Embryonic stem (ES) cells recapitulate normal developmental processes and serve as an attractive source for routine access to a large number of cells for research and therapies. We previously reported that ES cells cultured on M15 cells, or a synthesized basement membrane (sBM) substratum, efficiently differentiated into an endodermal fate and subsequently adopted fates of various digestive organs, such as the pancreas and liver. Here, we established a novel hepatic differentiation procedure using the synthetic nanofiber (sNF) as a cell culture scaffold. We first compared endoderm induction and hepatic differentiation between murine ES cells grown on sNF and several other substrata. The functional assays for hepatocytes reveal that the ES cells grown on sNF were directed into hepatic differentiation. To clarify the mechanisms for the promotion of ES cell differentiation in the sNF system, we focused on the function of Rac1, which is a Rho family member protein known to regulate the actin cytoskeleton. We observed the activation of Rac1 in undifferentiated and differentiated ES cells cultured on sNF plates, but not in those cultured on normal plastic plates. We also show that inhibition of Rac1 blocked the potentiating effects of sNF on endoderm and hepatic differentiation throughout the whole differentiation stages. Taken together, our results suggest that morphological changes result in cellular differentiation controlled by Rac1 activation, and that motility is not only the consequence, but is also able to trigger differentiation. In conclusion, we believe that sNF is a promising material that might contribute to tissue engineering and drug delivery.

The liver is an important organ that performs many complex functions, including the metabolism of carbohydrates, proteins and lipids, as well as storage of essential nutrients and biotransformation of drugs. Drug biotransformation involves detoxification and bioactivation, where the metabolite becomes more toxic. Therefore, drug biotransformation plays an important role in the early stages of drug discovery processes. Primary hepatocyte cultures are often used for pharmacological assays, but they are short-lived and cannot be maintained in long-term culture. In addition, there are considerable donor-dependent variations. By contrast, embryonic stem (ES) cells or induced pluripotent stem (iPS) cells can proliferate infinitely and maintain their pluripotent ability to differentiate into various cell types. There is evidence that ES or iPS cells recapitulate normal developmental processes, and can serve as an alternative resource for hepatological researches, drug development and clinical uses. Through our present knowledge of developmental biology, efficient induction of hepatic lineage cells has been established. For example, based on the evidence that TGFβ–activin–Smad2 signaling is involved in definitive endoderm formation in the mouse (Tremblay et al., 2000), the activation of Activin–Nodal signaling was used for endoderm induction (D'Amour et al., 2005; Kubo et al., 2004). Fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) were added for the specification of liver lineages (Jung, 1999; Mfopou et al., 2010; Shiraki et al., 2008a); this helped to mimic the mesodermal signals from the septum transversum mesenchyme in normal development (Katsumoto et al., 2010; Shin et al., 2007; Rossi et al., 2001). Because hepatocyte growth factors are known to be important effectors in the specification of cell fate and organogenesis of the liver (Schmidt et al., 1995; Sonnenberg et al., 1993), hepatocyte growth factor (HGF), dexamethasone and oncostatin M have been used for induction of hepatocyte maturation (Basma et al., 2009; Kamiya et al., 1999; Si-Tayeb et al., 2010). Compared with the factors described above that direct hepatic differentiation, the role of extracellular matrices (ECMs) and scaffolds remains unclear.

We have previously reported that culturing ES/iPS cells on the mesonephric M15 cell line, in the presence of specific growth factors, resulted in an efficient induction of endoderm-derived tissues, such as the liver or pancreas (Shiraki et al., 2008a; Shiraki et al., 2008b; Umeda et al., 2013). We further suggested that the basement membrane components, including lama5, play an important role in guiding the differentiation of ES cells into regional-specific lineages of the definitive endoderm (Higuchi et al., 2010). We also successfully established an alternative hepatic differentiation procedure without using feeder cells, but with a synthesized basement membrane (sBM) substratum (Higuchi et al., 2010; Shiraki et al., 2011). Together, these results revealed the importance of the ECM for differentiation of ES cells.

The basement membrane, a highly integrated three-dimensional structure composed of ECM molecules, is known to regulate various cellular processes. It is known that electrospun nanofibers provide not only three-dimensional microenvironments mimicking the ECM, but also appropriate guidance cues to modulate cell behavior. Here, we tested the effects of synthetic nanofiber (sNF) matrices on ES/iPS cell differentiation. We found that ES/iPS cells grown on the sNF were induced into endoderm and then hepatic fates. Overall, we conclude that the sNF is more potent in promoting hepatic differentiation, compared with the traditional two-dimensional culture surfaces, and is able to substitute for the sBM or M15 cells.

Differentiation of murine and human ES cells into the hepatic lineages on the sNF matrix

We first tested the sNF matrix for its potency to mimic the basement membrane substratum of the cells. Murine SK7 ES cells (Shiraki et al., 2008a) were seeded onto the sNF matrix, and allowed to differentiate into the hepatic lineage by sequential changes of medium containing specific growth factors (Fig. 1A). We found that the expression of the pluripotent marker Oct3/4 was downregulated, whereas the mesendoderm marker Gsc and definitive endoderm markers Sox17 and Foxa2 were expressed at day 4 (d4) of differentiation (Fig. 1B). Whereas Gsc was downregulated rapidly, Sox17 and Foxa2 showed peak expressions around d7 and were downregulated afterwards. Notably, the hepatic progenitor marker gene, alpha-fetoprotein (Afp), and the mature hepatocyte marker, albumin (Alb1) were detectable from d12 and d16, respectively, and their expression levels were increased with time. Although the Afp transcript level was decreased after d22, the Alb1 expression continued increasing beyond d22. The immunocytochemical analysis further confirms that ALB and AFP were present in the cytoplasm of differentiated ES cells (Fig. 1C). In addition, periodic-acid–Schiff (PAS) staining and the Indocyanin Green (ICG) test were also conducted to investigate the hepatocyte functions of differentiated ES cells. The former reflects glycogen storage by showing positive populations as magenta in the cytoplasm, and the latter is used to examine cellular uptake activities, which are regarded as a hepatocyte detoxification function. As shown in Fig. 1D, glycogen storage was observed as the accumulation of magenta staining in the cytoplasm of the differentiated cells (top panel) and the ICG test also shows a similar result (bottom panel).

Fig. 1.

Differentiation of murine ES cells into the hepatocyte lineage on nanofiber scaffolds. (A) Schematic diagram of the differentiation procedure for mouse ES cells. (B) Time-dependent expression levels of endoderm and hepatic marker genes. β-actin was used as a control. FL, fetal liver on embryonic day 12.5; P0, neonatal liver on postnatal day 0. (C) The immunochemical analysis of differentiated ES cells on day 22 (d22) for α-fetoprotein (AFP, green) and albumin (ALB, red) with nuclear counterstaining (DAPI). (D) Hepatocyte functional tests for PAS and ICG on d26 differentiated ES cells (top panels) and undifferentiated (bottom left and right panels) and d9 differentiated (bottom middle panel) ES cells as negative controls (bottom panels). Nuclei are counterstained with hematoxylin (blue). Scale bars: 250 µm.

