232-1239. 10.1182/blood-2004-11-4422.

    CAS  PubMed  Google Scholar 

  21. Botquin V, Hess H, Fuhrmann G, Anastassiadis C, Gross MK, Vriend G, Scholer HR: New POU dimer configuration mediates antagonistic control of an osteopontin preimplantation enhancer by Oct-4 and Sox-2. Genes Dev. 1998, 12 (13): 2073-2090.

    CAS  PubMed Central  PubMed  Google Scholar 

  22. Jin J, Cardozo T, Lovering RC, Elledge SJ, Pagano M, Harper JW: Systematic analysis and nomenclature of mammalian F-box proteins. Genes Dev. 2004, 18 (21): 2573-2580. 10.1101/gad.1255304.

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Tokuzawa Y, Kaiho E, Maruyama M, Takahashi K, Mitsui K, Maeda M, Niwa H, Yamanaka S: Fbx15 is a novel target of Oct3/4 but is dispensable for embryonic stem cell self-renewal and mouse development. Mol Cell Biol. 2003, 23 (8): 2699-2708. 10.1128/MCB.23.8.2699-2708.2003.

    CAS  PubMed Central  PubMed  Google Scholar 

  24. Takahashi K, Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006, 126 (4): 663-676. 10.1016/j.cell.2006.07.024.

    CAS  PubMed  Google Scholar 

  25. Nichols J, Zevnik B, Anastassiadis K, Niwa H, Klewe-Nebenius D, Chambers I, Scholer H, Smith A: Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell. 1998, 95 (3): 379-391. 10.1016/S0092-8674(00)81769-9.

    CAS  PubMed  Google Scholar 

  26. Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R: Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003, 17 (1): 126-140. 10.1101/gad.224503.

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A: Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003, 113 (5): 643-655. 10.1016/S0092-8674(03)00392-1.

    CAS  PubMed  Google Scholar 

  28. Rosner MH, Vigano MA, Ozato K, Timmons PM, Poirier F, Rigby PW, Staudt LM: A POU-domain transcription factor in early stem cells and germ cells of the mammalian embryo. Nature. 1990, 345 (6277): 686-692. 10.1038/345686a0.

    CAS  PubMed  Google Scholar 

  29. Wilkinson DG, Bhatt S, Herrmann BG: Expression pattern of the mouse T gene and its role in mesoderm formation. Nature. 1990, 343 (6259): 657-659. 10.1038/343657a0.

    CAS  PubMed  Google Scholar 

  30. Hart AH, Hartley L, Sourris K, Stadler ES, Li R, Stanley EG, Tam PP, Elefanty AG, Robb L: Mixl1 is required for axial mesendoderm morphogenesis and patterning in the murine embryo. Development. 2002, 129 (15): 3597-3608.

    CAS  PubMed  Google Scholar 

  31. Tsang TE, Shawlot W, Kinder SJ, Kobayashi A, Kwan KM, Schughart K, Kania A, Jessell TM, Behringer RR, Tam PP: Lim1 activity is required for intermediate mesoderm differentiation in the mouse embryo. Dev Biol. 2000, 223 (1): 77-90. 10.1006/dbio.2000.9733.

    CAS  PubMed  Google Scholar 

  32. Treichel D, Becker MB, Gruss P: The novel transcription factor gene Sp5 exhibits a dynamic and highly restricted expression pattern during mouse embryogenesis. Mech Dev. 2001, 101 (1-2): 175-179. 10.1016/S0925-4773(00)00544-X.

    CAS  PubMed  Google Scholar 

  33. Ciruna BG, Rossant J: Expression of the T-box gene Eomesodermin during early mouse development. Mech Dev. 1999, 81 (1-2): 199-203. 10.1016/S0925-4773(98)00243-3.

    CAS  PubMed  Google Scholar 

  34. Peifer M, Berg S, Reynolds AB: A repeating amino acid motif shared by proteins with diverse cellular roles. Cell. 1994, 76 (5): 789-791. 10.1016/0092-8674(94)90353-0.

    CAS  PubMed  Google Scholar 

  35. Haub O, Goldfarb M: Expression of the fibroblast growth factor-5 gene in the mouse embryo. Development. 1991, 112 (2): 397-406.

    CAS  PubMed  Google Scholar 

  36. Gofflot F, Hall M, Morriss-Kay GM: Genetic patterning of the developing mouse tail at the time of posterior neuropore closure. Dev Dyn. 1997, 210 (4): 431-445. 10.1002/(SICI)1097-0177(199712)210:4<431::AID-AJA7>3.0.CO;2-H.

    CAS  PubMed  Google Scholar 

  37. Okano M, Bell DW, Haber DA, Li E: DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999, 99 (3): 247-257. 10.1016/S0092-8674(00)81656-6.

    CAS  PubMed  Google Scholar 

  38. Chen Y, Amende I, Hampton TG, Yang Y, Ke Q, Min JY, Xiao YF, Morgan JP: Vascular endothelial growth factor promotes cardiomyocyte differentiation of embryonic stem cells. Am J Physiol Heart Circ Physiol. 2006, 291 (4): H1653-8. 10.1152/ajpheart.00363.2005.

    CAS  PubMed  Google Scholar 

  39. Black BL: Transcriptional pathways in second heart field development. Semin Cell Dev Biol. 2007, 18 (1): 67-76. 10.1016/j.semcdb.2007.01.001.

    CAS  PubMed Central  PubMed  Google Scholar 

  40. Andrews NC, Kotkow KJ, Ney PA, Erdjument-Bromage H, Tempst P, Orkin SH: The ubiquitous subunit of erythroid transcription factor NF-E2 is a small basic-leucine zipper protein related to the v-maf oncogene. Proc Natl Acad Sci U S A. 1993, 90 (24): 11488-11492. 10.1073/pnas.90.24.11488.

