Establishment of Tsc2‑deficient rat embryonic stem cells

  • Authors:
    • Yoshitaka Ito
    • Haruna Kawano
    • Fumio Kanai
    • Eri Nakamura
    • Norihiro Tada
    • Setsuo Takai
    • Shigeo Horie
    • Hajime Arai
    • Toshiyuki Kobayashi
    • Okio Hino
  • View Affiliations

  • Published online on: March 3, 2015     https://doi.org/10.3892/ijo.2015.2913
  • Pages: 1944-1952
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tuberous sclerosis complex (TSC) is an autosomal dominant disorder caused by TSC1 or TSC2 mutations. TSC causes the development of tumors in various organs such as the brain, skin, kidney, lung, and heart. The protein complex TSC1/2 has been reported to have an inhibitory function on mammalian target of rapamycin complex 1 (mTORC1). Treatment with mammalian target of rapamycin (mTOR) inhibitors has demonstrated tumor‑reducing effects in patients with TSC but is also associated with various adverse effects. In recent years, experiments involving in vivo differentiation of pluripotent stem cells have been reported as useful in elucidating mechanisms of pathogenesis and discovering new therapeutic targets for several diseases. To reveal the molecular basis of the pathogenesis caused by the Tsc2 mutation, we derived embryonic stem cells (ESCs) from Eker rats, which have the Tsc2 mutation and develop brain lesions and renal tumors. Although several studies have reported the necessity of Tsc1 and Tsc2 regulation to maintain ESCs and hematopoietic stem cells, we successfully established not only Tsc2+/+ and Tsc2+/- ESCs but also Tsc2-/- ESCs. We confirmed that these cells express pluripotency markers and retain the ability to differentiate into all three germ layers. Comprehensive gene expression analysis of Tsc2+/+ and Tsc2+/- ESCs revealed similar profiles, whereas the profile of Tsc2-/- ESCs was distinct from these two. In vitro differentiation experiments using these ESCs combined with in vivo experiments may reveal the mechanism of the tissue‑specific pathogenesis caused by the Tsc2 mutation and identify specific new therapeutic targets.

Introduction

Tuberous sclerosis complex (TSC) is a genetic disorder characterized by multisystem involvement and wide phenotypic variability. This condition results in the development of non-cancerous tumors in various organs and most frequently affects the brain, skin, kidney, lung, heart, and retina. TSC manifestations in the central nervous system include cortical tubers, subependymal nodules, subependymal giant cell astrocytomas, and scattered abnormal cells throughout the brain (1). A majority of patients with TSC reveal neurological and/or psychiatric symptoms, including epilepsy, intellectual disability, autism spectrum disorder (ASD), attention deficit, depression, and anxiety disorder, which range from mild to severe and may impair their ability to live an independent life.

Mutation of either TSC1 or TSC2 causes TSC (2,3). Protein products of TSC1 (hamartin) and TSC2 (tuberin) form a complex that inhibits the Ras homologue enriched in the brain (Rheb), a small G protein that activates mammalian target of rapamycin complex 1 (mTORC1). Defects of TSC1 or TSC2 cause excessive mTORC1 activation, which in turn provokes abnormal regulation of important cellular processes such as cellular growth and proliferation (4,5). The Knudson’s ‘two-hit’ model (6) has been the working molecular model for tumor development in TSC for several years. In fact, loss of heterozygosity (LOH) of TSC1 or TSC2 has been demonstrated in renal angiomyolipomas (79) and in subependymal giant cell astrocytomas (10). However, evidence for LOH in TSC cortical tubers is limited (11). On the other hand, haploinsufficiency of these genes is also speculated to be involved in TSC pathogenesis. To reveal Tsc mutation-related mechanisms of the pathogenesis, rodents harboring a defect of the Tsc1 or Tsc2 gene have been extensively investigated (1215). For instance, Tsc1+/− and Tsc2+/− mouse models exhibit learning and memory deficits (16,17). Eker rats are heterozygous for a mutation of Tsc2 and develop hereditary kidney cancer by the age of 1 year (1820). Although kidney cancer is rare in human patients with TSC, it is the only cancer known to occur at an increased incidence in TSC. The embryonic lethality of Tsc2−/− Eker rat embryos is characterized by disrupted neuroepithelial growth (21). Although cortical tubers are rare (22), 63% of Eker rats develop brain lesions comprising a mixture of large and elongated cells in both subependymal and subcortical regions (23,24). In contrast, among Tsc1 and Tsc2 knock-out mouse models, only conditional ablation in the brain can induce such lesions (25). Consequently, with regard to brain lesions, the Eker rat model is more similar to the human patients compared with other mouse models.