Fig. 1.

Differentiation of murine ES cells into the hepatocyte lineage on nanofiber scaffolds. (A) Schematic diagram of the differentiation procedure for mouse ES cells. (B) Time-dependent expression levels of endoderm and hepatic marker genes. β-actin was used as a control. FL, fetal liver on embryonic day 12.5; P0, neonatal liver on postnatal day 0. (C) The immunochemical analysis of differentiated ES cells on day 22 (d22) for α-fetoprotein (AFP, green) and albumin (ALB, red) with nuclear counterstaining (DAPI). (D) Hepatocyte functional tests for PAS and ICG on d26 differentiated ES cells (top panels) and undifferentiated (bottom left and right panels) and d9 differentiated (bottom middle panel) ES cells as negative controls (bottom panels). Nuclei are counterstained with hematoxylin (blue). Scale bars: 250 µm.

We next investigated whether human ES or iPS cells could differentiate in the sNF system. We used khES3 human ES cells (supplementary material Fig. S1A–D), as well as human iPS cell lines, such as Toe (supplementary material Fig. S1E,G) and 201B7 (supplementary material Fig. S1F), and found that these cells were able to differentiate into hepatocyte-like cells, thereby producing ALB and taking up ICG (supplementary material Fig. S1D–G). Together, these results indicate that sNF is a suitable matrix for potentiating hepatic differentiation, not only in murine cells, but also in human ES cells and iPS cells.

sNF is more potent than normal plates precoated with other matrices

To compare the supportive effects of NFs and other substrata, we seeded murine ES cells onto either the sNF matrix or normal plates precoated with other stubstrata, including collagen I, Matrigel, gelatin and fibronectin and then performed the differentiation experiment as described in Fig. 1A. On differentiation day 22 (d22), quantitative PCR analyses were carried out to quantify the expression profiles of hepatic function marker genes in differentiated cells. Our results indicate that ES cells grown on sNF showed higher expression levels of proteins secreted by hepatocytes, such as serine peptidase inhibitor a1 (Serpina1), Ttr and Alb1, compared with those grown on other substrata (Fig. 2). Similar results were observed for the expression of several other genes, including glucose 6-phosphatase (G6p) and fatty-acid binding protein (Fabp1), or transporters, such as organic anion-transporting polypeptide 1 (Oatp1), Na+-taurocholate cotransporting polypeptide (Ntcp) and UDP-glucuronosyltransferase (Ugt1a1), as well as multidrug resistance-associated protein family proteins 2 and 3 (Mrp2 and Mrp3) (Fig. 2). By contrast, little change was found in the expression of glucose transporter 2 (Glut2).

Fig. 2.

Expression of hepatic markers in differentiated murine ES cells on sNF or other substrata. Expression levels of various gene transcripts quantified by real time PCR in d22 differentiated ES cells cultured on sNF, collagen I (Col), Matrigel (Mat), fibronectin (FN) or gelatin (Gel). ES, undifferentiated ES cells. FL (E12.5 fetal liver) and P0 (neonatal liver on postnatal day 0) are used as references. For differentiated ES cells, values represent mean ± s.e.m. (n = 3). *P<0.05 and **P<0.01, by one-way ANOVA with the post-hoc Dunnett's test.

Fig. 2.

Expression of hepatic markers in differentiated murine ES cells on sNF or other substrata. Expression levels of various gene transcripts quantified by real time PCR in d22 differentiated ES cells cultured on sNF, collagen I (Col), Matrigel (Mat), fibronectin (FN) or gelatin (Gel). ES, undifferentiated ES cells. FL (E12.5 fetal liver) and P0 (neonatal liver on postnatal day 0) are used as references. For differentiated ES cells, values represent mean ± s.e.m. (n = 3). *P<0.05 and **P<0.01, by one-way ANOVA with the post-hoc Dunnett's test.

To determine the hepatic functions of differentiated ES cells grown on Matrigel, collagen I or sNF, we next measured their ALB secretions, ICG uptake and cytochrome p450 (CYP) activities. Our results show that ES cells grown on sNF secreted ALB at a higher level compared with those on Matrigel or collagen I (Fig. 3A). By day 26, the percentage of ICG-positive ES cells on sNF was also higher than in ES cells grown on the other two substrates (Fig. 3B,C).

Fig. 3.

Liver functional assays of differentiated murine ES cells grown on nanofiber scaffolds versus other substrata. (A) ELISA analysis of time-dependent albumin secretion for 24 hours by ES cells grown on Matrigel (Mat), collagen I (Col) or NF. (B,C) ICG tests performed on d26 differentiatied ES cells. The percentage of cells taking up ICG in culture was calculated (B) and representative images are shown (C). Values represent means ± s.e.m. (n = 6). *P<0.05, **P<0.01, by two-tailed Student's t-test. Scale bar: 250 µm.

Fig. 3.

Liver functional assays of differentiated murine ES cells grown on nanofiber scaffolds versus other substrata. (A) ELISA analysis of time-dependent albumin secretion for 24 hours by ES cells grown on Matrigel (Mat), collagen I (Col) or NF. (B,C) ICG tests performed on d26 differentiatied ES cells. The percentage of cells taking up ICG in culture was calculated (B) and representative images are shown (C). Values represent means ± s.e.m. (n = 6). *P<0.05, **P<0.01, by two-tailed Student's t-test. Scale bar: 250 µm.

To measure CYP activities, the differentiated ES cells were treated with a CYP1A inducer, 3-methylcholantrene (3MC), for 48 hours during d22–d24 or d64–d66, as shown in supplementary material Fig. S2A. We found that the differentiated cells cultured on sNF had higher CYP1A1 activities and responses to the inducer than those cultured on fibronectin, Matrigel or collagen I (supplementary material Fig. S2B). We also assayed the effects of sNF on the maintenance of the mature hepatic cells. ES cells cultured on sNF were able to maintain their CYP1A1 activities and responses to 3MC even on d66, whereas those cultured on other matrices did not survive in long-term cultures (supplementary material Fig. S2C). It is also worth noting that the differentiated cells on sNF could be maintained in culture for more than 100 days. Specifically, we show that the ES cells cultured on sNF for 129 days were able to uptake and secrete ICG (supplementary material Fig. S2D). Based on these findings, we conclude that sNF is an excellent matrix, not only for the differentiation of ES cells into the hepatic lineage but also for maintaining the mature state of ES-cell-derived hepatocytes.