    CAS  PubMed Central  PubMed  Google Scholar 

  41. Kihm AJ, Kong Y, Hong W, Russell JE, Rouda S, Adachi K, Simon MC, Blobel GA, Weiss MJ: An abundant erythroid protein that stabilizes free alpha-haemoglobin. Nature. 2002, 417 (6890): 758-763. 10.1038/nature00803.

    CAS  PubMed  Google Scholar 

  42. Miller IJ, Bieker JJ: A novel, erythroid cell-specific murine transcription factor that binds to the CACCC element and is related to the Kruppel family of nuclear proteins. Mol Cell Biol. 1993, 13 (5): 2776-2786.

    CAS  PubMed Central  PubMed  Google Scholar 

  43. Shaw GC, Cope JJ, Li L, Corson K, Hersey C, Ackermann GE, Gwynn B, Lambert AJ, Wingert RA, Traver D, Trede NS, Barut BA, Zhou Y, Minet E, Donovan A, Brownlie A, Balzan R, Weiss MJ, Peters LL, Kaplan J, Zon LI, Paw BH: Mitoferrin is essential for erythroid iron assimilation. Nature. 2006, 440 (7080): 96-100. 10.1038/nature04512.

    CAS  PubMed  Google Scholar 

  44. Philipsen S, Suske G: A tale of three fingers: the family of mammalian Sp/XKLF transcription factors. Nucleic Acids Res. 1999, 27 (15): 2991-3000. 10.1093/nar/27.15.2991.

    CAS  PubMed Central  PubMed  Google Scholar 

  45. Perkins A: Erythroid Kruppel like factor: from fishing expedition to gourmet meal. Int J Biochem Cell Biol. 1999, 31 (10): 1175-1192. 10.1016/S1357-2725(99)00083-7.

    CAS  PubMed  Google Scholar 

  46. Suske G, Bruford E, Philipsen S: Mammalian SP/KLF transcription factors: bring in the family. Genomics. 2005, 85 (5): 551-556. 10.1016/j.ygeno.2005.01.005.

    CAS  PubMed  Google Scholar 

  47. Anderson KP, Kern CB, Crable SC, Lingrel JB: Isolation of a gene encoding a functional zinc finger protein homologous to erythroid Kruppel-like factor: identification of a new multigene family. Mol Cell Biol. 1995, 15 (11): 5957-5965.

    CAS  PubMed Central  PubMed  Google Scholar 

  48. Shields JM, Christy RJ, Yang VW: Identification and characterization of a gene encoding a gut-enriched Kruppel-like factor expressed during growth arrest. J Biol Chem. 1996, 271 (33): 20009-20017. 10.1074/jbc.271.33.20009.

    CAS  PubMed Central  PubMed  Google Scholar 

  49. Kuo CT, Veselits ML, Barton KP, Lu MM, Clendenin C, Leiden JM: The LKLF transcription factor is required for normal tunica media formation and blood vessel stabilization during murine embryogenesis. Genes Dev. 1997, 11 (22): 2996-3006.

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Segre JA, Bauer C, Fuchs E: Klf4 is a transcription factor required for establishing the barrier function of the skin. Nat Genet. 1999, 22 (4): 356-360. 10.1038/11926.

    CAS  PubMed  Google Scholar 

  51. Li Y, McClintick J, Zhong L, Edenberg HJ, Yoder MC, Chan RJ: Murine embryonic stem cell differentiation is promoted by SOCS-3 and inhibited by the zinc finger transcription factor Klf4. Blood. 2005, 105 (2): 635-637. 10.1182/blood-2004-07-2681.

    CAS  PubMed  Google Scholar 

  52. Crossley M, Whitelaw E, Perkins A, Williams G, Fujiwara Y, Orkin SH: Isolation and characterization of the cDNA encoding BKLF/TEF-2, a major CACCC-box-binding protein in erythroid cells and selected other cells. Mol Cell Biol. 1996, 16 (4): 1695-1705.

    CAS  PubMed Central  PubMed  Google Scholar 

  53. Turner J, Crossley M: Cloning and characterization of mCtBP2, a co-repressor that associates with basic Kruppel-like factor and other mammalian transcriptional regulators. Embo J. 1998, 17 (17): 5129-5140. 10.1093/emboj/17.17.5129.

    CAS  PubMed Central  PubMed  Google Scholar 

  54. Shields JM, Yang VW: Identification of the DNA sequence that interacts with the gut-enriched Kruppel-like factor. Nucleic Acids Res. 1998, 26 (3): 796-802. 10.1093/nar/26.3.796.

    CAS  PubMed Central  PubMed  Google Scholar 

  55. Pavletich NP, Pabo CO: Zinc finger-DNA recognition: crystal structure of a Zif268-DNA complex at 2.1 A. Science. 1991, 252 (5007): 809-817. 10.1126/science.2028256.

    CAS  PubMed  Google Scholar 

  56. Oka S, Shiraishi Y, Yoshida T, Ohkubo T, Sugiura Y, Kobayashi Y: NMR structure of transcription factor Sp1 DNA binding domain. Biochemistry. 2004, 43 (51): 16027-16035. 10.1021/bi048438p.

    CAS  PubMed  Google Scholar 

  57. Feng WC, Southwood CM, Bieker JJ: Analyses of beta-thalassemia mutant DNA interactions with erythroid Kruppel-like factor (EKLF), an erythroid cell-specific transcription factor. J Biol Chem. 1994, 269 (2): 1493-1500.

    CAS  PubMed  Google Scholar 

  58. van Vliet J, Crofts LA, Quinlan KG, Czolij R, Perkins AC, Crossley M: Human KLF17 is a new member of the Sp/KLF family of transcription factors. Genomics. 2006, 87 (4): 474-482. 10.1016/j.ygeno.2005.12.011.