We observed that the tumorigenicity of Tsc2−/− cells derived from mice was effectively inhibited by rapamycin treatment (26). Other groups reported a similar effect when Eker rats or knock-out mice were treated with rapamycin, although some residual tumors were detected (27,28). These findings have provided the rationale for therapy with rapalogues to treat TSC lesions such as lymphangioleiomyomatosis, SEGAs, and angiomyolipomas, directed at the abnormal activation of mTORC1 (2931). Although decreased tumor volume has been documented, complete cure was not achieved in most cases. In addition, there are several problems associated with long-term use of rapalogues, including various undesirable side-effects. Consequently additional therapeutic molecular targets are required. The pathogenesis of TSC is assumed to be related to abnormal differentiation as a result of TSC1/2 deficiency. For instance, abnormal giant cells that appear in brain lesions of patients with TSC express both neuronal and glial lineage markers (32). In recent years, a number of articles have revealed differentiation- and cell type-specific abnormalities using in vitro differentiation protocols to investigate differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). To evaluate roles of Tsc2 from the viewpoint of differentiation and tissue-specific pathogenesis as well as to compare and combine in vivo and in vitro models, we established ESCs from Eker rats.

In 2008, authentic rat ESCs were established for the first time (33,34), lagging behind the establishment of mouse (35,36) and human ESCs (37). Using methods described by Buehr et al (33), we generated ESCs from blastocysts of Eker rats to establish an in vivo experimental system to explore the role of Tsc2 in TSC pathogenesis. Although several reports have indicated the necessity of Tsc1 and Tsc2 regulation to maintain ESCs (38) and somatic stem cells (39) or to establish iPS cells (40), we were able to establish not only Tsc2+/+ and Tsc2+/− ESCs but also Tsc2−/− ESCs. To our knowledge, this is the first report describing the generation of Tsc2-deficient ESCs.

Materials and methods

Ethics statement

All animal experiments were conducted in strict accordance with the institutional guidelines of Juntendo University for animal experiments. The protocol was approved by the Animal Experimentation Committee of Juntendo University (Tokyo, Japan) (approval no. 250105). All surgical procedures were performed under isoflurane anesthesia, and all efforts were made to minimize animal suffering.

Animals

Genetic homogeneity of Eker rats was maintained in our laboratory by brother-sister mating. Wistar rats, Brown Norway rats, C57BL/6J mice, and nude mice were purchased from Charles River Laboratories Japan, Inc. (Kanagawa, Japan). All animals were housed under specific pathogen-free conditions.

Mouse embryonic fibroblasts (MEFs)

MEFs were derived from embryonic day 14.5 C57BL6/J mouse embryos. MEFs were cultured in Knockout DMEM supplemented with 10% fetal bovine serum, 1% L-glutamine, and penicillin streptomycin (all from Gibco Life technologies, Carlsbad, CA, USA) on gelatin-coated dishes. MEFs were treated with mitomycin C (Sigma-Aldrich, St. Louis, MO, USA) for use as feeder cells.

Culture of ESCs

We generated ESCs from Eker rats according to the method reported by Buehr et al (33). After double heterozygous mating of Eker rats, E4.5 blastocysts were gently flushed out from uteri using the N2B27 medium (StemCells, Inc., Newark, CA, USA). After removal of zonae pellucidae with acid Tyrode’s solution, whole blastocysts were plated and cultured on mitomycin C-treated MEFs in N2B27 medium supplemented with 3 μM of CHIR99021, 1 μM of PD0325901 (both from Axon Medchem BV, Groningen, The Netherlands), 1,000 U/ml rat leukemia inhibitory factor (LIF) (ESGRO®; Millipore, Bedford, MA, USA) [two inhibitors (2i) + LIF condition]. After 5–7 days, blastocyst outgrowths were cut into pieces and replated in the same 2i + LIF medium. Thereafter, emerging ESC colonies were dissociated using Accutase (Innovative Cell Technologies, Inc., San Diego, CA, USA) and passaged every 2–4 days.

Alkaline phosphatase staining

Alkaline phosphatase staining was performed with an alkaline phosphatase kit (85L3R; Sigma-Aldrich) according to the manufacturer’s instructions.

Chromosomal analysis

A standard chromosome preparation method using colchicine treatment was employed. Chromosome preparations were analyzed after Giemsa staining. At least 30 metaphase chromosome sets were analyzed for each line.

Genotyping polymerase chain reaction (PCR)

Genotyping of ESCs was conducted using PCR on ESC DNA. To discriminate Tsc2 mutant or wild-type alleles, the following primers were used: 5MFJ (5′-ACC ATC AGG ATG CTG CTG AA-3′), 3MFJ2 (5′-CTA TGG CCA CAT GTG ACC AA-3′), and TSR27 (5′-GCG CCA GAT TCA CCT CAT TA-3′) (41). PCR was used to identify the gender of ESCs by amplification of the rat Y chromosome-specific Sry gene using the primer pair Sry-F (5′-CAT CGA AGG GTT AAA GTG CCA-3′) and Sry-R (5′-ATA GTG TGT AGG TTG TTG TCC-3′) (33).