High Rac1 activities in undifferentiated and differentiated ES cells grown on sNF

Next, we investigated the effects of sNF on hepatic differentiation of ES cells. It was previously reported that the undifferentiated murine ES cells cultured on sNF exhibited spheroid morphologies and formed dome-like structures, and proliferated well (Nur-E-Kamal et al., 2006). Therefore, we checked the morphological changes between the undifferentiated and differentiated states of the murine ES cells. To exclude the effect of the extracellular matrix, we compared gelatin-coated normal two-dimensional (2D) plates with either gelatin-coated or uncoated sNF. We found that the undifferentiated ES cells grown on gelatin-coated 2D plates, with cytoplasmic spreading morphologies and attached to the plate surface in large areas (Fig. 4A, left). By contrast, the ES cells grown on sNF showed aggregated morphologies (Fig. 4A, middle and right).

Fig. 4.

NF induces Rac1 hydroxylation in both undifferentiated and day 9 differentiated murine ES cells. (A,B) Representative images of undifferentiated (A) and day 9 (d9) differentiated (B) ES cells grown on gelatin-precoated normal plates (2D-gel), gelatin-coated NF plates (NF-gel) and uncoated NF plates (NF). Insets are higher magnifications of the boxed regions. (C,D) Western blot analysis of GTP-bound active Rac1, total Rac1 and GAPDH expression in undifferentiated (C) or differentiated (D) ES cells described in A,B.

Fig. 4.

NF induces Rac1 hydroxylation in both undifferentiated and day 9 differentiated murine ES cells. (A,B) Representative images of undifferentiated (A) and day 9 (d9) differentiated (B) ES cells grown on gelatin-precoated normal plates (2D-gel), gelatin-coated NF plates (NF-gel) and uncoated NF plates (NF). Insets are higher magnifications of the boxed regions. (C,D) Western blot analysis of GTP-bound active Rac1, total Rac1 and GAPDH expression in undifferentiated (C) or differentiated (D) ES cells described in A,B.

In the differentiated state, the ES cells were found to form a monolayer on sNF. However, the morphological differences were still observed between ES cells cultured on sNF or normal 2D plates (Fig. 4B). Because these morphological changes are known to be regulated by cytoskeletal molecules, such as small Rho GTPase family member proteins, we next examined Rac1 activities in undifferentiated and differentiated ES cells. Our western blot analyses demonstrate that the GTP-bound active form of Rac1 was expressed at a higher level in ES cells cultured on sNF than those cultured on normal 2D plates, even though total Rac1 expression levels were similar (Fig. 4C,D). Interestingly, both the undifferentiated (Fig. 4C) and the differentiated ES cells on d8 (Fig. 4D) showed higher Rac-GTP activities. These results not only agree with the morphological differences of the ES cells, but also suggest that activated Rac1 plays a crucial role in potentiating the differentiation activity of ES cells cultured on sNF into hepatic lineages.

A crucial role of Rac activation in potentiating the differentiation of ES cells into the definitive endoderm and hepatocyte lineages

NSC23766 is a selective inhibitor of Rac1 activation that is mediated by the Rac-specific guanine nucleotide exchange factors (GEFs) TrioN and Tiam1, without affecting other Rho family members, such as RhoA or Cdc42 (Gao et al., 2004). We confirmed that 100 µM NSC23766 inhibited Rac1 hydroxylation (supplementary material Fig. 3A). To test whether sNF potentiates the differentiation of ES cells into the hepatic lineages through Rac1 activation, we treated murine ES cells with 100 µM NSC23766 at various stages and then determined the expression of stage-specific markers (Fig. 5A–C). We first added NSC23766 for 4 days at stage I to examine the effect of Rac1 activation on endoderm induction (Fig. 5A). We found that Foxa2 expression was downregulated by the Rac1 inhibitor in ES cells cultured on sNF (Fig. 5A). We next examined the effects of Rac1 inhibition on hepatic differentiation. Our results show that the Rac1 inhibitor added at stage II (Fig. 5B) or stage III (Fig. 5C) downregulated the expression of hepatic markers, Afp or Alb1, on d10 or d14, respectively.

Fig. 5.

Inhibition of the Rac1 pathway blocks the differentiation-potentiating activity of NF. (A–C) Quantitative PCR analysis of the gene expression of Foxa2 (A), Afp (B) and Alb1 (C), in differentiated cells cultured on sNF with (+) or without (−) the Rac1 inhibitor NSC23766 (100 µM), at the end of stage I (A), stage II (B) or stage III (C). (D) The expression of Alb1 on day 18 in differentiated cells, treated with (+) or without (−) the Rac1 inhibitor at indicated stages. Data shown represent mean ± s.e.m. (n = 3); *P<0.05 and **P<0.01, compared with untreated cells on sNF with the Rac1 inhibitor by two-tailed Student's t-test or one-way ANOVA with the post-hoc Dunnett's test.

Fig. 5.

Inhibition of the Rac1 pathway blocks the differentiation-potentiating activity of NF. (A–C) Quantitative PCR analysis of the gene expression of Foxa2 (A), Afp (B) and Alb1 (C), in differentiated cells cultured on sNF with (+) or without (−) the Rac1 inhibitor NSC23766 (100 µM), at the end of stage I (A), stage II (B) or stage III (C). (D) The expression of Alb1 on day 18 in differentiated cells, treated with (+) or without (−) the Rac1 inhibitor at indicated stages. Data shown represent mean ± s.e.m. (n = 3); *P<0.05 and **P<0.01, compared with untreated cells on sNF with the Rac1 inhibitor by two-tailed Student's t-test or one-way ANOVA with the post-hoc Dunnett's test.

These results suggested that Rac1 activation is crucial for endoderm and hepatic differentiation. We subsequently examined the stage dependency of hepatic differentiation on Rac1 activities. The Rac1 inhibitor was added at different stages (I, II, III or IV) and Alb1 expression was assayed on day 18 (Fig. 5D). We found that Rac1 inhibition at all four stages blocked the potentiating effects of sNF, and resulted in decreases in Alb1 expression. These results further confirm the important role of Rac1 and demonstrate that continuous activation of Rac1 is crucial for the potentiation of hepatic differentiation.

Then we confirmed whether NSC23766 had any effect on the proliferation of ES cells. NSC23766 decreased the proportion of EdU-positive cells in stages I, II and III, particularly in stage I, without apparent toxicity (supplementary material Fig. S3C). Interestingly, the total numbers of cells in the NSC23766-treated groups was smaller in stages I and II, which became greater than that of control groups used in stages III and IV (supplementary material Fig. S3B). Taken together, these findings suggest that Rac1 differentially contributes to proliferation in the early differentiation stages and promotes differentiation in the late stage.