    CAS  PubMed  Google Scholar 

  59. Fu Y, Frith MC, Haverty PM, Weng Z: MotifViz: an analysis and visualization tool for motif discovery. Nucleic Acids Res. 2004, 32 (Web Server issue): W420-3. 10.1093/nar/gkh426.

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, Bourque G, George J, Leong B, Liu J, Wong KY, Sung KW, Lee CW, Zhao XD, Chiu KP, Lipovich L, Kuznetsov VA, Robson P, Stanton LW, Wei CL, Ruan Y, Lim B, Ng HH: The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet. 2006, 38 (4): 431-440. 10.1038/ng1760.

    CAS  PubMed  Google Scholar 

  61. Perkins AC, Sharpe AH, Orkin SH: Lethal beta-thalassaemia in mice lacking the erythroid CACCC- transcription factor EKLF. Nature. 1995, 375 (6529): 318-322. 10.1038/375318a0.

    CAS  PubMed  Google Scholar 

  62. Ben-Shushan E, Thompson JR, Gudas LJ, Bergman Y: Rex-1, a gene encoding a transcription factor expressed in the early embryo, is regulated via Oct-3/4 and Oct-6 binding to an octamer site and a novel protein, Rox-1, binding to an adjacent site. Mol Cell Biol. 1998, 18 (4): 1866-1878.

    CAS  PubMed Central  PubMed  Google Scholar 

  63. Wu DY, Yao Z: Functional analysis of two Sp1/Sp3 binding sites in murine Nanog gene promoter. Cell Res. 2006, 16 (3): 319-322. 10.1038/sj.cr.7310040.

    CAS  PubMed  Google Scholar 

  64. Pesce M, Marin Gomez M, Philipsen S, Scholer HR: Binding of Sp1 and Sp3 transcription factors to the Oct-4 gene promoter. Cell Mol Biol (Noisy-le-grand). 1999, 45 (5): 709-716.

    CAS  Google Scholar 

  65. Nakatake Y, Fukui N, Iwamatsu Y, Masui S, Takahashi K, Yagi R, Yagi K, Miyazaki J, Matoba R, Ko MS, Niwa H: Klf4 cooperates with Oct3/4 and Sox2 to activate the Lefty1 core promoter in embryonic stem cells. Mol Cell Biol. 2006, 26 (20): 7772-7782. 10.1128/MCB.00468-06.

    CAS  PubMed Central  PubMed  Google Scholar 

  66. Hosler BA, Rogers MB, Kozak CA, Gudas LJ: An octamer motif contributes to the expression of the retinoic acid-regulated zinc finger gene Rex-1 (Zfp-42) in F9 teratocarcinoma cells. Mol Cell Biol. 1993, 13 (5): 2919-2928.

    CAS  PubMed Central  PubMed  Google Scholar 

  67. Johansson BM, Wiles MV: Evidence for involvement of activin A and bone morphogenetic protein 4 in mammalian mesoderm and hematopoietic development. Mol Cell Biol. 1995, 15 (1): 141-151.

    CAS  PubMed Central  PubMed  Google Scholar 

  68. Hubner K, Fuhrmann G, Christenson LK, Kehler J, Reinbold R, De La Fuente R, Wood J, Strauss JF, Boiani M, Scholer HR: Derivation of oocytes from mouse embryonic stem cells. Science. 2003, 300 (5623): 1251-1256. 10.1126/science.1083452.

    PubMed  Google Scholar 

  69. Clark AT, Bodnar MS, Fox M, Rodriquez RT, Abeyta MJ, Firpo MT, Pera RA: Spontaneous differentiation of germ cells from human embryonic stem cells in vitro. Hum Mol Genet. 2004, 13 (7): 727-739. 10.1093/hmg/ddh088.

    CAS  PubMed  Google Scholar 

  70. Lacham-Kaplan O, Chy H, Trounson A: Testicular cell conditioned medium supports differentiation of embryonic stem cells into ovarian structures containing oocytes. Stem Cells. 2006, 24 (2): 266-273. 10.1634/stemcells.2005-0204.

    PubMed  Google Scholar 

  71. Schultheiss TM, Burch JB, Lassar AB: A role for bone morphogenetic proteins in the induction of cardiac myogenesis. Genes Dev. 1997, 11 (4): 451-462. 10.1101/gad.11.4.451.

    CAS  PubMed  Google Scholar 

  72. Schultheiss TM, Xydas S, Lassar AB: Induction of avian cardiac myogenesis by anterior endoderm. Development. 1995, 121 (12): 4203-4214.

    CAS  PubMed  Google Scholar 

  73. Sugi Y, Lough J: Anterior endoderm is a specific effector of terminal cardiac myocyte differentiation of cells from the embryonic heart forming region. Dev Dyn. 1994, 200 (2): 155-162.

    CAS  PubMed  Google Scholar 

  74. Zhang H, Bradley A: Mice deficient for BMP2 are nonviable and have defects in amnion/chorion and cardiac development. Development. 1996, 122 (10): 2977-2986.

    CAS  PubMed  Google Scholar 

  75. Tzahor E, Lassar AB: Wnt signals from the neural tube block ectopic cardiogenesis. Genes Dev. 2001, 15 (3): 255-260. 10.1101/gad.871501.

    CAS  PubMed Central  PubMed  Google Scholar 

  76. Kawai T, Takahashi T, Esaki M, Ushikoshi H, Nagano S, Fujiwara H, Kosai K: Efficient cardiomyogenic differentiation of embryonic stem cell by fibroblast growth factor 2 and bone morphogenetic protein 2. Circ J. 2004, 68 (7): 691-702. 10.1253/circj.68.691.