Reverse transcription (RT)-PCR

Total RNA was obtained using a NucleoSpin® RNA II kit (Macherey-Nagel GmbH & Co. KG, Düren, Germany) according to the manufacturer’s instructions. Complementary DNA was synthesized using a SuperScript III First-Strand Synthesis SuperMix kit (Invitrogen Life Technologies, Carlsbad, CA, USA) and an oligo-dT primer, according to the manufacturer’s instructions. PCR was performed in a thermal cycler (Hybaid MBS 0.2G Thermal Cycler; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The following primer pairs were used: Oct4-F (5′-GGG ATG GCA TAC TGT GGA C-3′), Oct4-R (5′-CTT CCT CCA CCC ACT TCT C-3′), Sox2-F (5′-GGC GGC AAC CAG AAG AAC AG-3′), Sox2-R (5′-GTT GCT CCA GCC GTT CAT GTG-3′), rat Nanog-F (5′-GCC CTG AGA AGA AAG AAG AG-3′), rat Nanog-R (5′-CGT ACT GCC CCA TAC TGG AA-3′) (33), rat nestin-F (5′-AGC CAT TGT GGT CTA CTG A-3′), rat nestin-R (5′-TGC AAC TCT GCC TTA TCC-3′), Sox17-F (5′-AGG AGA GGT GGT GGC GAG TAG-3′), and Sox17-R (5′-GTT GGG ATG GTC CTG CAT GTG-3′) (34).

Western blotting

Cells were harvested and lysed in sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) sample buffer (50 mM Tris-HCl, pH 6.8, 2% SDS, and 10% glycerol). Proteins were separated by SDS-PAGE and transferred onto a polyvinylidene fluoride (PVDF) membrane (Millipore). The membrane was blocked with 1% skimmed milk in Tris-buffered saline containing 0.05% Tween-20 and probed with appropriate antibodies using the EnVision System (DakoCytomation, Glostrup, Denmark). Antibody signals were developed using ECL reagents and Hyperfilm ECL film (both from GE Healthcare, Little Chalfont, UK), which were then scanned using CEPROS SV (Fujifilm, Tokyo, Japan). The following primary antibodies were used: anti-Tsc2 antibody (C20; 1:200; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), anti-Tsc1 primary antibody (c-Tsc1, 1:500), anti-phospho-S6 ribosomal protein (Ser235/236) rabbit polyclonal antibody (1:1,000, no. 2211), anti-S6 ribosomal protein rabbit monoclonal antibody (1:1,000, no. 2217) (both from Cell Signaling Technology, Inc., Danvers, MA, USA), and anti-β-actin mouse monoclonal antibody (1:1,000; Sigma-Aldrich).

Embryoid body (EB) formation

ESCs were plated into low-adhesion 96-well dishes (MS-9096; Sumitomo Bakelite Co., Ltd., Tokyo, Japan). After 10 days of suspension culture, EBs were plated onto Matrigel-coated dishes in GMEM/10% fetal bovine serum medium (both from Gibco Life Technologies).

Immunocytochemistry

Cells were fixed and permeabilized with 4% paraformaldehyde and 0.25% Triton X-100 (both from Wako Pure Chemical Industries, Ltd., Osaka, Japan) in PBS for 30 min at 4°C and then washed (3×5 min) with PBS/0.1% bovine serum albumin (BSA) (Iwai Kagaku Co., Tokyo, Japan). Cells were incubated with a primary antibody in PBS with 1% BSA for 1 h at room temperature. Thereafter, cells were washed and incubated with fluorophore-conjugated secondary antibodies and 4′,6-diamidino-2-phenylindole (DAPI) for 1 h at room temperature. Immunofluorescent images were captured using a Leica TCS SP5 v2.0 system (Leica, Heidelberg, Germany). The following primary antibodies were used: anti-Oct3/4 mouse monoclonal antibody (1:50, C-10; Santa Cruz Biotechnology, Inc.), anti-Sox2 rabbit polyclonal antibody (1:100, poly6309; BioLegend, San Diego, CA, USA), anti-β-III tubulin mouse monoclonal antibody (1:500, Tuj-1; Covance Laboratories, Princeton, NJ, USA), anti-myosin heavy chain mouse monoclonal antibody (1:50, MF20; R&D Systems, Minneapolis, MN, USA), and anti-Gata4 mouse monoclonal antibody (1:50; Santa Cruz Biotechnology, Inc.). Alexa Fluor (488 or 568)-conjugated goat anti-mouse or goat anti-rabbit secondary antibodies (Invitrogen Life Technologies) were used at 1:1,000 dilutions.

Teratoma formation

Approximately 5×105 cells were injected under kidney capsules of nude mice. Tumors were dissected after 4–5 weeks and fixed in 10% buffered formalin. Tumor tissues were embedded in paraffin wax, sectioned, and examined after hematoxylin and eosin staining.

Blastocyst injection and generation of chimeric rats

Because collection of many blastocysts from Brown Norway rats is inefficient, we attempted the chimera formation assay using Brown Norway as well as Wistar rats. Rat blastocysts at E4.5 days were collected on the day of injection and cultured for 2–3 h to ensure cavitation. ESCs were disaggregated using Accutase, and 10–12 cells were injected into blastocyst cavities. Injected embryos were transferred into uteri of pseudopregnant rats.