Our previous study suggested that although addition of soluble growth factors is sufficient to promote the differentiation of ES cells into the definitive endoderm, further differentiation from the definitive endoderm into hepatic and pancreatic fates appears to require a direct contact with M15 cells (Shiraki et al., 2008a). We previously showed the importance of basement membrane substratum by culturing ES cells on sBM (Higuchi et al., 2010; Shiraki et al., 2011). Specifically, ES cells grown on sBM were able to differentiate into hepatic and pancreatic lineages. These results imply that the basement membrane structure plays a major role in the differentiation of ES cells. Although the sBM used previously was constructed by overexpressing recombinant laminin-511 (laminin α5, laminin β1 and laminin γ1) in H293 cells (Doi et al., 2002), ES cells or iPS cells could be induced into the hepatic and pancreatic lineages. The efficacy of such an sBM for differentiation was high, and the differentiated cells could perform liver-specific functions, such as protein secretion, detoxification and glycogen storage (Higuchi et al., 2010; Shiraki et al., 2011).

The nanofiber produced by the electrospinning technique is a chemically and physically stable synthetic three-dimensional surface that mimics the structural geometry and porosity of the basement membrane or ECM (Schindler et al., 2005; Schindler et al., 2006). NF scaffolds have been shown to recapitulate the structural features of stem cell niche (Lim and Mao, 2009) and have been used for the ex vivo expansion of various types of stem cells such as murine ES cells (Hashemi et al., 2011; Nur-E-Kamal et al., 2006) and human tissue stem cells. In addition, the ECM was found to deposit as an extensive scaffold on the basal surface of the cells attached to NFs (Shih et al., 2006; Chua et al., 2007; Ma et al., 2008). Importantly, the sNF system has been reported to enhance not only the differentiation of murine ES cells into neural lineages (Lim et al., 2010; Purcell et al., 2012; Xie et al., 2009), but also differentiation from human MSCs into hepatoblasts (Ghaedi et al., 2012; Kazemnejad et al., 2009).

Taken together, these previous observations revealed that the sNF matrix is useful as a substratum to replace feeder cells and that it has the ability to potentiate hepatic differentiation. In this study, we show that both murine and human ES cells, as well as human iPS cells, could differentiate on sNF and exhibit liver-specific functions. Furthermore, we demonstrate that Rac1 activation was involved in hepatic differentiation. Rac1, a member of the Rho family GTP-binding proteins, including Rho and Cdc42 (Heasman and Ridley, 2008), functions by activating actin-rich lamellipodial protrusion and membrane ruffling, which are thought to be a major part of the driving force for cell movement (Nobes and Hall, 1995; Ridley et al., 1992). Although Rho family proteins were reported to be expressed by ES cells cultured on sNF (Nur-E-Kamal et al., 2005; Nur-E-Kamal et al., 2006; Schindler et al., 2006), their roles have never been investigated.

In vivo developmental processes occurring in the endoderm and its derivatives cause dynamic migration during gastrulation and later stages of organogenesis (Woo et al., 2012), suggesting that motility and differentiation are closely inter-related. In this study, we observed that ES cells cultured on sNF showed greater Rac1 activation than did cells cultured on the normal 2D surface. Indeed, Rac1 is known to be involved in not only endoderm induction but also hepatic specification and maturation. In particular, Rac1 mutant mice died by mouse embryonic day 9.5 (E9.5) because of severe developmental abnormalities, and Rac1-deficient embryos showed numerous cell deaths in the space between the ectoderm and endoderm at the primitive streak stage (Sugihara et al., 1998). Rac1 is also important for cellular differentiation, for example, epithelial differentiation in the small intestine (Stappenbeck and Gordon, 2000), pancreatic islet morphogenesis (Greiner et al., 2009), myogenic differentiation (Heller et al., 2001), maintenance of the thymic epithelial cells (Hunziker et al., 2011), formation of the lens (Maddala et al., 2011) and neuronal development (Corbetta et al., 2009; Leone et al., 2010). In addition, Rac1 has been shown to crosstalk with many downstream signaling pathways such as Wnt (Clarke, 2006; Malliri et al., 2006), TGF-β1 (Varon et al., 2008), Nodal (Woo et al., 2012), retinoic acid (Lee et al., 2008) and Myc (Hunziker et al., 2011; Nikolova et al., 2008). Interestingly, Rac1 is also known to mediate stem cell-shape-dependent regulation of differentiation to a chondrogenic versus myogenic fate (Gao et al., 2010). On the basis of these studies, we postulate that the sNF system might potentiate ES cells to differentiate into hepatic lineages by interacting downstream of certain growth factors during differentiation processes.

In conclusion, we show that Rac1 was activated in both undifferentiated and differentiated ES cells cultured on sNF plates and that Rac1 inhibition blocked the potentiating effects of sNF on endoderm and hepatic differentiation. These results suggest that continuous activation of Rac1 throughout the differentiation stage is crucial for potentiating differentiation. Our results also highlight the morphological changes during differentiation along the Rac1 pathway, which controls cellular morphology, motility and differentiation into the hepatic lineage. Here, we established a completely chemically defined method that requires no serum or no xenogenic substrata, thereby eliminating the risk of contamination with unknown factors. We believe that this novel method could be an attractive culture model for pharmacological research and research on stem cell biology and therapeutic strategies.

ES and iPS cell lines

The murine ES cell line, SK7 (Shiraki et al., 2008a) was maintained on mouse embryonic fibroblast (MEF) feeders in Glasgow minimum essential medium (Invitrogen, Glasgow, UK) supplemented with 1000 units/ml leukemia inhibitory factor (LIF; Chemicon, Temecula, CA), 15% knocked-out serum replacement (KSR; Invitrogen), 1% fetal bovine serum (FBS; Hyclone, Logan, UT), 100 µM nonessential amino acids (NEAA; Invitrogen), 2 mM L-glutamine (L-Gln; Invitrogen), 1 mM sodium pyruvate (Invitrogen), 50 units/ml penicillin and 50 µg/ml streptomycin (PS; Invitrogen) and 100 µM β-mercaptoethanol (β-ME; Sigma-Aldrich, St Louis, MO).

Human ES cells (KhES-3) (Suemori et al., 2006) were from Dr Norio Nakatsuji and Dr Hirofumi Suemori (Kyoto University, Kyoto, Japan). They were used in accordance with the human ES cell guidelines of the Japanese government. This human ES work was approved by Kumamoto University institutional review board. Human iPS 201B7 cells were a gift from Dr Yamanaka (Kyoto University, Kyoto, Japan). The human iPS Toe cell line was established by M. Toyoda and colleagues (National Institute for Child Health and Development, Tokyo, Japan). Undifferentiated human ES and iPS cells were maintained as described previously (Shiraki et al., 2008b).