    CAS  PubMed  Google Scholar 

  77. Hosseinkhani M, Hosseinkhani H, Khademhosseini A, Bolland F, Kobayashi H, Prat Gonzalez S: Bone morphogenetic protein-4 enhances cardiomyocyte differentiation of cynomolgus monkey ES cells in Knockout Serum Replacement medium. Stem Cells. 2006

    Google Scholar 

  78. Nishii K, Shibata Y: Mode and determination of the initial contraction stage in the mouse embryo heart. Anat Embryol (Berl). 2006, 211 (2): 95-100. 10.1007/s00429-005-0065-x.

    Google Scholar 

  79. Zandstra PW, Bauwens C, Yin T, Liu Q, Schiller H, Zweigerdt R, Pasumarthi KB, Field LJ: Scalable production of embryonic stem cell-derived cardiomyocytes. Tissue Eng. 2003, 9 (4): 767-778. 10.1089/107632703768247449.

    CAS  PubMed  Google Scholar 

  80. Kouskoff V, Lacaud G, Schwantz S, Fehling HJ, Keller G: Sequential development of hematopoietic and cardiac mesoderm during embryonic stem cell differentiation. Proc Natl Acad Sci U S A. 2005, 102 (37): 13170-13175. 10.1073/pnas.0501672102.

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Drissen R, von Lindern M, Kolbus A, Driegen S, Steinlein P, Beug H, Grosveld F, Philipsen S: The erythroid phenotype of EKLF-null mice: defects in hemoglobin metabolism and membrane stability. Mol Cell Biol. 2005, 25 (12): 5205-5214. 10.1128/MCB.25.12.5205-5214.2005.

    CAS  PubMed Central  PubMed  Google Scholar 

  82. Hodge D, Coghill E, Keys J, Maguire T, Hartmann B, McDowall A, Weiss M, Grimmond S, Perkins A: A global role for EKLF in definitive and primitive erythropoiesis. Blood. 2006, 107 (8): 3359-3370. 10.1182/blood-2005-07-2888.

    CAS  PubMed Central  PubMed  Google Scholar 

  83. Adelman CA, Chattopadhyay S, Bieker JJ: The BMP/BMPR/Smad pathway directs expression of the erythroid-specific EKLF and GATA1 transcription factors during embryoid body differentiation in serum-free media. Development. 2002, 129 (2): 539-549.

    CAS  PubMed  Google Scholar 

  84. Shie JL, Chen ZY, O'Brien MJ, Pestell RG, Lee ME, Tseng CC: Role of gut-enriched Kruppel-like factor in colonic cell growth and differentiation. Am J Physiol Gastrointest Liver Physiol. 2000, 279 (4): G806-14.

    CAS  PubMed  Google Scholar 

  85. Wani MA, Means RT, Lingrel JB: Loss of LKLF function results in embryonic lethality in mice. Transgenic Res. 1998, 7 (4): 229-238. 10.1023/A:1008809809843.

    CAS  PubMed  Google Scholar 

  86. Kuo CT, Veselits ML, Leiden JM: LKLF: A transcriptional regulator of single-positive T cell quiescence and survival. Science. 1997, 277 (5334): 1986-1990. 10.1126/science.277.5334.1986.

    CAS  PubMed  Google Scholar 

  87. Meno C, Shimono A, Saijoh Y, Yashiro K, Mochida K, Ohishi S, Noji S, Kondoh H, Hamada H: lefty-1 is required for left-right determination as a regulator of lefty-2 and nodal. Cell. 1998, 94 (3): 287-297. 10.1016/S0092-8674(00)81472-5.

    CAS  PubMed  Google Scholar 

  88. Ghaleb AM, Nandan MO, Chanchevalap S, Dalton WB, Hisamuddin IM, Yang VW: Kruppel-like factors 4 and 5: the yin and yang regulators of cellular proliferation. Cell Res. 2005, 15 (2): 92-96. 10.1038/sj.cr.7290271.

    CAS  PubMed Central  PubMed  Google Scholar 

  89. Funnell AP, Maloney CA, Thompson LJ, Keys J, Tallack M, Perkins AC, Crossley M: Erythroid Kruppel-like Factor Directly Activates the Basic Kruppel-like Factor Gene in Erythroid Cells. Mol Cell Biol. 2007

    Google Scholar 

  90. Wei D, Kanai M, Huang S, Xie K: Emerging role of KLF4 in human gastrointestinal cancer. Carcinogenesis. 2006, 27 (1): 23-31. 10.1093/carcin/bgi243.

    CAS  PubMed  Google Scholar 

  91. Bateman NW, Tan D, Pestell RG, Black JD, Black AR: Intestinal tumor progression is associated with altered function of KLF5. J Biol Chem. 2004, 279 (13): 12093-12101. 10.1074/jbc.M311532200.

    CAS  PubMed  Google Scholar 

  92. Heyer LJ, Kruglyak S, Yooseph S: Exploring expression data: identification and analysis of coexpressed genes. Genome Res. 1999, 9 (11): 1106-1115. 10.1101/gr.9.11.1106.

    CAS  PubMed Central  PubMed  Google Scholar 

  93. Stepchenko AG, Luchina NN, Polanovsky OL: Conservative Val47 residue of POU homeodomain: role in DNA recognition. FEBS Lett. 1997, 412 (1): 5-8. 10.1016/S0014-5793(97)00508-5.

    CAS  PubMed  Google Scholar 

  94. Merika M, Orkin SH: DNA-binding specificity of GATA family transcription factors. Mol Cell Biol. 1993, 13 (7): 3999-4010.

    CAS  PubMed Central  PubMed  Google Scholar 

  95. Ovcharenko I, Nobrega MA, Loots GG, Stubbs L: ECR Browser: a tool for visualizing and accessing data from comparisons of multiple vertebrate genomes. Nucleic Acids Res. 2004, 32 (Web Server issue): W280-6. 10.1093/nar/gkh355.

    CAS  PubMed Central  PubMed  Google Scholar 

  96. Liaw L, Birk DE, Ballas CB, Whitsitt JS, Davidson JM, Hogan BL: Altered wound healing in mice lacking a functional osteopontin gene (spp1). J Clin Invest. 1998, 101 (7): 1468-1478.

