Oncostatin M in William's E medium is suitable for initiation of hepatocyte differentiation in human induced pluripotent stem cells

  • Authors:
    • Minoru Tomizawa
    • Fuminobu Shinozaki
    • Yasufumi Motoyoshi
    • Takao Sugiyama
    • Shigenori Yamamoto
    • Naoki Ishige
  • View Affiliations

  • Published online on: March 28, 2017     https://doi.org/10.3892/mmr.2017.6406
  • Pages: 3088-3092
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

William's E (WE) is a suitable medium for the differentiation of human induced pluripotent stem (iPS) cells to the hepatocyte lineage. The aim of the present study was to investigate various growth factors in their ability to promote hepatocyte differentiation of iPS cells in WE medium. Human iPS 201B7 cells were cultured in WE medium supplemented with growth factors, and mRNA expression levels and promoter activities of α‑fetoprotein (AFP) and albumin were examined by reverse transcription‑quantitative polymerase chain reaction and luciferase assay, respectively. In addition, time course analysis of AFP mRNA expression was performed in 201B7 cells cultured in WE medium supplemented with oncostatin M. The results demonstrated that mRNA expression levels of AFP were significantly elevated by most growth factors tested as supplements in WE medium, except all‑trans retinoic acid, compared with cells cultured in ReproFF (a medium that maintains pluripotency). The highest increase in AFP mRNA expression levels was observed by oncostatin M stimulation. Albumin mRNA expression levels were increased by all‑trans retinoic acid and insulin‑transferrin‑selenium supplementation in WE medium compared with cells cultured in ReproFF. Oncostatin M supplementation significantly stimulated the promoter activity of the AFP gene, but no growth factor tested significantly stimulated the promoter activity of the albumin gene. By time course analysis, significant increase of AFP mRNA expression was observed on the sixth day post‑stimulation, compared with cells cultured in WE medium alone. In conclusion, the present study demonstrated that oncostatin M supplementation in WE medium was sufficient to initiate hepatocyte differentiation in iPS cells.

Introduction

Human induced pluripotent stem (iPS) cells are established with the introduction of reprogramming factors (1). iPS cells have the potential to differentiate into hepatocyte-like cells by stimulation with growth factors or by introduction of transcription factors (2,3). It is hypothesized that hepatocytes generated from iPS cells could be used in the future to treat liver failure, a fatal condition due to major loss of hepatocytes, by transplantation into patients (4). Current protocols, however, have many limitations, including that the hepatocytes generated from iPS cells remain at an immature state (5). In addition, current protocols require several weeks to obtain hepatocyte-like cells.

Growth factors and transcription factors are important for differentiation of iPS cells to hepatocyte lineage. Transcription factor protocols efficacy depends on the efficiency of the method used for the introduction of the gene of interest to target cells. This problem has been, partly, overcome by the use of adenovirus vectors (6). Transcription factors have also been introduced into iPS cells with conventional reagents (7,8). Alternatively, a combination of growth factors and media has been demonstrated to affect the differentiation of iPS cells to hepatocyte lineage. A recent study reported that William's E medium (WE) is suitable for hepatocyte differentiation of iPS cells when followed by culturing in hepatocyte differentiation inducer medium (9). WE medium was originally formulated to culture primary hepatocytes, and it has been demonstrated to maintain hepatocyte-specific drug metabolism (10,11). However, whether growth factor supplementation in WE medium is sufficient to initiate hepatocyte differentiation of iPS cells remains unkown.

Oncostatin M, a member of interleukin 6 family, is expressed in fetal liver, and its expression decreases after birth (12). Oncostatin M promotes maturation of hepatocyte differentiation from fetal hepatocytes in vitro (13). The resulting hepatocytes express hepatocyte-specific genes, accumulate glycogen, and remove ammonia (13). Based on these properties, it is hypothesized that oncostatin M may promote hepatocyte differentiation of iPS cells, but its effect as a supplement in WE medium remains unknown.

The present study, therefore, investigated whether supplementation of WE medium with various growth factors, including oncostatin M, was a suitable method for hepatocyte differentiation of iPS cells. The results may provide a novel method for the successful differentiation of iPS cells to hepatocytes, which could be potentially useful in the future for therapeutic transplantation into patients.