Culture plates

Synthetic nanofiber (sNF) matrices were purchased from Corning Coster (Ultra-Web Synthetic Polyamine Surface #3873XX1; Cambridge, MA). Plate surfaces were coated with electrospun polyamide nanofibers. sNF matrices consisted of two kinds of polyamide polymers, A (C28O4N4H47)n and B (C28O4.4N4H47)n, which were crosslinked in the presence of an acid catalyst, and were 200–400 nm in diameter (average 280 nm). Pore sizes, similar to those of the cell basement membrane, were ∼700 nm. For comparison, Corning 96-well plates were pretreated for 3 hours at 37°C with 0.1% gelatin (Sigma-Aldrich), Matrigel (BD, Franklin Lakes, NJ) or CellStart (Invitrogen). Collagen I (Nitta Gelatin, Japan) was diluted with Dulbecco's modified Eagle's medium (DMEM; Invitrogen) at a concentration of 1 mg/ml and plate surfaces were treated for 15 minutes, then dried until use.

Differentiation of murine ES cells into hepatic lineages on sNF

Murine ES cells plated at a density of 1.5×104 cells/cm2 in culture plates described above were grown for 8 days in DMEM containing 4,500 mg/l glucose, s NEAA, L-Gln, PS, β-ME, 10 mg/ml insulin, 5.5 mg/ml transferrin, 6.7 pg/ml selenium (Insulin-Transferrin-Selenium-G Supplement; ITS, Invitrogen), 0.25% AlbuMAX II (Invitrogen), 10 ng/ml recombinant human activin-A (R&D Systems, Minneapolis, MN), 5 ng/ml; recombinant human bFGF, and cultured for 8 days. On day 9 (d9), the medium was changed to RPMI-1640 (Invitrogen) containing 10−6 M retinoic acid (RA; Stemolecule all-trans retinoic acid; Stemgent, Cambridge, MA) and B27 supplement (Invitrogen). On d10, medium was switched to 2000 mg/l glucose DMEM (Invitrogen), 10% KSR, 10 ng/ml recombinant human hepatocyte growth factor (Peprotech, Rocky Hill, NJ) and 10 µM dexamethasone (Sigma-Aldrich), and cultured until d14. Next, 1 mM nicotinamide (NA; Sigma-Aldrich) and 1% dimethylsufoxide (DMSO; Sigma-Aldrich), were added to medium and KSR was removed. Medium was replaced every 2 days with fresh medium and growth factors.

Human ES/iPS cells were pretreated with the ROCK inhibitor Y27632 (Wako, Japan) 1 day before trypsinization. Cells were plated at a density of 3×105 cells/cm2 on Matrigel-coated sNF matrices with Y27632. The following two procedures were subsequently used to induce hepatic differentiation of various human ES/iPS cells; simplified (two-step) protocol, KhES3 and 201B7 cells; or conventional (three-step) protocol, Toe cells. In the simplified protocol, medium used at first contained B27 and 100 ng/ml activin-A in RPMI-1640, which was then, switched to 10 ng/ml HGF, 10 µM dexamethasone, 0.5% DMSO, 0.5 mM NA. In the conventional protocol, medium used first was the same as that in the simplified protocol, followed by 1% DMSO and 20% KSR in knockout DMEM/F12 (Invitrogen) for 6 days and, then DMEM containing HGF, dexamethasone and 10% KSR. Finally, the above medium was added with 0.5 mM NA. Medium was replaced every 2 days with fresh medium and growth factors. KhES3 and 201B7 cells were induced hepatic differentiation with simplified protocol, and Toe cells were treated with conventional protocol.

Periodic-acid–Schiff's staining

For detection of glycogen storage in the differentiated cells, periodic-acid–Schiff's (PAS) staining kit (Muto Pure Chemicals, Tokyo, Japan) was used. Cells cultured for 9 and 26 days, and undifferentiated ES cells were fixed in 3.3% formalin for 10 minutes, and stained following the manufacturer's instructions, then nuclear counterstaining with hematoxylin (blue) was performed.

Albumin secretion assay

The culture medium was replaced with fresh medium every 2 days, and supernatants were collected 24 hours after replacing the medium. The mouse (human) albumin secreted in the supernatant was determined using a mouse (human) ELISA quantification kit (Bethyl, Montgomery, TX).

Indocyanine Green (ICG) test

Indocyanine Green (Daiichi-Sankyo Pharm., Japan) was diluted with the above culture medium to a final concentration of 1 mg/dl. The ICG test solution was added to the differentiated ES cells after the appropriate culture periods and undifferentiated ES cells were used as controls, and incubated at 37°C for 30 minutes. Then, after three washes with phosphate-buffered saline (PBS), the cellular uptake of ICG was examined by microscopy. The percentage ICG-positive areas represent the proportion of ICG-positive area versus total cell area, which were determined using ImageJ software (US National Institutes of Health, Bethesda, MD).

CYP inductions

To check the inducibilities of cytochrome P450 activities in response to inducers, we used the P450-Glo CYP Assay Kit (Promega, Madison, WI). The differentiated ES cells were treated with 5 µM 3-methylcholantrene as inducers of CYP1A. The medium containing the inducers was changed every 24 hours. 48 hours after treatment, we changed the medium and used the appropriate luminogenic CYP substrates (Luciferine-CEE for CYP1A). The cells were incubated at 37°C for 3 hours, and then the supernatants were mixed with equal amount of detection reagent, according to the manufacturer's instructions. The luminescence was measured using a GloMax 96 microplate luminometer (Promega), and luminometer settings were as in the manufacturer's instructions. Cell numbers were calculated using CellTiter-Glo luminescent cell viability assays (Promega) to normalize P450-Glo assay values to cell number.

Immunocytochemistry

After culture for the appropriate times, cells were fixed in 4% paraformaldehyde in PBS for 30 minutes at room temperature. After removal of paraformaldehyde solution, the fixed cells were permeabilized with 0.1% Triton X-100 for 10 minutes. The permeabilized cells were rinsed several times with PBS and were then incubated with 20% Blocking One (Nacalai Tesque, Japan) in PBST (0.1% Tween-20 in PBS) for blocking. After blocking, the cells were incubated with the diluted antibody in 20% Blocking One in PBST (0.1% Tween-20 in PBS) in a humidified chamber overnight at 4°C. After washing the cells in PBST, cells were incubated with the secondary antibody in 20% Blocking One for 2 hours at room temperature in the dark. After washing off the secondary antibody in PBST, cells were counterstained with 6-diamidino-2-phenylindole (DAPI) (Roche Diagnostics, Switzerland). The following antibodies were used as primary antibodies: rabbit anti-alphafeto protein (Dako, Denmark), goat anti-albumin (Sigma-Aldrich), goat anti-Sox17, mouse anti-FoxA2 (R&D systems); secondary antibodies used were conjugated to Alexa Fluor 568, Alexa Fluor 488 and Alexa Fluor 633 (Invitrogen). For human ES cell cultures, goat antibodies against human albumin (Bethyl) were used as primary antibodies.