    CAS  PubMed Central  PubMed  Google Scholar 

  97. Luo J, Sladek R, Bader JA, Matthyssen A, Rossant J, Giguere V: Placental abnormalities in mouse embryos lacking the orphan nuclear receptor ERR-beta. Nature. 1997, 388 (6644): 778-782. 10.1038/42022.

    CAS  PubMed  Google Scholar 

  98. Rodda S, Sharma S, Scherer M, Chapman G, Rathjen P: CRTR-1, a developmentally regulated transcriptional repressor related to the CP2 family of transcription factors. J Biol Chem. 2001, 276 (5): 3324-3332. 10.1074/jbc.M008167200.

    CAS  PubMed  Google Scholar 

  99. Rogers MB, Hosler BA, Gudas LJ: Specific expression of a retinoic acid-regulated, zinc-finger gene, Rex-1, in preimplantation embryos, trophoblast and spermatocytes. Development. 1991, 113 (3): 815-824.

    CAS  PubMed  Google Scholar 

  100. Shi W, Wang H, Pan G, Geng Y, Guo Y, Pei D: Regulation of the pluripotency marker rex-1 by nanog and sox2. J Biol Chem. 2006, 281 (33): 23319-23325. 10.1074/jbc.M601811200.

    CAS  PubMed  Google Scholar 

  101. Edgar AJ: Molecular cloning and tissue distribution of mammalian L-threonine 3-dehydrogenases. BMC Biochem. 2002, 3: 19-10.1186/1471-2091-3-19.

    PubMed Central  PubMed  Google Scholar 

  102. Arima T, Hata K, Tanaka S, Kusumi M, Li E, Kato K, Shiota K, Sasaki H, Wake N: Loss of the maternal imprint in Dnmt3Lmat-/- mice leads to a differentiation defect in the extraembryonic tissue. Dev Biol. 2006, 297 (2): 361-373. 10.1016/j.ydbio.2006.05.003.

    CAS  PubMed  Google Scholar 

  103. Yoshikawa T, Piao Y, Zhong J, Matoba R, Carter MG, Wang Y, Goldberg I, Ko MS: High-throughput screen for genes predominantly expressed in the ICM of mouse blastocysts by whole mount in situ hybridization. Gene Expr Patterns. 2006, 6 (2): 213-224. 10.1016/j.modgep.2005.06.003.

    CAS  PubMed Central  PubMed  Google Scholar 

  104. Franz WM, Breves D, Klingel K, Brem G, Hofschneider PH, Kandolf R: Heart-specific targeting of firefly luciferase by the myosin light chain-2 promoter and developmental regulation in transgenic mice. Circ Res. 1993, 73 (4): 629-638.

    CAS  PubMed  Google Scholar 

  105. Tonini GP, Casalaro A, Cara A, Di Martino D: Inducible expression of calcyclin, a gene with strong homology to S-100 protein, during neuroblastoma cell differentiation and its prevalent expression in Schwann-like cell lines. Cancer Res. 1991, 51 (6): 1733-1737.

    CAS  PubMed  Google Scholar 

  106. Wang PJ, McCarrey JR, Yang F, Page DC: An abundance of X-linked genes expressed in spermatogonia. Nat Genet. 2001, 27 (4): 422-426. 10.1038/86927.

    PubMed  Google Scholar 

  107. Wulf P, Suter U: Embryonic expression of epithelial membrane protein 1 in early neurons. Brain Res Dev Brain Res. 1999, 116 (2): 169-180. 10.1016/S0165-3806(99)00092-9.

    CAS  PubMed  Google Scholar 

  108. Ko MS, Kitchen JR, Wang X, Threat TA, Hasegawa A, Sun T, Grahovac MJ, Kargul GJ, Lim MK, Cui Y, Sano Y, Tanaka T, Liang Y, Mason S, Paonessa PD, Sauls AD, DePalma GE, Sharara R, Rowe LB, Eppig J, Morrell C, Doi H: Large-scale cDNA analysis reveals phased gene expression patterns during preimplantation mouse development. Development. 2000, 127 (8): 1737-1749.

    PubMed  Google Scholar 

  109. Meno C, Takeuchi J, Sakuma R, Koshiba-Takeuchi K, Ohishi S, Saijoh Y, Miyazaki J, ten Dijke P, Ogura T, Hamada H: Diffusion of nodal signaling activity in the absence of the feedback inhibitor Lefty2. Dev Cell. 2001, 1 (1): 127-138. 10.1016/S1534-5807(01)00006-5.

    CAS  PubMed  Google Scholar 

  110. Sakaguchi T, Nishimoto M, Miyagi S, Iwama A, Morita Y, Iwamori N, Nakauchi H, Kiyonari H, Muramatsu M, Okuda A: Putative "stemness" gene jam-B is not required for maintenance of stem cell state in embryonic, neural, or hematopoietic stem cells. Mol Cell Biol. 2006, 26 (17): 6557-6570. 10.1128/MCB.00729-06.

    CAS  PubMed Central  PubMed  Google Scholar 

  111. Scott LM, Mueller L, Collins SJ: E3, a hematopoietic-specific transcript directly regulated by the retinoic acid receptor alpha. Blood. 1996, 88 (7): 2517-2530.

    CAS  PubMed  Google Scholar 

  112. Vogel W, Gish GD, Alves F, Pawson T: The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cell. 1997, 1 (1): 13-23. 10.1016/S1097-2765(00)80003-9.

    CAS  PubMed  Google Scholar 

  113. Mootha VK, Bunkenborg J, Olsen JV, Hjerrild M, Wisniewski JR, Stahl E, Bolouri MS, Ray HN, Sihag S, Kamal M, Patterson N, Lander ES, Mann M: Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria. Cell. 2003, 115 (5): 629-640. 10.1016/S0092-8674(03)00926-7.