Materials and methods

Cell culture

Human iPS 201B7 cells (RIKEN BioResource Center, Tsukuba, Japan) were cultured in ReproFF (Reprocell, Inc., Yokohama, Japan) medium in 6-well plates coated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA) under feeder-free conditions. The cells were then harvested using Accutase (Innovative Cell Technologies, Inc., San Diego, CA, USA) and centrifuged at 100 × g for 3 min at 4°C. The cells were spread onto new 6-well plates or 96-well plates coated with Matrigel. For experiments, cells were cultured in ReproFF or in WE (Thermo Fisher Scientific, Inc., Waltham, MA, USA) media supplemented with nicotinamide (1.2 mg/ml; Wako Pure Chemical Industries, Ltd., Osaka, Japan), proline (30 ng/ml; Wako Pure Chemical Industries, Ltd.) and 10% Knockout serum replacement (Thermo Fisher Scientific, Inc.).

Growth factors

The growth factors used in the present study were: Basic fibroblast growth factor (5 ng/ml; Wako Pure Chemical Industries, Ltd.), bone morphogenetic protein 4 (20 ng/ml; Wako Pure Chemical Industries, Ltd.), oncostatin M (20 ng/mg; Wako Pure Chemical Industries, Ltd.), epidermal growth factor (20 ng/ml; Wako Pure Chemical Industries, Ltd.), β-nerve growth factor (100 ng/ml; R&D Systems, Inc., Minneapolis, MN, USA), all-trans retinoic acid (1 µM; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), transforming growth factor-β1 (2 ng/ml; R&D Systems, Inc.), hepatocyte growth factor (20 ng/ml; Wako Pure Chemical Industries, Ltd.), dexamethasone (10−7 M; Wako Pure Chemical Industries, Ltd.), and insulin-transferrin-sodium selenite media supplement (100x; Sigma-Aldrich; Merck KGaA).

Plasmid construction

The EcoR1-Sal1 fragment of the human α-fetoprotein (AFP) promoter (Switchgear Genomics, Carlsbad, CA, USA) was subcloned into the pMetLuc2-reporter plasmid (Clontech Laboratories, Inc., Mountainview, CA, USA) to produce the pMetLuc2/AFP promoter reporter plasmid. Similarly, the Sac1-Hind3 fragment of the human albumin promoter (Switchgear Genomics) was subcloned into the pMetLuc2-reporter plasmid to produce the pMetLuc2/albumin promoter reporter plasmid.

Transfection and luciferase assay

201B7 cells were cultured in 96-well plates coated with Matrigel. Cells in each well (5×105 cells/well) were transfected with 100 ng pMetLuc2/AFP or pMetLuc2/albumin plasmid using FuGENE HD transfection reagent (Promega Corporation, Madison, WI, USA), according to the manufacturer's instructions. As a control, 10 ng pSEAP2-control plasmid (Clontech Laboratories, Inc.), which expresses secreted alkaline phosphatase (SEAP), was transfected into the cells in parallel to monitor the transfection efficiency. The transfected cells were then cultured in ReproFF (FF) or WE media, with or without growth factors. Following two days of culture, media samples were evaluated by a Ready-To-Glow secreted luciferase assay (Clontech Laboratories, Inc.) and a SEAP assay (Clontech Laboratories, Inc.). The luciferase measurement was divided by the SEAP measurement to calculate the relative gene promoter activity.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA (~5 µg) was isolated from cultured cells using Isogen (Nippon Gene Co., Ltd., Tokyo, Japan) and was utilized for the synthesis of first-strand cDNA using SuperScript III reverse transcriptase and oligo (dT) primers (Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Total RNA from human fetal liver was purchased from Clontech Laboratories, Inc. RT-qPCR was performed using Fast SYBR-Green Master Mix (Thermo Fisher Scientific, Inc.) and the results were analyzed using the MiniOpticon Real-Time PCR System (Bio-Rad Laboratories, Inc., Hercules, CA, USA). qPCR was performed in a volume of 20 µl for 40 cycles, with thermocyclining conditions according to the Fast SYBR-Green Master Mix suggested protocol. Primer sequences are listed in Table I. RPL19 was used as an endogenous reference control because it is a constitutively expressed housekeeping gene (14). The gene expression levels were analyzed automatically using the MiniOpticon System, based on the 2−ΔΔCq method (15). The relative expression was calculated as the expression level of a specific gene divided by that of RPL19.

Table I.

Primer sequences used for quantitative polymerase chain reaction.

Table I.

