Skip to main content
Erschienen in: BMC Cancer 1/2016

Open Access 01.12.2016 | Research article

Induction of artificial cancer stem cells from tongue cancer cells by defined reprogramming factors

verfasst von: Koji Harada, Tarannum Ferdous, Dan Cui, Yasuhiro Kuramitsu, Takuya Matsumoto, Eiji Ikeda, Hideyuki Okano, Yoshiya Ueyama

Erschienen in: BMC Cancer | Ausgabe 1/2016

Abstract

Background

The cancer stem cells (CSCs), a small subpopulation of cells in tumor are responsible for the tumor initiation, growth, recurrence and metastasis of cancer, as well as resistance of cancers to drugs or radiotherapy. CSCs are an important target for the development of novel strategies in cancer treatment. However, CSCs-targeted new anti-cancer drug discovery is currently hindered by the lack of easy and reliable methods for isolating, collecting and maintaining sufficient number of CSCs. Here, we examined whether introduction of defined reprogramming factors (Oct4, shp53, Sox2, Klf4, l-Myc and Lin28) into HSC2 tongue cancer cells could transform the HSC2 into HSC2 with CSCs properties.

Methods

We introduced the defined reprogramming factors into HSC2 tongue cancer cells via episomal vectors by electroporation method to generate transfectant cells. We investigated the malignant properties of the transfectant cells by cell proliferation assay, migration assay, wound healing assay, sphere formation assay, chemosensitivity and radiosensitivity assay in vitro; and also examined the tumorigenic potential of the transfectants in vivo.

Results

The transfectant cells (HSC2/hOCT3/4-shp53-F, HSC2/hSK, HSC2/hUL, HSC2/hOCT3/4-shp53-F + hSK, HSC2/hOCT3/4-shp53-F + hUL, HSC2/hSK + hUL, HSC2/hOCT3/4-shp53-F + hSK + hUL) displayed a malignant phenotype in culture and form tumors on the back of nude mice more efficiently than parental HSC2 and control HSC2/EGFP transfectant cells. They exhibited increased resistance to chemotherapeutic agents; 5-fluorouracil, cisplatin, docetaxel, trifluorothymidine, zoledronic acid, cetuximab, bortezomib and radiation when compared with HSC2 and HSC2/EGFP. Among all the transfected cells, HSC2/hOCT3/4-shp53-F + hSK + hUL cell containing all of the reprogramming factors showed the most aggressive and malignant properties and presented the highest number of spheres in the culture medium containing human recombinant fibroblast Growth Factor-2 (FGF-2) and epidermal Growth Factor (EGF).

Conclusion

These findings suggest that artificial cancer stem cells obtained by the induction of cellular reprogramming may be useful for investigating the acquisition of potential malignancy as well as screening the CSCs-targeting drugs.