    CAS  PubMed  Google Scholar 

  114. Wassarman KM, Lewandoski M, Campbell K, Joyner AL, Rubenstein JL, Martinez S, Martin GR: Specification of the anterior hindbrain and establishment of a normal mid/hindbrain organizer is dependent on Gbx2 gene function. Development. 1997, 124 (15): 2923-2934.

    CAS  PubMed  Google Scholar 

  115. Ikonomidis JS, Gibson WC, Butler JE, McClister DM, Sweterlitsch SE, Thompson RP, Mukherjee R, Spinale FG: Effects of deletion of the tissue inhibitor of matrix metalloproteinases-1 gene on the progression of murine thoracic aortic aneurysms. Circulation. 2004, 110 (11 Suppl 1): II268-73.

    PubMed  Google Scholar 

  116. Katz JP, Perreault N, Goldstein BG, Lee CS, Labosky PA, Yang VW, Kaestner KH: The zinc-finger transcription factor Klf4 is required for terminal differentiation of goblet cells in the colon. Development. 2002, 129 (11): 2619-2628.

    CAS  PubMed Central  PubMed  Google Scholar 

  117. Shindo T, Manabe I, Fukushima Y, Tobe K, Aizawa K, Miyamoto S, Kawai-Kowase K, Moriyama N, Imai Y, Kawakami H, Nishimatsu H, Ishikawa T, Suzuki T, Morita H, Maemura K, Sata M, Hirata Y, Komukai M, Kagechika H, Kadowaki T, Kurabayashi M, Nagai R: Kruppel-like zinc-finger transcription factor KLF5/BTEB2 is a target for angiotensin II signaling and an essential regulator of cardiovascular remodeling. Nat Med. 2002, 8 (8): 856-863.

    CAS  PubMed  Google Scholar 

  118. Falender AE, Lanz R, Malenfant D, Belanger L, Richards JS: Differential expression of steroidogenic factor-1 and FTF/LRH-1 in the rodent ovary. Endocrinology. 2003, 144 (8): 3598-3610. 10.1210/en.2002-0137.

    CAS  PubMed  Google Scholar 

  119. Vasseur S, Hoffmeister A, Garcia-Montero A, Barthet M, Saint-Michel L, Berthezene P, Fiedler F, Closa D, Dagorn JC, Iovanna JL: Mice with targeted disruption of p8 gene show increased sensitivity to lipopolysaccharide and DNA microarray analysis of livers reveals an aberrant gene expression response. BMC Gastroenterol. 2003, 3: 25-10.1186/1471-230X-3-25.

    PubMed Central  PubMed  Google Scholar 

  120. Xu H, Gonzalo JA, St Pierre Y, Williams IR, Kupper TS, Cotran RS, Springer TA, Gutierrez-Ramos JC: Leukocytosis and resistance to septic shock in intercellular adhesion molecule 1-deficient mice. J Exp Med. 1994, 180 (1): 95-109. 10.1084/jem.180.1.95.

    CAS  PubMed  Google Scholar 

  121. Kobayashi K, Hernandez LD, Galan JE, Janeway CA, Medzhitov R, Flavell RA: IRAK-M is a negative regulator of Toll-like receptor signaling. Cell. 2002, 110 (2): 191-202. 10.1016/S0092-8674(02)00827-9.

    CAS  PubMed  Google Scholar 

  122. McGowan KM, Coulombe PA: Onset of keratin 17 expression coincides with the definition of major epithelial lineages during skin development. J Cell Biol. 1998, 143 (2): 469-486. 10.1083/jcb.143.2.469.

    CAS  PubMed Central  PubMed  Google Scholar 

  123. Schubart UK, Yu J, Amat JA, Wang Z, Hoffmann MK, Edelmann W: Normal development of mice lacking metablastin (P19), a phosphoprotein implicated in cell cycle regulation. J Biol Chem. 1996, 271 (24): 14062-14066. 10.1074/jbc.271.24.14062.

    CAS  PubMed  Google Scholar 

  124. Takahashi Y, Dominici M, Swift J, Nagy C, Ihle JN: Trophoblast stem cells rescue placental defect in SOCS3-deficient mice. J Biol Chem. 2006, 281 (17): 11444-11445. 10.1074/jbc.C600015200.

    CAS  PubMed  Google Scholar 

  125. Jin W, Broedl UC, Monajemi H, Glick JM, Rader DJ: Lipase H, a new member of the triglyceride lipase family synthesized by the intestine. Genomics. 2002, 80 (3): 268-273. 10.1006/geno.2002.6837.

    CAS  PubMed  Google Scholar 

  126. Ishiguro H, Furukawa Y, Daigo Y, Miyoshi Y, Nagasawa Y, Nishiwaki T, Kawasoe T, Fujita M, Satoh S, Miwa N, Fujii Y, Nakamura Y: Isolation and characterization of human NBL4, a gene involved in the beta-catenin/tcf signaling pathway. Jpn J Cancer Res. 2000, 91 (6): 597-603.

    CAS  PubMed  Google Scholar 

  127. Jiang N, Wang X, Jhanwar-Uniyal M, Darzynkiewicz Z, Dai W: Polo box domain of Plk3 functions as a centrosome localization signal, overexpression of which causes mitotic arrest, cytokinesis defects, and apoptosis. J Biol Chem. 2006, 281 (15): 10577-10582. 10.1074/jbc.M513156200.

    CAS  PubMed  Google Scholar 

  128. Kim M, Geum D, Khang I, Park YM, Kang BM, Lee KA, Kim K: Expression pattern of HSP25 in mouse preimplantation embryo: heat shock responses during oocyte maturation. Mol Reprod Dev. 2002, 61 (1): 3-13. 10.1002/mrd.1125.