Dead embryos were collected from uteri by cesarean section. For Tsc2 genotyping PCR, genomic DNA was obtained from several parts of each embryo or pup.

Gene expression microarray analysis

The Rat Affymetrix GeneChip Gene 1.0 ST Array (Affymetrix, Inc., Santa Clara, CA, USA) was used for microarray analysis. Amplification and labeling of probes and hybridization were performed according to the manufacturer’s instructions. Hierarchical clustering analysis was performed using GeneSpring software version 12.1 (Agilent Technologies, Inc., Santa Clara, CA, USA).

Results

Establishment of Tsc2-deficient stem cells from Eker rat embryos

After mating of double heterozygous Eker rats, a total of 34 blastocysts were collected. Zonae pellucidae were removed, and most blastocysts were successfully cultured on feeder cells, revealing outgrowths from embryonic fibroblasts (MEFs) in N2B27 medium supplemented with 2i + LIF (33). After several passages, a total of 26 cell lines were established. We routinely passaged these cells every 2–4 days by dissociating them into single cells and replating onto new feeder cells. They grew as dome-shaped or spherical colonies and were maintained for >25 passages without losing their morphology (Fig. 1A). A majority of colonies expressed alkaline phosphatase, an indicator of stem cell character (Fig. 1A). Thereafter, we checked Tsc2 genotypes of established cell lines by PCR. Surprisingly, we identified that not only Tsc2+/+ and Tsc2+/− cell lines but also Tsc2−/− cell lines had been established (Fig. 1B). Considering that previous reports had indicated that Tsc2 is necessary for the maintenance of stem cell characteristics, this result was unexpected. Both male and female cell lines were established for each genotype (Fig. 1C). Chromosome analysis revealed that most cell lines had normal ploidy (n=42, data not shown). RT-PCR analysis revealed that Tsc2−/− cells expressed the pluripotency markers Oct4, Sox2, and Nanog (Fig. 2A). Oct4 and Sox2 expressions were confirmed by immunofluorescence microscopy (Fig. 2B). These results indicate that Tsc2-deficient stem cells could be established from Eker rat embryos. We performed further experiments using two independent cell lines of each genotype.

Activation of the mTORC1 pathway in Tsc2−/− ESCs

To evaluate the mTORC1 activation status, we analyzed ESCs by western blotting. Tsc2 protein was detected in Tsc2+/+ and Tsc2+/− cells but not in Tsc2−/− cells, thereby confirming results of the genotype analysis. Tsc1 protein levels were decreased in Tsc2−/− ESCs, thereby reflecting the reciprocal stabilization between Tsc1 and Tsc2 proteins (42). As expected, an increase in S6 phosphorylation was detected in Tsc2−/− cells compared with that in Tsc2+/+ and Tsc2+/− cells, which indicates abnormal activation of the mTORC1 pathway in Tsc2−/− ESCs (Fig. 3). These results indicate that despite abnormal activation of the mTORC1 pathway, Tsc2−/− ESCs can be established.

In vitro differentiation of Tsc2−/− ESCs into three germ layers

Using the EB formation assay, we evaluated the differentiation potential of the established cell lines. We assessed the expression of differentiation markers by RT-PCR. Expression of markers for ectoderm (Nestin), endoderm (Sox17), and mesoderm (Flk1) were all observed in EBs (Fig. 4A). We plated EBs onto Matrigel-coated dishes and assessed their differentiation status by immunofluorescent staining for β-III tubulin (neuroectoderm), myosin (mesoderm), and Gata4 (endoderm) (Fig. 4B). Not only Tsc2+/+ and Tsc2+/− cells but also Tsc2−/− cells demonstrated the potential to differentiate into all three germ layers. In addition, we observed spontaneously beating areas in EBs of all Tsc2 genotypes (data not shown). These results suggest that most differentiation processes of ESCs were not blocked by Tsc2 deficiency.

Differentiation of Tsc2−/− ESCs into multiple lineages in teratomas

When Tsc2−/− ESCs were transplanted under the kidney capsule of nude mice, they differentiated into tissues derived from all three germ layers, including gut-like epithelium (endoderm), cartilage and adipocytes (mesoderm), stratified squamous epithelium, and neuroepithelium (ectoderm) (Fig. 5). These results indicate that Tsc2−/− ESCs are multipotent, although detailed characterization of each of the differentiated tissues remains to be elucidated. Interestingly, we observed that abnormal ductal structures appeared in Tsc2−/− teratomas (Kawano H, et al, unpublished data). Further characterization of these abnormal structures is described in another report (Kawano H, et al, unpublished data).