Primer sequences used for quantitative polymerase chain reaction.

GenePrimerSequence (5′-3′)Product size (bp)GenBank accession number
AFP
F ACACAAAAAGCCCACTCCAG147NM_001134
R GGTGCATACAGGAAGGGATG
RPL19
F CGAATGCCAGAGAAGGTCAC157BC095445
R CCATGAGAATCCGCTTGTTT
Albumin
F GCTCGTGAAACACAAGCCCAAG114NM_000477
R GCAAAGCAGGTCTCCTTATCGTC

[i] Primers were designed to have an annealing temperature of 60°C and underwent 40 cycles of quantitative polymerase chain reaction. AFP, α-fetoprotein; RPL19, ribosomal protein L19.

Statistical analysis

Data are presented as means ± standard deviation of three independent repeats. Results were analyzed for statistical significance by one-way analysis of variance followed by Turkey's post hoc test, using JMP version 10.0.2 software (SAS Institute, Inc., Cary, NC, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Human iPS 201B7 cells were cultured in WE medium supplemented with various growth factors for 7 days and then mRNA expression levels of AFP and albumin were analyzed by RT-qPCR. As a control, cells were also cultured in WE medium alone and in ReproFF, a medium that maintains pluripotency. The mRNA expression levels of AFP were significantly increased in cells cultured in WE with most of the growth factors tested, except all-trans retinoic acid, compared with cells cultured in ReproFF (Fig. 1A). Out of all the factors tested, Oncostatin M supplementation resulted in the highest increase in AFP mRNA (Fig. 1A). The mRNA expression levels of albumin was increased in cells cultured in WE medium supplemented with all-trans retinoic acid and insulin-transferrin-selenium compared with cells cultured in ReproFF (Fig. 1B).

A luciferase reporter assay was performed to investigate the effect of the growth factors on the promoter activity of the AFP and albumin genes. 201B7 cells were transfected with either the pMetLuc2/AFP (Fig. 2A) or pMetLuc2/albumin (Fig. 2B) promoter reporter plasmid, in parallel with pSEAP2-control plasmid, which expresses alkaline phosphatase, as a control for transfection efficiency. Following 2 days of culturing the transfected cells in ReproFF or WE with or without growth factors, samples from the culture media were subjected to luciferase and SEAP assays. Oncostatin M supplementation in WE media was observed to stimulate AFP promoter activity most strongly and significantly (Fig. 1A), while no growth factor tested significantly affected the promoter activity of the albumin gene (Fig. 1B).

The present results suggested that oncostatin M may be suitable for the initiation of hepatocyte differentiation in iPS cells. Time course analysis of the AFP mRNA expression pattern was performed to identify the timing of the stimulation of 201B7 cells towards the initiation of hepatocyte lineage differentiation (Fig. 3). The mRNA expression levels of AFP increased on the sixth day following the initiation of culture in the oncostatin M-supplemented WE medium, compared with cells cultured in WE medium alone (Fig. 3).

Discussion

Oncostatin M belongs to the interleukin-6 subfamily (16) and it is secreted by hematopoietic cells during endoderm formation (17). Oncostatin M is important in the maturation of immature hepatocytes (18) and in the long-term culture of human primary hepatocytes (19). In the present study, it was demonstrated that oncostatin M initiated the differentiation of iPS cells to hepatocyte lineage. The mechanism of the initiation of hepatocyte differentiation of iPS cells remains unclear. The present data is consistent with previous studies from our own group (5,20) and a study from another group that has demonstrated that oncostatin M promotes differentiation of iPS cells to hepatocytes (21). Therefore, oncostatin M is suggested to be suitable for the initiation of hepatocyte differentiation from iPS cells.

There is the potential that some of the immature iPS cells cultured in WE medium supplemented with oncostatin M remain after differentiation initiation. To counter this, hepatocyte selection medium (HSM) could be used to eliminate undifferentiated iPS cells from the culture. Cells do not survive in media without glucose or arginine, because these two ingredients are essential for their survival. However, the gluconeogenesis pathway and the urea cycle occur in hepatocytes, thereby allowing them to produce glucose from galactose and arginine from ornithine, respectively (18,19). Hence, HSM is routinely prepared without glucose or arginine, but supplemented with galactose and ornithine (22), and culturing in HSM results is elimination of undifferentiated iPS cells, however survival of hepatocytes (23). Since iPS cells cannot survive in media without glucose, it is hypothesized that the potentially remaining iPS cells in the differentiation conditions tested in the present study may be eliminated by subsequent culture in HSM.