Background

Head and neck squamous cell carcinoma (HNSCC), including oral squamous cell carcinoma (OSCC) is a major health problem throughout the world. Despite advances in treatment of HNSCC, mortality from this disease remains high because of local and regional recurrences of tumors, acquired resistance of patients to chemotherapy or radiotherapy and high rate of metastases at the advanced stage of the disease [1]. Therefore, understanding the initial steps of the tumorigenic processes, as well as tumor progression and metastasis is required for the improvement of prognosis of OSCC and survival of the patients [2].
There are accumulating evidences that cancer cells are functionally heterogeneous and they go through proliferation, differentiation and maturation to a certain extent [3]. A solid tumor contains a distinct subpopulation of cells with stem cell properties (CSCs) that play important roles in cancer initiation, progression, recurrence and metastasis [47]. These CSCs are capable of long-term self-renewal and can generate phenotypically diverse tumor cell populations. In addition, they show resistance to radiation and chemotherapeutic agents [810]. Therefore, the development of new strategies for CSCs-targeted therapy has attracted attention in these years. Identification and characterization of CSC populations are important for the development of novel strategies for cancer treatment. Separation or generation of a sufficient number of CSCs from tumor tissues and amplification of the CSCs while stably maintaining them in an undifferentiated state in vitro is a pre-requisite to ensure a large-scale drug screening process [11]. CSCs could be identified and isolated through functional assays such as detection of the expression of the cell surface markers specific for CSCs or the sphere body formation method [1214]. Until now, a number of CSC markers are identified in HNSCC, eg - CD44, CD44v4, CD44v6, ALDH1, CD166, and CD133 [1519]. There are several other CSC markers reported in different cancer types, eg- CD34, CD133, CD90, CD13, EpCAM, ABCG, ALDH1 etc. [8]. Moreover, Side Population (SP) cells, which are a subpopulation of tumor cells with increased efflux capacity are also considered as a source of CSCs specially where CSC molecular markers are unknown [20, 21]. However, these methods that are dependent upon the isolation of CSCs from tumor tissues by flow cytometry or generation of spheres bodies have some real limitations [1214]. Therefore, the isolation of sufficient number CSCs from tumor tissues is still difficult. Until now, an easy, cost-saving and optimal cell culture method for successfully amplifying and maintaining pure CSC populations is also unknown. Thus, screening of CSCs-targeting drugs in vitro and in animal models remain in a difficult situation. The development of an easy and efficient method to manufacture artificial CSCs is crucial to overcome these research obstacles.
It is believed that CSCs undergo differentiation into non-CSCs (cancer progeny cells), and that CSCs reach to the top of hierarchical system composed of cancer cells [2225]. If non-CSCs can be dedifferentiated, CSCs may be generated artificially. Like normal pluripotent stem cells, CSCs are long-lived, and display quiescent potentials in a dormant state, and are responsible for angiogenic induction, apoptotic resistance and differentiation. CSC cells express stem cell marker genes, including Oct4, Sox2, Nanog, c-kit, ABCG2, and ALDH [3, 2628]. Among these genes, Oct4 is the key transcription factor that maintains the pluripotency and self-renewal in undifferentiated embryonic stem cells [27]. Additionally, the essential role of c-Myc and Klf4 in the regulation and maintenance of the stem cell-like features of tumor CSCs was reported by Wang et al. and Yu et al. respectively [29, 30].
The well-known work of generation of induced pluripotent stem cells (iPSCs) by Takahashi and Yamanaka showed that adult somatic cells can be reprogrammed to become pluripotent by the introduction of the pluripotent stem cell genes Oct4, Sox2, Klf4 and c-Myc [31, 32]. Additionally, Okita et al. mentioned the importance Lin28 and l-myc for the generation of human iPSCs from blood cells [33, 34]. The iPSCs development process shares many features with cancer development. Such similarities indicate that iPSCs reprogramming processes and carcinogenesis might be promoted by overlapping mechanisms; during which, somatic differentiated cell undergoes transcriptional changes and acquires self-renewal and unlimited proliferation capabilities [3537]. Ohnishi et al. showed that, somatic cells that deviated successful reprogramming failed to develop iPSCs, but behaved similarly to cancer cells and developed Wilms tumor, a childhood blastoma in the kidney [38]. Thus, the same reprogramming factors that generate iPSCs could be also involved in carcinogenic transformation of normal somatic cells. Additionally, in neurosphere culture conditions, introduction of Oct4, Sox2, c-Myc and Klf4 directly induced neural stem cells (NSCs) properties in somatic cells such as skin fibroblasts, which suggests that these reprogramming factors might possess the ability to induce stemness in somatic cells [3942].
In this study, we followed the iPSCs-generation protocol obtained from the Center for iPS cell research and application (CiRA) website to reprogram HSC2 tongue cancer cells into CSCs [43]. We introduced l-Myc instead of c-Myc and two other factors (shp53 and Lin28) along with Oct4, Sox2 and Klf4 into HSC2 cells via episomal vector; instead of using only Oct4, Sox2, Klf4 and c-Myc with retroviral vectors as initially described by Takahashi and Yamanaka [3133, 43]. The resultant cells possess the hallmarks of CSCs and could efficiently generate tumors in a nude mouse model. These results suggest that introduction of defined reprogramming factors can possibly dedifferentiate oral cancer cells into CSCs and can provide a potentially valuable system for the study of CSCs.

Methods

Cell culture

HSC2 cells were purchased from Cell Bank, RIKEN BioResource Center (Ibaraki, Japan). Cells were cultured in a 1:1 mixture of Dulbecco’s modified Eagle’s medium (D-MEM)/Ham’s F-12 (Wako Pure Chemical Industries, Ltd. Osaka, Japan) supplemented with 10 % fetal bovine serum (FBS) (Thermo Fisher scientific Inc., Waltham, MA, USA), 100 μg/ml streptomycin, 100 units/ml penicillin (Thermo Fisher scientific) in a humidified atmosphere containing 5 % CO2 at 37 °C. The electroporated cells, ie - HSC2/EGFP, HSC2/hOCT3/4-shp53-F, HSC2/hSK, HSC2/hUL, HSC2/hOCT3/4-shp53-F + hSK, HSC2/hOCT3/4-shp53-F + hUL, HSC2/hSK + hUL, HSC2/hOCT3/4-shp53-F + hSK + hUL were cultured in the same culture medium without any selection agents.