Cell proliferation assay

Cell proliferation was evaluated using Click-iT EdU assay kit (invitrogen). The cells cultured with or without NSC23766 were exposed to 10 µM of 5-ethnyl-2′-deoxyuridine (EdU) for 1 hour at 37°C before fixation. The fixed cells were processed for immunocytochemistry as described above, with an additional step for EdU detection. Before incubation with secondary antibodies, the cells were incubated with EdU in the Click-iT reaction cocktail and Alexa Fluor 488 for 30 minutes at room temperature, following the manufacturer's instructions. Images were collected using ImageXpress Micro (Molecular Devices) and EdU-positive nuclei per total number of nuclei were counted.

RT-PCR analysis

RNA was extracted from ES cells or mouse liver using an RNeasy mini-kit (Qiagen, Germany) and then treated with DNase (Qiagen). For reverse transcription reactions, 3 µg RNA was reverse-transcribed using ReverTra Ace (Toyobo, Japan) and oligo dT primers (Toyobo). One µl of fivefold-diluted cDNA (1% of the RT product) was used for PCR analyses. The primer sequences for each primer set are shown in supplementary material Table S1. For real-time PCR analysis, mRNA expression was quantified with SyberGreen on an ABI 7500 thermal cycler (Applied Biosystems, Foster City, CA). The PCR conditions were as follows: denaturation at 95°C for 15 seconds, annealing and extention at 60°C for 60 seconds, for up to 40 cycles. Each measurement was normalized to β-actin (mouse) and GAPDH (human) for each sample by subtracting the average β-actin (mouse) and GAPDH (human) Ct values (Threshold Cycle) from the average Ct for each gene. Target mRNA levels, expressed as arbitrary units, were determined using a standard curve method.

Rac pull-down assay

Murine ES cells were trypsinized and suspended at a density of 5×104 cells/ml. Cells were then plated onto sNF either with or without 0.1% gelatin pretreatment; control plates were pretreated with 0.1% gelatin. Undifferentiated cells were harvested 48 hours after incubation under ES cell maintenance culture conditions at 37°C, whereas differentiated cells were harvested 9 days after hepatic differentiation started. The activation of Rac was determined using a Rac1 Activation Assay Kit purchased from Millipore. Briefly, cells were washed with PBS and suspended in lysis buffer provided by the supplier. Aliquots were taken from each cell lysate, and the amount of GAPDH proteins present in the lysates was determined and used for normalization. GTP-bound forms of Rac were then pulled down from lysates using reagents provided by the supplier, following the recommended instructions. Proteins present in total cell lysates or Rac pull-down samples were separated by SDS-PAGE (12%) and transferred onto a nylon membrane. Western blotting was performed using antibodies against Rac1, according to the ECL protocol provided by the suppliers. Luminescence of Rac1 bands was quantified using the GE ImageQuant LAS 4000 (GE Healthcare Life Science, Sweden).

We thank members of Gene Technology Center in Kumamoto University for their technical assistance.

Author contributions

T.Y. performed cellular and biochemical analyses; T.Y. and N.S. established the ES cell differentiation system; M.T., N.K., H.O., Y.M., H.A. and A.U. established human iPS Toe cell line; Y.S., K.K. and S.K. provided technical advice, designed the experiments and wrote the paper. All authors discussed the results and commented on the manuscript.

Funding

This work was supported by a grant (to S.K.) from the Realization of Regenerative Medicine from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) Japan; a grant from National Institute of Biomedical Innovation (to N.S.); a funding program for Next Generation World-Leading Researchers (NEXT Program) from the Japan Society for the Promotion of Science (JSPS) [grant number LS099 to S.K.] (to S.K.); and the Program for Leading Graduate Schools “HIGO” (to S.K.) a global COE grant (Cell Fate Regulation Research and Education Unit) from MEXT. S.K. was a member of Program (Cell Fate Regulation Research and Education Unit), MEXT, Japan.