    CAS  PubMed  Google Scholar 

  129. Zhu M, Lovell KL, Patterson JS, Saunders TL, Hughes ED, Friderici KH: Beta-mannosidosis mice: a model for the human lysosomal storage disease. Hum Mol Genet. 2006, 15 (3): 493-500. 10.1093/hmg/ddi465.

    CAS  PubMed  Google Scholar 

  130. Sato M, Kimura T, Kurokawa K, Fujita Y, Abe K, Masuhara M, Yasunaga T, Ryo A, Yamamoto M, Nakano T: Identification of PGC7, a new gene expressed specifically in preimplantation embryos and germ cells. Mech Dev. 2002, 113 (1): 91-94. 10.1016/S0925-4773(02)00002-3.

    CAS  PubMed  Google Scholar 

  131. Maki JM, Rasanen J, Tikkanen H, Sormunen R, Makikallio K, Kivirikko KI, Soininen R: Inactivation of the lysyl oxidase gene Lox leads to aortic aneurysms, cardiovascular dysfunction, and perinatal death in mice. Circulation. 2002, 106 (19): 2503-2509. 10.1161/01.CIR.0000038109.84500.1E.

    PubMed  Google Scholar 

  132. Sitzmann J, Noben-Trauth K, Kamano H, Klempnauer KH: Expression of B-Myb during mouse embryogenesis. Oncogene. 1996, 12 (9): 1889-1894.

    CAS  PubMed  Google Scholar 

  133. Tang CJ, Lin CY, Tang TK: Dynamic localization and functional implications of Aurora-C kinase during male mouse meiosis. Dev Biol. 2006, 290 (2): 398-410. 10.1016/j.ydbio.2005.11.036.

    CAS  PubMed  Google Scholar 

  134. Beer HD, Bittner M, Niklaus G, Munding C, Max N, Goppelt A, Werner S: The fibroblast growth factor binding protein is a novel interaction partner of FGF-7, FGF-10 and FGF-22 and regulates FGF activity: implications for epithelial repair. Oncogene. 2005, 24 (34): 5269-5277. 10.1038/sj.onc.1208560.

    CAS  PubMed  Google Scholar 

  135. Razani B, Engelman JA, Wang XB, Schubert W, Zhang XL, Marks CB, Macaluso F, Russell RG, Li M, Pestell RG, Di Vizio D, Hou H, Kneitz B, Lagaud G, Christ GJ, Edelmann W, Lisanti MP: Caveolin-1 null mice are viable but show evidence of hyperproliferative and vascular abnormalities. J Biol Chem. 2001, 276 (41): 38121-38138.

    CAS  PubMed  Google Scholar 

  136. Beddington RS, Rashbass P, Wilson V: Brachyury--a gene affecting mouse gastrulation and early organogenesis. Dev Suppl. 1992, 157-165.

    Google Scholar 

  137. Sun X, Meyers EN, Lewandoski M, Martin GR: Targeted disruption of Fgf8 causes failure of cell migration in the gastrulating mouse embryo. Genes Dev. 1999, 13 (14): 1834-1846.

    CAS  PubMed Central  PubMed  Google Scholar 

  138. Saga Y, Hata N, Kobayashi S, Magnuson T, Seldin MF, Taketo MM: MesP1: a novel basic helix-loop-helix protein expressed in the nascent mesodermal cells during mouse gastrulation. Development. 1996, 122 (9): 2769-2778.

    CAS  PubMed  Google Scholar 

  139. Behrens J, Jerchow BA, Wurtele M, Grimm J, Asbrand C, Wirtz R, Kuhl M, Wedlich D, Birchmeier W: Functional interaction of an axin homolog, conductin, with beta-catenin, APC, and GSK3beta. Science. 1998, 280 (5363): 596-599. 10.1126/science.280.5363.596.

    CAS  PubMed  Google Scholar 

  140. Bouckson-Castaing V, Moudjou M, Ferguson DJ, Mucklow S, Belkaid Y, Milon G, Crocker PR: Molecular characterisation of ninein, a new coiled-coil protein of the centrosome. J Cell Sci. 1996, 109 ( Pt 1): 179-190.

    CAS  Google Scholar 

  141. Fehling HJ, Swat W, Laplace C, Kuhn R, Rajewsky K, Muller U, von Boehmer H: MHC class I expression in mice lacking the proteasome subunit LMP-7. Science. 1994, 265 (5176): 1234-1237. 10.1126/science.8066463.

    CAS  PubMed  Google Scholar 

  142. Cole TB, Wenzel HJ, Kafer KE, Schwartzkroin PA, Palmiter RD: Elimination of zinc from synaptic vesicles in the intact mouse brain by disruption of the ZnT3 gene. Proc Natl Acad Sci U S A. 1999, 96 (4): 1716-1721. 10.1073/pnas.96.4.1716.

    CAS  PubMed Central  PubMed  Google Scholar 

  143. Meissner A, Zilles O, Varona R, Jozefowski K, Ritter U, Marquez G, Hallmann R, Korner H: CC chemokine ligand 20 partially controls adhesion of naive B cells to activated endothelial cells under shear stress. Blood. 2003, 102 (8): 2724-2727. 10.1182/blood-2003-01-0007.

    CAS  PubMed  Google Scholar 

  144. Li Y, Chambers J, Pang J, Ngo K, Peterson PA, Leung WP, Yang Y: Characterization of the mouse proteasome regulator PA28b gene. Immunogenetics. 1999, 49 (3): 149-157. 10.1007/s002510050476.

    CAS  PubMed  Google Scholar 

  145. Ridley SH, Ktistakis N, Davidson K, Anderson KE, Manifava M, Ellson CD, Lipp P, Bootman M, Coadwell J, Nazarian A, Erdjument-Bromage H, Tempst P, Cooper MA, Thuring JW, Lim ZY, Holmes AB, Stephens LR, Hawkins PT: FENS-1 and DFCP1 are FYVE domain-containing proteins with distinct functions in the endosomal and Golgi compartments. J Cell Sci. 2001, 114 (Pt 22): 3991-4000.