Contribution of Tsc2+/+ ESCs in chimeras

Next, to determine the ability of established ESCs to form chimeras, we injected Tsc2+/+ and Tsc2−/− ESCs into blastocysts of Wistar rats or Brown Norway rats (Materials and methods). Although ratios were low, four chimeras with black coat color were born from Wistar blastocysts injected with Tsc2+/+ ESCs, indicating the contribution of ESCs from the Eker rat strain (Fig. 6A). In contrast, we were unable to obtain pups demonstrating chimeric coat color in repeated trials using two independent Tsc2−/− ESCs. However, in these trials, we detected dead embryos in the uterus of recipient mother rats at term (Fig. 6B1 and 2). The appearance of dead embryos suggested developmental retardation. In addition, one live pup was delivered by cesarean section but died shortly after birth (Fig. 6B3). This pup revealed various morphological abnormalities such as an enlarged trunk. PCR genotyping of dead embryos and the pup indicated the contribution of Tsc2−/− ESCs in their tissues (Fig. 6C). On the basis of the band pattern of two dead embryos, we concluded that they had a greater contribution of Tsc2−/− ESCs compared with the live pup. These results suggest that a greater contribution of Tsc2−/− ESCs in the chimera results in embryonic lethality. Although germline transmission has not been confirmed yet, the contribution in chimeras suggests that ESCs established in this study possessed characteristics of multipotent stem cells.

Distinct gene expression pattern in Tsc2−/− ESCs on microarray analysis

To compare gene expression profiles of established ESCs, we employed microarray analysis (Fig. 7). Similar expression levels of pluripotency-related genes were identified in all these cells. Moreover, hierarchical clustering analysis revealed that gene expression profiles of Tsc2+/+ and Tsc2+/− ESCs resembled each other, but those of Tsc2−/− ESCs revealed an apparently distinct pattern. These results suggest that the homozygous Tsc2 mutation causes extensive gene expression changes in rat ESCs.

Discussion

In this study, we successfully established Tsc2−/− ESCs from Eker rats. These cells possessed characteristic features of ESCs, including expression of pluripotency markers, long-term self-renewal, and the capacity to differentiate into derivatives of all three germ layers. Although detailed mechanisms are still not clear, there have been several reports indicating the importance of the Tsc2-mTOR pathway in stem cell maintenance and differentiation (38,39,43). Gan et al reported that Tsc1 is a critical regulator of self-renewal, mobilization, and multilineage development in hematopoietic stem cells and that it executes these phenomena via both mTORC1-dependent and -independent pathways (39). Further, it was reported that the activation of S6K by expression of the constitutively active S6K1 or siRNA-mediated knockdown of TSC2 and RICTOR induced differentiation of human ESCs (38). Recently, Betschinger et al reported that siRNA-mediated knockdown of Tsc2 or Flcn inhibits differentiation of ESCs (43). In contrast to results in these reports, we successfully established Tsc2−/− ESCs possessing multipotent differentiation capacity despite the presence of the activated mTORC1 pathway. There are several possible reasons why the derivation of Tsc2−/− ESCs was possible in this study. Previous studies utilized siRNA- or shRNA-mediated knockdown of Tsc2 in already established ESCs or conditional knockout of Tsc1 in somatic stem cells. Such ‘acute’ downregulation of Tsc1/Tsc2 may cause some aberrant gene regulation that restrains the maintenance of the multipotent nature and differentiation capacity of stem cells. Microarray analysis revealed a distinct gene expression pattern in Tsc2−/− ESCs compared with their Tsc2+/+ and Tsc2+/− counterparts. Our system enables comparison of gene expression profiles between Tsc2−/− ESCs and Tsc2+/− ESCs with Tsc2 knockdown. Such analysis is of interest to further explore Tsc2 mutation-related pathogenesis.

We were unable to obtain pups demonstrating chimeric coat color using Tsc2−/− ESCs. Results of dead embryos suggested that higher contribution of Tsc2−/− cells in chimeras induced embryonic lethality. Further, it has been reported that when human TSC2-deficient fibroblast-like cells were grafted into mice, differentiated tissues revealed features of TSC skin tumors and that TSC2-deficient cells directly or indirectly induce abnormal follicular neogenesis and epidermal proliferation (44). Because Tsc2−/− ESCs may cause abnormal differentiation of hair in chimeras, it may not be appropriate to determine the contribution of ESCs on the basis of hair color of chimeras.

He et al reported that reprogramming of somatic cells derived from Tsc2−/− mouse embryos to iPSCs was not possible (40). In this study, we provide evidence that the derivation of Tsc2−/− ESCs from Eker rat embryos is possible. In somatic cells, some epigenetic abnormalities caused by Tsc2 deficiency may not be corrected even under reprogramming conditions. Conversely, during early embryogenesis, epigenetic abnormalities in Tsc2−/− cells may be tuned to maintain the stemness. With reprogramming experiments using Eker rat-derived embryonic fibroblasts, ESCs established in this study will serve as useful tools to compare effects of Tsc2 deficiency on epigenetic status in reprogramming and ESC derivation.