Cell reprogramming and transfection

Episomal vectors (pCXLE-hOCT3/4-shp53-F, pCXLE-hSK, pCXLE-hUL and pCXLE-EGFP) were obtained from Addgene (Cambridge, MA, USA) and introduced into HSC2 cells in various combinations. An expression plasmid mixture containing one or more of these episomal vectors (1 μg of each vector) were electroporated into 6 × 105 HSC2 cells with Neon Transfection System (Thermo Fisher scientific) using a 100 μl kit according to the manufacturer’s instructions (conditions for electroporation: pulse voltage: 1550 or 1650 V, pulse width: 10 ms, pulse number: 3). In the same way, we inserted pCXLE-EGFP only into HSC2 cells to obtain HSC2/EGFP as a control. The list of expression plasmid mixtures used in the experiments and the resultant cells is shown in Table 1.
Table 1
Summary of plasmid mixtures for electroporation
Mix-ture
Plasmid name
Amount (μg)
Genes
Resultant cell
1
pCXLE-EGFP
1
Egfp
HSC2/EGFP
2
pCXLE-hOCT3/4-shp53-F
1
Oct4, shp53, Egfp
HSC2/hOCT3/4-shp53-F
pCXLE-EGFP
1
3
pCXLE-hSK
1
Sox2, Klf4, Egfp
HSC2/hSK
pCXLE-EGFP
1
4
pCXLE-hUL
1
l-Myc, Lin28, Egfp
HSC2/hUL
pCXLE-EGFP
1
5
pCXLE-hOCT3/4-shp53-F
1
Oct4, shp53,Sox2, Klf4, Egfp
HSC2/hOCT3/4-shp53-F + hSK
pCXLE-hSK
1
pCXLE-EGFP
1
6
pCXLE-hOCT3/4-shp53-F
1
Oct4, shp53, l-Myc, Lin28, Egfp
HSC2/hOCT3/4-shp53-F + hUL
pCXLE-hUL
1
pCXLE-EGFP
1
7
pCXLE-hSK
1
Sox2, Klf4, l-Myc, Lin28, Egfp
HSC2/hSK + hUL
pCXLE-hUL
1
pCXLE-EGFP
1
8
pCXLE-hOCT3/4-shp53-F
1
Oct4, shp53, Sox2, Klf4, l-Myc, Lin28, Egfp
HSC2/hOCT3/4-shp53-F + hSK + hUL
pCXLE-hSK
1
pCXLE-hUL
1
pCXLE-EGFP
1

Cell proliferation assay

HSC2 or each transfectant (5 × 103 cells per well) were seeded on 96-well plates (Becton Dickinson Labware, Franklin lakes, NJ, USA) in D-MEM/Ham’s F-12 medium supplemented with 10 % FBS and 1 % penicillin/streptomycin. After 48 h or 72 h, 3-(4, 5-dimethylthiazol- 2-yl)-2, 5- diphenyltetrazolium bromide (MTT; Sigma-Aldrich, St. Louis, MO, USA) was added to each well (25 μl/well) and incubated for 4 h. Then dimethyl sulfoxide (100 μl/well) was added to each well and a spectrophotometer (BioRad Laboratories, Hercules, CA, USA) was used to measure the absorbance at 490 nm (Optical Density 490 or OD490). All assays were run in triplicate.

Cell migration assay

Cell migration assay was performed using a Boyden chamber according to the manufacturer’s instructions, (Neuro Probe, Gaithersburg, MD, USA). 5 × 103 cells in 50 μl D-MEM/Ham’s F-12 medium without FBS were seeded on a gelatin coated polycarbonate membrane. In the lower chamber, 25 μl D-MEM/Ham’s F-12 with 10 % FBS was added as chemoattractant. After the cells were incubated for 24 h at 37 °C in a 5 % CO2 atmosphere, the polycarbonate membrane was washed with PBS, and cells on the top surface of the polycarbonate membrane were removed with a cotton swab. Cells adhering to the lower surface were fixed with methanol, stained with Hematoxylin solution and counted under a microscope in five predetermined fields (200×). All assays were independently repeated at least three times.