    CAS  PubMed  Google Scholar 

  146. Ossendorp F, Fu N, Camps M, Granucci F, Gobin SJ, van den Elsen PJ, Schuurhuis D, Adema GJ, Lipford GB, Chiba T, Sijts A, Kloetzel PM, Ricciardi-Castagnoli P, Melief CJ: Differential expression regulation of the alpha and beta subunits of the PA28 proteasome activator in mature dendritic cells. J Immunol. 2005, 174 (12): 7815-7822.

    CAS  PubMed  Google Scholar 

  147. Mohan WS, Scheer E, Wendling O, Metzger D, Tora L: TAF10 (TAF(II)30) is necessary for TFIID stability and early embryogenesis in mice. Mol Cell Biol. 2003, 23 (12): 4307-4318. 10.1128/MCB.23.12.4307-4318.2003.

    CAS  PubMed  Google Scholar 

  148. Wagner DS, Gan L, Klein WH: Identification of a differentially expressed RNA helicase by gene trapping. Biochem Biophys Res Commun. 1999, 262 (3): 677-684. 10.1006/bbrc.1999.1208.

    CAS  PubMed  Google Scholar 

  149. Shah GN, Ulmasov B, Waheed A, Becker T, Makani S, Svichar N, Chesler M, Sly WS: Carbonic anhydrase IV and XIV knockout mice: roles of the respective carbonic anhydrases in buffering the extracellular space in brain. Proc Natl Acad Sci U S A. 2005, 102 (46): 16771-16776. 10.1073/pnas.0508449102.

    CAS  PubMed Central  PubMed  Google Scholar 

  150. Lau YF, Chan KM, Sparkes R: Male-enhanced antigen gene is phylogenetically conserved and expressed at late stages of spermatogenesis. Proc Natl Acad Sci U S A. 1989, 86 (21): 8462-8466. 10.1073/pnas.86.21.8462.

    CAS  PubMed Central  PubMed  Google Scholar 

  151. Kusumi K, Sun ES, Kerrebrock AW, Bronson RT, Chi DC, Bulotsky MS, Spencer JB, Birren BW, Frankel WN, Lander ES: The mouse pudgy mutation disrupts Delta homologue Dll3 and initiation of early somite boundaries. Nat Genet. 1998, 19 (3): 274-278. 10.1038/961.

    CAS  PubMed  Google Scholar 

  152. Spencer JA, Eliazer S, Ilaria RL, Richardson JA, Olson EN: Regulation of microtubule dynamics and myogenic differentiation by MURF, a striated muscle RING-finger protein. J Cell Biol. 2000, 150 (4): 771-784. 10.1083/jcb.150.4.771.

    CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge funding from Australian Stem Cell Centre (P021), Wesley Research Institute (2006_04), and scholarship support from Kidney Health Australia.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Andrew C Perkins.

Additional information

Competing interests

The author(s) declares that there are no competing interests.

Authors' contributions

SB established the EB differentiation assay, collected and purified RNA samples, designed and carried out the quantitative RT-PCR studies and drafted the manuscript. BG carried out the micro array protocol. LB Carried out experimentation of mesoderm specific gene expression. MG performed the array statistical analysis and annotation. SG participated in the design of the study and performed the statistical analysis. AP conceived the study, and participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.

Electronic supplementary material

12864_2007_1078_MOESM1_ESM.doc

Additional file 1: Oct4 gene list. The data provided lists all genes expressed during 16 days of embryoid body differentiation with similarity to Oct4 (Pearson correlation >0.9). (DOC 630 KB)

12864_2007_1078_MOESM2_ESM.doc

Additional file 2: Nanog gene list. The data provided lists all genes expressed during 16 days of embryoid body differentiation with similarity to Nanog (Pearson correlation >0.9). (DOC 111 KB)

12864_2007_1078_MOESM3_ESM.doc

Additional file 3: Sox2 gene list. The data provided lists all genes expressed during 16 days of embryoid body differentiation with similarity to Sox2 (Pearson correlation >0.9). (DOC 236 KB)

12864_2007_1078_MOESM4_ESM.doc

Additional file 4: Sequence alignment of RIKEN clone 8430415E04RIK. The data provides an alignment of RIKEN clone 8430415E04RIK using human, mouse, chick and zebrafish sequences. (DOC 30 KB)

12864_2007_1078_MOESM5_ESM.doc

Additional file 5: FGF5 gene list. The data provided lists all genes expressed during 16 days of embryoid body differentiation with similarity to FGF5 (Pearson correlation >0.9). (DOC 69 KB)

12864_2007_1078_MOESM6_ESM.doc

Additional file 6: Wnt5a gene list. The data provided lists all genes expressed during 16 days of embryoid body differentiation with similarity to Wnt5 (Pearson correlation >0.9). (DOC 76 KB)

12864_2007_1078_MOESM7_ESM.doc

Additional file 7: Position Weighted Matrices. Provides published data on position weighted matrices for Oct, Sox, KLF-A, KLF4, Nanog, E-box, Gata-1 and their variants. (DOC 116 KB)

12864_2007_1078_MOESM8_ESM.doc

Additional file 8: Oligonucleotide sequences used for qRT-PCR analysis. Lists the oligonucleotide sequences and cycling conditions used for the qRT-PCR analysis. (DOC 48 KB)

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Bruce, S.J., Gardiner, B.B., Burke, L.J. et al. Dynamic transcription programs during ES cell differentiation towards mesoderm in serum versus serum-freeBMP4 culture. BMC Genomics 8, 365 (2007). https://doi.org/10.1186/1471-2164-8-365

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2164-8-365

Keywords