In recent years, various patient-derived iPSCs have been used for in vitro differentiation experiments to mimic the pathogenesis of human diseases (45,46). Moreover, such cellular models are useful to research novel drug target molecules by high-throughput screening (47). With regard to tumorigenesis, tissue specificity and abnormal differentiation are relevant to its molecular basis. Lineage-specific in vitro differentiation of tumor suppressor-deficient ESCs will provide valuable experimental models to explore the mechanism of pathogenesis. However, in humans, establishment of tumor suppressor-deficient (i.e., homozygously inactivated) ESCs or iPSCs has been technically difficult. In rodents, homozygous mutant ESCs for tumor suppressors, including Rb, Tp53, and Apc, have been established (4850). To date, none of those ESCs have been extensively used for in vitro differentiation experiments. For example, Apc-deficient ESCs failed to differentiate into multiple lineages in the teratoma formation assay, suggesting that the induction of various cell types was not applicable to these ESCs (50). In contrast, Tsc2−/− ESCs exhibited the potential to differentiate into all germ layers and multiple cell lineages, both in vitro and in vivo. We already observed development of abnormal ductal structures in Tsc2−/− teratomas, suggesting that cell type-specific effects of Tsc2 deficiency could be reproduced in differentiation of ESCs (Kawano H, et al, unpublished data). Combined with in vivo experiments, in vitro differentiation models using ESCs established in this study will facilitate understanding of Tsc2 mutation-related pathogenesis as well as aid in the search for therapeutic target pathways.

Acknowledgements

We thank Takako Ikegami, and Tomomi Ikeda, Laboratory of Molecular and Biochemical Research, Research Support Center, Juntendo University Graduate School of Medicine (Tokyo, Japan) for technical assistance. The authors would like to thank Enago (www.enago.jp) for the English language review. This study was supported in part by the following grants: Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) (Japan); MEXT-Supported Program for the Strategic Research Foundation at Private Universities; Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (Japan); and Grants-in-Aid for Scientific Research from the Ministry of Health, Labour and Welfare (Japan). This study was also supported by the Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine.

References

1 

Kwiatkowski DJ, Whittemore VH and Thiele EA: Tuberous Sclerosis Complex: Genes, Clinical Features, and Therapeutics. Wiley-VCH Verlag GmbH & Co. KGaA; Weinheim: 2010, View Article : Google Scholar

2 

European Chromosome 16 Tuberous Sclerosis Consortium. Identification and characterization of the tuberous sclerosis gene on chromosome 16. Cell. 75:1305–1315. 1993. View Article : Google Scholar : PubMed/NCBI

3 

van Slegtenhorst M, de Hoogt R, Hermans C, et al: Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science. 277:805–808. 1997. View Article : Google Scholar : PubMed/NCBI

4 

Curatolo P, Bombardieri R and Jozwiak S: Tuberous sclerosis. Lancet. 372:657–668. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Laplante M and Sabatini DM: mTOR signaling in growth control and disease. Cell. 149:274–293. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Knudson AG Jr: Mutation and cancer: Statistical study of retino-blastoma. Proc Natl Acad Sci USA. 68:820–823. 1971. View Article : Google Scholar

7 

Henske EP, Scheithauer BW, Short MP, Wollmann R, Nahmias J, Hornigold N, van Slegtenhorst M, Welsh CT and Kwiatkowski DJ: Allelic loss is frequent in tuberous sclerosis kidney lesions but rare in brain lesions. Am J Hum Genet. 59:400–406. 1996.PubMed/NCBI

8 

Au KS, Hebert AA, Roach ES and Northrup H: Complete inactivation of the TSC2 gene leads to formation of hamartomas. Am J Hum Genet. 65:1790–1795. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Tucker T and Friedman JM: Pathogenesis of hereditary tumors: Beyond the ‘two-hit’ hypothesis. Clin Genet. 62:345–357. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Chan JA, Zhang H, Roberts PS, Jozwiak S, Wieslawa G, Lewin-Kowalik J, Kotulska K and Kwiatkowski DJ: Pathogenesis of tuberous sclerosis subependymal giant cell astrocytomas: Biallelic inactivation of TSC1 or TSC2 leads to mTOR activation. J Neuropathol Exp Neurol. 63:1236–1242. 2004.PubMed/NCBI

11 

Niida Y, Stemmer-Rachamimov AO, Logrip M, Tapon D, Perez R, Kwiatkowski DJ, Sims K, MacCollin M, Louis DN and Ramesh V: Survey of somatic mutations in tuberous sclerosis complex (TSC) hamartomas suggests different genetic mechanisms for pathogenesis of TSC lesions. Am J Hum Genet. 69:493–503. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Onda H, Lueck A, Marks PW, Warren HB and Kwiatkowski DJ: Tsc2(+/−) mice develop tumors in multiple sites that express gelsolin and are influenced by genetic background. J Clin Invest. 104:687–695. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Kobayashi T, Minowa O, Kuno J, Mitani H, Hino O and Noda T: Renal carcinogenesis, hepatic hemangiomatosis, and embryonic lethality caused by a germ-line Tsc2 mutation in mice. Cancer Res. 59:1206–1211. 1999.PubMed/NCBI