Basma
H.
,
Soto-Gutiérrez
A.
,
Yannam
G. R.
,
Liu
L.
,
Ito
R.
,
Yamamoto
T.
,
Ellis
E.
,
Carson
S. D.
,
Sato
S.
,
Chen
Y.
 et al. (
2009
).
Differentiation and transplantation of human embryonic stem cell-derived hepatocytes.
Gastroenterology
136
,
990
999
.
Chua
K-N.
,
Chai
C.
,
Lee
P-C.
,
Ramakrishna
S.
,
Leong
K. W.
,
Mao
H-Q.
(
2007
).
Functional nanofiber scaffolds with different spacers modulate adhesion and expansion of cryopreserved umbilical cord blood hematopoietic stem/progenitor cells.
Exp. Hematol.
35
,
771
781
.
Clarke
R.
(
2006
).
Wnt signalling in the mouse intestine.
Oncogene
25
,
7512
7521
.
Corbetta
S.
,
Gualdoni
S.
,
Ciceri
G.
,
Monari
M.
,
Zuccaro
E.
,
Tybulewicz
V. L. J.
,
de Curtis
I.
(
2009
).
Essential role of Rac1 and Rac3 GTPases in neuronal development.
FASEB J.
23
,
1347
1357
.
D'Amour
K. A.
,
Agulnick
A. D.
,
Eliazer
S.
,
Kelly
O. G.
,
Kroon
E.
,
Baetge
E. E.
(
2005
).
Efficient differentiation of human embryonic stem cells to definitive endoderm.
Nat. Biotechnol.
23
,
1534
1541
.
Doi
M.
,
Thyboll
J.
,
Kortesmaa
J.
,
Jansson
K.
,
Iivanainen
A.
,
Parvardeh
M.
,
Timpl
R.
,
Hedin
U.
,
Swedenborg
J.
,
Tryggvason
K.
(
2002
).
Recombinant human laminin-10 (alpha5beta1gamma1). Production, purification, and migration-promoting activity on vascular endothelial cells.
J. Biol. Chem.
277
,
12741
12748
.
Gao
Y.
,
Dickerson
J. B.
,
Guo
F.
,
Zheng
J.
,
Zheng
Y.
(
2004
).
Rational design and characterization of a Rac GTPase-specific small molecule inhibitor.
Proc. Natl. Acad. Sci. USA
101
,
7618
7623
.
Gao
L.
,
McBeath
R.
,
Chen
C. S.
(
2010
).
Stem cell shape regulates a chondrogenic versus myogenic fate through Rac1 and N-cadherin.
Stem Cells
28
,
564
572
.
Ghaedi
M.
,
Soleimani
M.
,
Shabani
I.
,
Duan
Y.
,
Lotfi
A. S.
(
2012
).
Hepatic differentiation from human mesenchymal stem cells on a novel nanofiber scaffold.
Cell. Mol. Biol. Lett.
17
,
89
106
.
Greiner
T. U.
,
Kesavan
G.
,
Ståhlberg
A.
,
Semb
H.
(
2009
).
Rac1 regulates pancreatic islet morphogenesis.
BMC Dev. Biol.
9
,
2
.
Hashemi
S. M.
,
Soudi
S.
,
Shabani
I.
,
Naderi
M.
,
Soleimani
M.
(
2011
).
The promotion of stemness and pluripotency following feeder-free culture of embryonic stem cells on collagen-grafted 3-dimensional nanofibrous scaffold.
Biomaterials
32
,
7363
7374
.
Heasman
S. J.
,
Ridley
A. J.
(
2008
).
Mammalian Rho GTPases: new insights into their functions from in vivo studies.
Nat. Rev. Mol. Cell Biol.
9
,
690
701
.
Heller
H.
,
Gredinger
E.
,
Bengal
E.
(
2001
).
Rac1 inhibits myogenic differentiation by preventing the complete withdrawal of myoblasts from the cell cycle.
J. Biol. Chem.
276
,
37307
37316
.
Higuchi
Y.
,
Shiraki
N.
,
Yamane
K.
,
Qin
Z.
,
Mochitate
K.
,
Araki
K.
,
Senokuchi
T.
,
Yamagata
K.
,
Hara
M.
,
Kume
K.
 et al. (
2010
).
Synthesized basement membranes direct the differentiation of mouse embryonic stem cells into pancreatic lineages.
J. Cell Sci.
123
,
2733
2742
.
Hunziker
L.
,
Benitah
S. A.
,
Braun
K. M.
,
Jensen
K.
,
McNulty
K.
,
Butler
C.
,
Potton
E.
,
Nye
E.
,
Boyd
R.
,
Laurent
G.
 et al. (
2011
).
Rac1 deletion causes thymic atrophy.
PLoS ONE
6
,
e19292
.
Jung
J.
(
1999
).
Initiation of mammalian liver development from endoderm by fibroblast growth factors.
Science
284
,
1998
2003
.
Kamiya
A.
,
Kinoshita
T.
,
Ito
Y.
,
Matsui
T.
,
Morikawa
Y.
,
Senba
E.
,
Nakashima
K.
,
Taga
T.
,
Yoshida
K.
,
Kishimoto
T.
 et al. (
1999
).
Fetal liver development requires a paracrine action of oncostatin M through the gp130 signal transducer.
EMBO J.
18
,
2127
2136
.
Katsumoto
K.
,
Shiraki
N.
,
Miki
R.
,
Kume
S.
(
2010
).
Embryonic and adult stem cell systems in mammals: ontology and regulation.
Dev. Growth Differ.
52
,
115
129
.
Kazemnejad
S.
,
Allameh
A.
,
Soleimani
M.
,
Gharehbaghian
A.
,
Mohammadi
Y.
,
Amirizadeh
N.
,
Jazayery
M.
(
2009
).
Biochemical and molecular characterization of hepatocyte-like cells derived from human bone marrow mesenchymal stem cells on a novel three-dimensional biocompatible nanofibrous scaffold.
J. Gastroenterol. Hepatol.
24
,
278
287
.
Kubo
A.
,
Shinozaki
K.
,
Shannon
J. M.
,
Kouskoff
V.
,
Kennedy
M.
,
Woo
S.
,
Fehling
H. J.
,
Keller
G.
(
2004
).
Development of definitive endoderm from embryonic stem cells in culture.
Development
131
,
1651
1662
.
Lee
Y. M.
,
Lee
J. O.
,
Jung
J-H.
,
Kim
J. H.
,
Park
S-H.
,
Park
J. M.
,
Kim
E-K.
,
Suh
P-G.
,
Kim
H. S.
(
2008
).
Retinoic acid leads to cytoskeletal rearrangement through AMPK-Rac1 and stimulates glucose uptake through AMPK-p38 MAPK in skeletal muscle cells.
J. Biol. Chem.
283
,
33969
33974
.
Leone
D. P.
,
Srinivasan
K.
,
Brakebusch
C.
,
McConnell
S. K.
(
2010
).
The rho GTPase Rac1 is required for proliferation and survival of progenitors in the developing forebrain.
Dev. Neurobiol.
70
,
659
678
.
Lim
S. H.
,
Mao
H-Q.
(
2009
).
Electrospun scaffolds for stem cell engineering.
Adv. Drug Deliv. Rev.
61
,
1084
1096
.
Lim
S. H.
,
Liu
X. Y.
,
Song
H.
,
Yarema
K. J.
,
Mao
H-Q.
(
2010
).
The effect of nanofiber-guided cell alignment on the preferential differentiation of neural stem cells.
Biomaterials
31
,
9031
9039
.
Ma
K.
,
Chan
C. K.
,
Liao
S.
,
Hwang
W. Y. K.
,
Feng
Q.
,
Ramakrishna
S.
(
2008
).
Electrospun nanofiber scaffolds for rapid and rich capture of bone marrow-derived hematopoietic stem cells.
Biomaterials
29
,
2096
2103
.
Maddala
R.
,
Chauhan
B. K.
,
Walker
C.
,
Zheng
Y.
,
Robinson
M. L.
,
Lang
R. A.
,
Rao
P. V.
(
2011
).
Rac1 GTPase-deficient mouse lens exhibits defects in shape, suture formation, fiber cell migration and survival.
Dev. Biol.
360
,
30
43
.
Malliri
A.
,
Rygiel
T. P.
,
van der Kammen
R. A.
,
Song
J. Y.
,
Engers
R.
,
Hurlstone
A. F.
,
Clevers
H.
,
Collard
J. G.
(
2006
).