A limitation of the present study is that the expression levels of AFP and albumin were analyzed only at the mRNA level, and not at the protein level. In addition, the expression levels of liver-specific genes, including cytochrome P450 3A4, aldehyde dehydrogenase 2, and glucose-6-phosphatase were not analyzed. Finally, Fig. 3 demonstrated that AFP expression increased on the sixth day of culture post-stimulation with oncostatin M in WE medium compared with WE medium alone, however, the time course analysis was only performed up to 6 days and therefore, it remains unclear whether longer culture would increase AFP expression further or whether this was the maximum. Further studies are required to culture iPS cells in WE supplemented with oncostatin M for longer than 6 days, and to analyze the protein expression of AFP and albumin, in addition to the expression of liver-specific gene markers.

Acknowledgements

The present study was supported by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (grant no. 15K09032).

References

1 

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K and Yamanaka S: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 131:861–872. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Takayama K, Inamura M, Kawabata K, Sugawara M, Kikuchi K, Higuchi M, Nagamoto Y, Watanabe H, Tashiro K, Sakurai F, et al: Generation of metabolically functioning hepatocytes from human pluripotent stem cells by FOXA2 and HNF1α transduction. J Hepatol. 57:628–636. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Takayama K, Inamura M, Kawabata K, Katayama K, Higuchi M, Tashiro K, Nonaka A, Sakurai F, Hayakawa T, Furue Kusuda M and Mizuguchi H: Efficient generation of functional hepatocytes from human embryonic stem cells and induced pluripotent stem cells by HNF4α transduction. Mol Ther. 20:127–137. 2012. View Article : Google Scholar

4 

Basma H, Soto-Gutiérrez A, Yannam GR, Liu L, Ito R, Yamamoto T, Ellis E, Carson SD, Sato S, Chen Y, et al: Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology. 136:990–999. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Tomizawa M, Shinozaki F, Sugiyama T, Yamamoto S, Sueishi M and Yoshida T: Single-step protocol for the differentiation of human-induced pluripotent stem cells into hepatic progenitor-like cells. Biomed Rep. 1:18–22. 2013.PubMed/NCBI

6 

Inamura M, Kawabata K, Takayama K, Tashiro K, Sakurai F, Katayama K, Toyoda M, Akutsu H, Miyagawa Y, Okita H, et al: Efficient generation of hepatoblasts from human ES cells and iPS cells by transient overexpression of homeobox gene HEX. Mol Ther. 19:400–407. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Tanaka A, Woltjen K, Miyake K, Hotta A, Ikeya M, Yamamoto T, Nishino T, Shoji E, Sehara-Fujisawa A, Manabe Y, et al: Efficient and reproducible myogenic differentiation from human iPS cells: Prospects for modeling Miyoshi Myopathy in vitro. PLoS One. 8:e615402013. View Article : Google Scholar : PubMed/NCBI

8 

Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S and Sueishi M: Dual gene expression in embryoid bodies derived from human induced pluripotent stem cells using episomal vectors. Tissue Eng Part A. 20:3154–3162. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S and Ishige N: Improved survival and initiation of differentiation of human induced pluripotent stem cells to hepatocyte-like cells upon culture in William's E medium followed by hepatocyte differentiation inducer treatment. PLoS One. 11:e01534352016. View Article : Google Scholar : PubMed/NCBI

10 

Wu D, Ramin SA and Cederbaum AI: Effect of pyridine on the expression of cytochrome P450 isozymes in primary rat hepatocyte culture. Mol Cell Biochem. 173:103–111. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Takeba Y, Matsumoto N, Takenoshita-Nakaya S, Harimoto Y, Kumai T, Kinoshita Y, Nakano H, Ohtsubo T and Kobayashi S: Comparative study of culture conditions for maintaining CYP3A4 and ATP-binding cassette transporters activity in primary cultured human hepatocytes. J Pharmacol Sci. 115:516–524. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Kamiya A, Kojima N, Kinoshita T, Sakai Y and Miyaijma A: Maturation of fetal hepatocytes in vitro by extracellular matrices and oncostatin M: induction of tryptophan oxygenase. Hepatology. 35:1351–1359. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Kamiya A, Kinoshita T, Ito Y, Matsui T, Morikawa Y, Senba E, Nakashima K, Taga T, Yoshida K, Kishimoto T and Miyajima A: Fetal liver development requires a paracrine action of oncostatin M through the gp130 signal transducer. EMBO J. 18:2127–2136. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Davies B and Fried M: The L19 ribosomal protein gene (RPL19): Gene organization, chromosomal mapping and novel promoter region. Genomics. 25:372–380. 1995. View Article : Google Scholar : PubMed/NCBI