14 

Kobayashi T, Minowa O, Sugitani Y, Takai S, Mitani H, Kobayashi E, Noda T and Hino O: A germ-line Tsc1 mutation causes tumor development and embryonic lethality that are similar, but not identical to, those caused by Tsc2 mutation in mice. Proc Natl Acad Sci USA. 98:8762–8767. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Meikle L, Pollizzi K, Egnor A, Kramvis I, Lane H, Sahin M and Kwiatkowski DJ: Response of a neuronal model of tuberous sclerosis to mammalian target of rapamycin (mTOR) inhibitors: Effects on mTORC1 and Akt signaling lead to improved survival and function. J Neurosci. 28:5422–5432. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Goorden SMI, van Woerden GM, van der Weerd L, Cheadle JP and Elgersma Y: Cognitive deficits in Tsc1+/− mice in the absence of cerebral lesions and seizures. Ann Neurol. 62:648–655. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Ehninger D, Han S, Shilyansky C, Zhou Y, Li W, Kwiatkowski DJ, Ramesh V and Silva AJ: Reversal of learning deficits in a Tsc2+/− mouse model of tuberous sclerosis. Nat Med. 14:843–848. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Eker R and Mossige J: A dominant gene for renal adenomas in the rat. Nature. 189:858–859. 1961. View Article : Google Scholar

19 

Hino O, Mitani H and Knudson AG: Genetic predisposition to transplacentally induced renal cell carcinomas in the Eker rat. Cancer Res. 53:5856–5858. 1993.PubMed/NCBI

20 

Kobayashi T, Hirayama Y, Kobayashi E, Kubo Y and Hino O: A germline insertion in the tuberous sclerosis (Tsc2) gene gives rise to the Eker rat model of dominantly inherited cancer. Nat Genet. 9:70–74. 1995. View Article : Google Scholar : PubMed/NCBI

21 

Rennebeck G, Kleymenova EV, Anderson R, Yeung RS, Artzt K and Walker CL: Loss of function of the tuberous sclerosis 2 tumor suppressor gene results in embryonic lethality characterized by disrupted neuroepithelial growth and development. Proc Natl Acad Sci USA. 95:15629–15634. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Mizuguchi M, Takashima S, Yamanouchi H, Nakazato Y, Mitani H and Hino O: Novel cerebral lesions in the Eker rat model of tuberous sclerosis: Cortical tuber and anaplastic ganglioglioma. J Neuropathol Exp Neurol. 59:188–196. 2000.PubMed/NCBI

23 

Yeung RS, Katsetos CD and Klein-Szanto A: Subependymal astrocytic hamartomas in the Eker rat model of tuberous sclerosis. Am J Pathol. 151:1477–1486. 1997.PubMed/NCBI

24 

Takahashi DK, Dinday MT, Barbaro NM and Baraban SC: Abnormal cortical cells and astrocytomas in the Eker rat model of tuberous sclerosis complex. Epilepsia. 45:1525–1530. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Prabhakar S, Goto J, Zhang X, et al: Stochastic model of Tsc1 lesions in mouse brain. PLoS One. 8:e642242013. View Article : Google Scholar : PubMed/NCBI

26 

Kobayashi T, Adachi H, Mitani H, Hirayama Y and Hino O: Toward chemotherapy for Tsc2-mutant renal tumor. Proc Jpn Acad. 79:22–25. 2003. View Article : Google Scholar

27 

Kenerson HL, Aicher LD, True LD and Yeung RS: Activated mammalian target of rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal tumors. Cancer Res. 62:5645–5650. 2002.PubMed/NCBI

28 

Lee L, Sudentas P, Donohue B, et al: Efficacy of a rapamycin analog (CCI-779) and IFN-γ in tuberous sclerosis mouse models. Genes Chromosomes Cancer. 42:213–227. 2005. View Article : Google Scholar

29 

McCormack FX, Inoue Y, Moss J, et al: National Institutes of Health Rare Lung Diseases Consortium; MILES Trial Group: Efficacy and safety of sirolimus in lymphangioleiomyomatosis. N Engl J Med. 364:1595–1606. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Franz DN, Belousova E, Sparagana S, et al: Efficacy and safety of everolimus for subependymal giant cell astrocytomas associated with tuberous sclerosis complex (EXIST-1): A multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 381:125–132. 2013. View Article : Google Scholar

31 

Bissler JJ, Kingswood JC, Radzikowska E, et al: Everolimus for angiomyolipoma associated with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis (EXIST-2): A multicentre, randomised, double-blind, placebo-controlled trial. Lancet. 381:817–824. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Mizuguchi M: Abnormal giant cells in the cerebral lesions of tuberous sclerosis complex. Congenit Anom (Kyoto). 47:2–8. 2007. View Article : Google Scholar

33 

Buehr M, Meek S, Blair K, Yang J, Ure J, Silva J, McLay R, Hall J, Ying QL and Smith A: Capture of authentic embryonic stem cells from rat blastocysts. Cell. 135:1287–1298. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Li P, Tong C, Mehrian-Shai R, et al: Germline competent embryonic stem cells derived from rat blastocysts. Cell. 135:1299–1310. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Evans MJ and Kaufman MH: Establishment in culture of pluripotential cells from mouse embryos. Nature. 292:154–156. 1981. View Article : Google Scholar : PubMed/NCBI