The rac activator Tiam1 is a Wnt-responsive gene that modifies intestinal tumor development.
J. Biol. Chem.
281
,
543
548
.
Mfopou
J. K.
,
Chen
B.
,
Mateizel
I.
,
Sermon
K.
,
Bouwens
L.
(
2010
).
Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells.
Gastroenterology
138
,
2233
2245, 2245.e1-14
.
Nikolova
E.
,
Mitev
V.
,
Minner
F.
,
Deroanne
C. F.
,
Poumay
Y.
(
2008
).
The inhibition of the expression of the small Rho GTPase Rac1 induces differentiation with no effect on cell proliferation in growing human adult keratinocytes.
J. Cell. Biochem.
103
,
857
864
.
Nobes
C. D.
,
Hall
A.
(
1995
).
Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia.
Cell
81
,
53
62
.
Nur-E-Kamal
A.
,
Ahmed
I.
,
Kamal
J.
,
Schindler
M.
,
Meiners
S.
(
2005
).
Three dimensional nanofibrillar surfaces induce activation of Rac.
Biochem. Biophys. Res. Commun.
331
,
428
434
.
Nur-E-Kamal
A.
,
Ahmed
I.
,
Kamal
J.
,
Schindler
M.
,
Meiners
S.
(
2006
).
Three-dimensional nanofibrillar surfaces promote self-renewal in mouse embryonic stem cells.
Stem Cells
24
,
426
433
.
Purcell
E. K.
,
Naim
Y.
,
Yang
A.
,
Leach
M. K.
,
Velkey
J. M.
,
Duncan
R. K.
,
Corey
J. M.
(
2012
).
Combining topographical and genetic cues to promote neuronal fate specification in stem cells.
Biomacromolecules
13
,
3427
3438
.
Ridley
J.
,
Paterson
H. F.
,
Johnston
C. L.
,
Diekmann
D.
,
Hall
A.
(
1992
).
The small GTP-binding protein rac regulates growth factor-induced membrane ruffling.
Cell
70
,
401
410
.
Rossi
J. M.
,
Dunn
N. R.
,
Hogan
B. L.
,
Zaret
K. S.
(
2001
).
Distinct mesodermal signals, including BMPs from the septum transversum mesenchyme, are required in combination for hepatogenesis from the endoderm.
Genes Dev.
15
,
1998
2009
.
Schindler
M.
,
Nur-E-Kamal
A.
,
Ahmed
I.
(
2006
).
Living in three dimensions.
Cell Biochem.
45
,
215
227
.
Schindler
M.
,
Ahmed
I.
,
Kamal
J.
,
Nur-E-Kamal
A.
,
Grafe
T. H.
,
Young Chung
H.
,
Meiners
S.
(
2005
).
A synthetic nanofibrillar matrix promotes in vivo-like organization and morphogenesis for cells in culture.
Biomaterials
26
,
5624
5631
.
Schmidt
C.
,
Bladt
F.
,
Goedecke
S.
,
Brinkmann
V.
,
Zschiesche
W.
,
Sharpe
M.
,
Gherardi
E.
,
Birchmeier
C.
(
1995
).
Scatter factor/hepatocyte growth factor is essential for liver development.
Nature
373
,
699
702
.
Shih
Y-R. V.
,
Chen
C-N.
,
Tsai
S-W.
,
Wang
Y. J.
,
Lee
O. K.
(
2006
).
Growth of mesenchymal stem cells on electrospun type I collagen nanofibers.
Stem Cells
24
,
2391
2397
.
Shin
D.
,
Shin
C. H.
,
Tucker
J.
,
Ober
E. A.
,
Rentzsch
F.
,
Poss
K. D.
,
Hammerschmidt
M.
,
Mullins
M. C.
,
Stainier
D. Y.
(
2007
).
Bmp and Fgf signaling are essential for liver specification in zebrafish.
Development
134
,
2041
2050
.
Shiraki
N.
,
Yoshida
T.
,
Araki
K.
,
Umezawa
A.
,
Higuchi
Y.
,
Goto
H.
,
Kume
K.
,
Kume
S.
(
2008a
).
Guided differentiation of embryonic stem cells into Pdx1-expressing regional-specific definitive endoderm.
Stem Cells
26
,
874
885
.
Shiraki
N.
,
Umeda
K.
,
Sakashita
N.
,
Takeya
M.
,
Kume
K.
,
Kume
S.
(
2008b
).
Differentiation of mouse and human embryonic stem cells into hepatic lineages.
Genes Cells
13
,
731
746
.
Shiraki
N.
,
Yamazoe
T.
,
Qin
Z.
,
Ohgomori
K.
,
Mochitate
K.
,
Kume
K.
,
Kume
S.
(
2011
).
Efficient differentiation of embryonic stem cells into hepatic cells in vitro using a feeder-free basement membrane substratum.
PLoS ONE
6
,
e24228
.
Si-Tayeb
K.
,
Noto
F. K.
,
Nagaoka
M.
,
Li
J.
,
Battle
M. A.
,
Duris
C.
,
North
P. E.
,
Dalton
S.
,
Duncan
S. A.
(
2010
).
Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells.
Hepatology
51
,
297
305
.
Sonnenberg
E.
,
Meyer
D.
,
Weidner
K. M.
,
Birchmeier
C.
(
1993
).
Scatter factor/hepatocyte growth factor and its receptor, the c-met tyrosine kinase, can mediate a signal exchange between mesenchyme and epithelia during mouse development.
J. Cell Biol.
123
,
223
235
.
Stappenbeck
T. S.
,
Gordon
J. I.
(
2000
).
Rac1 mutations produce aberrant epithelial differentiation in the developing and adult mouse small intestine.
Development
127
,
2629
2642
.
Suemori
H.
,
Yasuchika
K.
,
Hasegawa
K.
,
Fujioka
T.
,
Tsuneyoshi
N.
,
Nakatsuji
N.
(
2006
).
Efficient establishment of human embryonic stem cell lines and long-term maintenance with stable karyotype by enzymatic bulk passage.
Biochem. Biophys. Res. Commun.
345
,
926
932
.
Sugihara
K.
,
Nakatsuji
N.
,
Nakamura
K.
,
Nakao
K.
,
Hashimoto
R.
,
Otani
H.
,
Sakagami
H.
,
Kondo
H.
,
Nozawa
S.
,
Aiba
A.
 et al. (
1998
).
Rac1 is required for the formation of three germ layers during gastrulation.
Oncogene
17
,
3427
3433
.
Tremblay
K. D.
,
Hoodless
P. A.
,
Bikoff
E. K.
,
Robertson
E. J.
(
2000
).
Formation of the definitive endoderm in mouse is a Smad2-dependent process.
Development
127
,
3079
3090
.
Umeda
K.
,
Suzuki
K.
,
Yamazoe
T.
,
Shiraki
N.
,
Higuchi
Y.
,
Tokieda
K.
,
Kume
K.
,
Mitani
K.
,
Kume
S.
(
2013
).
Albumin gene targeting in human embryonic stem cells and induced pluripotent stem cells with helper-dependent adenoviral vector to monitor hepatic differentiation.
Stem Cell Res.
10
,
179
194
.
Varon
C.
,
Rottiers
P.
,
Ezan
J.
,
Reuzeau
E.
,
Basoni
C.
,
Kramer
I.
,
Génot
E.
(
2008
).
TGFbeta1 regulates endothelial cell spreading and hypertrophy through a Rac-p38-mediated pathway.
Biol. Cell
100
,
537
550
.
Woo
S.
,
Housley
M. P.
,
Weiner
O. D.
,
Stainier
D. Y. R.
(
2012
).
Nodal signaling regulates endodermal cell motility and actin dynamics via Rac1 and Prex1.
J. Cell Biol.
198
,
941
952
.
Xie
J.
,
Willerth
S. M.
,
Li
X.
,
Macewan
M. R.
,
Rader
A.
,
Sakiyama-Elbert
S. E.
,
Xia
Y.
(
2009
).
The differentiation of embryonic stem cells seeded on electrospun nanofibers into neural lineages.
Biomaterials
30
,
354
362
.

Supplementary information