15 

Tam S, Clavijo A, Engelhard EK and Thurmond MC: Fluorescence-based multiplex real-time RT-PCR arrays for the detection and serotype determination of foot-and-mouth disease virus. J Virol Methods. 161:183–191. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Urbańska-Ryś H, Wiersbowska A, Stepień H and Robak T: Relationship between circulating interleukin-10 (IL-10) with interleukin-6 (IL-6) type cytokines (IL-6, interleukin-11 (IL-11), oncostatin M (OSM)) and soluble interleukin-6 (IL-6) receptor (sIL-6R) in patients with multiple myeloma. Eur Cytokine Netw. 11:443–451. 2000.PubMed/NCBI

17 

Kinoshita T, Sekiguchi T, Xu MJ, Ito Y, Kamiya A, Tsuji K, Nakahata T and Miyajima A: Hepatic differentiation induced by oncostatin M attenuates fetal liver hematopoiesis. Proc Natl Acad Sci USA. 96:7265–7270. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Ye JS, Su XS, Stoltz JF, de Isla N and Zhang L: Signalling pathways involved in the process of mesenchymal stem cells differentiating into hepatocytes. Cell Prolif. 48:157–165. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Levy G, Bomze D, Heinz S, Ramachandran SD, Noerenberg A, Cohen M, Shibolet O, Sklan E, Braspenning J and Nahmias Y: Long-term culture and expansion of primary human hepatocytes. Nat Biotechnol. 33:1264–1271. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S and Ishige N: An optimal medium supplementation regimen for initiation of hepatocyte differentiation in human induced pluripotent stem cells. J Cell Biochem. 116:1479–1489. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Kondo Y, Iwao T, Nakamura K, Sasaki T, Takahashi S, Kamada N, Matsubara T, Gonzalez FJ, Akutsu H, Miyagawa Y, et al: An efficient method for differentiation of human induced pluripotent stem cells into hepatocyte-like cells retaining drug metabolizing activity. Drug Metab Pharmacokinet. 29:237–243. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Tomizawa M, Toyama Y, Ito C, Toshimori K, Iwase K, Takiguchi M, Saisho H and Yokosuka O: Hepatoblast-like cells enriched from mouse embryonic stem cells in medium without glucose, pyruvate, arginine and tyrosine. Cell Tissue Res. 333:17–27. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Tomizawa M, Shinozaki F, Sugiyama T, Yamamoto S, Sueishi M and Yoshida T: Survival of primary human hepatocytes and death of induced pluripotent stem cells in media lacking glucose and arginine. PLoS One. 8:e718972013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 15 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S and Ishige N: Oncostatin M in William's E medium is suitable for initiation of hepatocyte differentiation in human induced pluripotent stem cells. Mol Med Rep 15: 3088-3092, 2017
APA
Tomizawa, M., Shinozaki, F., Motoyoshi, Y., Sugiyama, T., Yamamoto, S., & Ishige, N. (2017). Oncostatin M in William's E medium is suitable for initiation of hepatocyte differentiation in human induced pluripotent stem cells. Molecular Medicine Reports, 15, 3088-3092. https://doi.org/10.3892/mmr.2017.6406
MLA
Tomizawa, M., Shinozaki, F., Motoyoshi, Y., Sugiyama, T., Yamamoto, S., Ishige, N."Oncostatin M in William's E medium is suitable for initiation of hepatocyte differentiation in human induced pluripotent stem cells". Molecular Medicine Reports 15.5 (2017): 3088-3092.
Chicago
Tomizawa, M., Shinozaki, F., Motoyoshi, Y., Sugiyama, T., Yamamoto, S., Ishige, N."Oncostatin M in William's E medium is suitable for initiation of hepatocyte differentiation in human induced pluripotent stem cells". Molecular Medicine Reports 15, no. 5 (2017): 3088-3092. https://doi.org/10.3892/mmr.2017.6406