36 

Martin GR: Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci USA. 78:7634–7638. 1981. View Article : Google Scholar : PubMed/NCBI

37 

Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS and Jones JM: Embryonic stem cell lines derived from human blastocysts. Science. 282:1145–1147. 1998. View Article : Google Scholar : PubMed/NCBI

38 

Easley CA IV, Ben-Yehudah A, Redinger CJ, Oliver SL, Varum ST, Eisinger VM, Carlisle DL, Donovan PJ and Schatten GP: mTOR-mediated activation of p70 S6K induces differentiation of pluripotent human embryonic stem cells. Cell Reprogram. 12:263–273. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Gan B, Sahin E, Jiang S, Sanchez-Aguilera A, Scott KL, Chin L, Williams DA, Kwiatkowski DJ and DePinho RA: mTORC1-dependent and -independent regulation of stem cell renewal, differentiation, and mobilization. Proc Natl Acad Sci USA. 105:19384–19389. 2008. View Article : Google Scholar : PubMed/NCBI

40 

He J, Kang L, Wu T, et al: An elaborate regulation of Mammalian target of rapamycin activity is required for somatic cell reprogramming induced by defined transcription factors. Stem Cells Dev. 21:2630–2641. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Shiono M, Kobayashi T, Takahashi R, et al: The G1556S-type tuberin variant suppresses tumor formation in tuberous sclerosis 2 mutant (Eker) rats despite its deficiency in mTOR inhibition. Oncogene. 27:6690–6697. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Benvenuto G, Li S, Brown SJ, Braverman R, Vass WC, Cheadle JP, Halley DJ, Sampson JR, Wienecke R and DeClue JE: The tuberous sclerosis-1 (TSC1) gene product hamartin suppresses cell growth and augments the expression of the TSC2 product tuberin by inhibiting its ubiquitination. Oncogene. 19:6306–6316. 2000. View Article : Google Scholar

43 

Betschinger J, Nichols J, Dietmann S, Corrin PD, Paddison PJ and Smith A: Exit from pluripotency is gated by intracellular redistribution of the bHLH transcription factor Tfe3. Cell. 153:335–347. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Li S, Thangapazham RL, Wang JA, Rajesh S, Kao TC, Sperling L, Moss J and Darling TN: Human TSC2-null fibroblast-like cells induce hair follicle neogenesis and hamartoma morphogenesis. Nat Commun. 2:2352011. View Article : Google Scholar : PubMed/NCBI

45 

Ebert AD, Yu J, Rose FF Jr, Mattis VB, Lorson CL, Thomson JA and Svendsen CN: Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature. 457:277–280. 2009. View Article : Google Scholar :

46 

Lee G, Papapetrou EP, Kim H, et al: Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature. 461:402–406. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Yang YM, Gupta SK, Kim KJ, et al: A small molecule screen in stem-cell-derived motor neurons identifies a kinase inhibitor as a candidate therapeutic for ALS. Cell Stem Cell. 12:713–726. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Williams BO, Schmitt EM, Remington L, Bronson RT, Albert DM, Weinberg RA and Jacks T: Extensive contribution of Rb-deficient cells to adult chimeric mice with limited histopathological consequences. EMBO J. 13:4251–4259. 1994.PubMed/NCBI

49 

Kawamata M and Ochiya T: Two distinct knockout approaches highlight a critical role for p53 in rat development. Sci Rep. 2:9452012. View Article : Google Scholar : PubMed/NCBI

50 

Kielman MF, Rindapää M, Gaspar C, van Poppel N, Breukel C, van Leeuwen S, Taketo MM, Roberts S, Smits R and Fodde R: Apc modulates embryonic stem-cell differentiation by controlling the dosage of beta-catenin signaling. Nat Genet. 32:594–605. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 46 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ito Y, Kawano H, Kanai F, Nakamura E, Tada N, Takai S, Horie S, Arai H, Kobayashi T, Hino O, Hino O, et al: Establishment of Tsc2‑deficient rat embryonic stem cells. Int J Oncol 46: 1944-1952, 2015
APA
Ito, Y., Kawano, H., Kanai, F., Nakamura, E., Tada, N., Takai, S. ... Hino, O. (2015). Establishment of Tsc2‑deficient rat embryonic stem cells. International Journal of Oncology, 46, 1944-1952. https://doi.org/10.3892/ijo.2015.2913
MLA
Ito, Y., Kawano, H., Kanai, F., Nakamura, E., Tada, N., Takai, S., Horie, S., Arai, H., Kobayashi, T., Hino, O."Establishment of Tsc2‑deficient rat embryonic stem cells". International Journal of Oncology 46.5 (2015): 1944-1952.
Chicago
Ito, Y., Kawano, H., Kanai, F., Nakamura, E., Tada, N., Takai, S., Horie, S., Arai, H., Kobayashi, T., Hino, O."Establishment of Tsc2‑deficient rat embryonic stem cells". International Journal of Oncology 46, no. 5 (2015): 1944-1952. https://doi.org/10.3892/ijo.2015.2913