Wound healing assay

Cells (15 × 103 cells per well) were seeded into 24-well plate (Becton Dickinson Labware) and were cultured in D-MEM/Ham’s F-12 with 10 % FBS and 1 % penicillin/streptomycin until a monolayer of cells were formed. A 200 μl pipette tip was used to gently wound cell layer through the central axis of the plate. The migration of cells into the wounded area was observed at 24 h by a microscope (BX-51-33-FLD2, OLYMPUS, PA, USA). The cell wound closure rate was calculated using the following equation: Wound closure = [1 − (wound area at Tt/wound area at T0) × 100, where Tt is the time passed since wounding (24 h) and T0 is the time of initial wounding. The experiments were performed in triplicate.

Tumor sphere formation assay

Cells were placed at a density of 1000 cells/well in sphere-culture medium consisting of serum-free DMEM/F-12 (Ham) 1:1 (Prototype) medium (Thermo Fisher scientific), N2 supplement (Thermo Fisher scientific), 10 ng/ml FGF-2 (Thermo Fisher scientific), and 10 ng/ml EGF (Thermo Fisher scientific) in a ultra-low attachment 96-well plate (Corning, New York, NY, USA) to generate primary sphere bodies. Every four days, 50 μL of the fresh growth medium was added. The numbers of spheres larger than 20 μm in diameter were counted after two weeks of culture.

Western blotting

Whole cell lysates were prepared using Radioimmunoprecipitation assay (RIPA) buffer (Thermo Fisher scientific) and were subjected to electrophoresis on 10 % SDS-polyacrylamide gels (Thermo Fisher scientific), and then transferred to a PVDF membrane (Thermo Fisher scientific). After blocking, the membranes were incubated with the anti-HCAM (CD44) mouse monoclonal antibody (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and anti-CD13 rabbit monoclonal antibody (Epitomics Inc., Burlingame, CA, USA) followed by Novex® alkaline-phosphatase conjugated (goat) anti-rabbit or (goat) anti-mouse immunoglobulin G (IgG) secondary antibody (Thermo Fisher scientific). The antibodies were detected using a chromogenic immunodetection system, WesternBreeze (Thermo Fisher scientific) according to the manufacturer's instructions. Also, anti- α- tubulin monoclonal antibody (Santa Cruz Biotech.) was used for normalization of Western blot analysis.

Chemosensitivity and radiosensitivity assessment

Cells (5 × 103 cells per well) were seeded on 96-well plates (Becton Dickinson Labware) in D-MEM/Ham’s F-12 with 10 % FBS and 1 % penicillin/streptomycin. Twenty four hours later, cells were either remained untreated or were treated with any one of the following drugs: 2 μg/ml 5-fluorouracil (5-FU), 1 μg/ml cisplatin (CDDP), 100 pg/ml docetaxel (DOC), 50 μg/ml trifluorothymidine (TFT), 1 μg/ml cetuximab, 5 ng/ml bortezomib or 10 μg/ml zoledronic acid. Cells were also exposed to 15 Gy radiation in an X-ray irradiator (MBR-1505R2, 150 kV, 5 mA, filter: 1.0 mm aluminum, Hitachi Medico, Tokyo, Japan). After 48 h, 25 μl MTT was added to each well. After 4 h, dimethyl sulfoxide (100 μl/well) was added and the absorbance was measured with a spectrophotometer (BioRad Laboratories) at 490 nm. All assays were run in triplicate.

In vivo tumor formation assay

Cells (1 × 104 ~ 1 × 106) were washed twice with antibiotic-free and serum-free D-MEM/Ham’s F-12 medium and finally re-suspended in 0.1 ml of saline. The cell suspension was injected subcutaneously into 5-week-old BALB/c nude mice (CLEA, Tokyo Japan). Tumor size was monitored and measured weekly for 4 weeks. The estimated tumor volume was calculated as 0.5 × length × width2. All mice were sacrificed at the end of 4 weeks/28 days. The tumors were dissected out, fixed in neutral-buffered formalin, embedded in paraffin and stained with hematoxylin and eosin. The mice were housed in a pathogen-free environment under a 12 h light/dark cycle, and provided with sterile water and food ad libitum. All studies and experiments conformed to the Guidelines for Animal Experimentation of Yamaguchi University (Ube, Japan).

Results

Establishment of transfectants by defined reprogramming factors

We successfully introduced the reprogramming factors (Oct4, shp53, Sox2, Klf4, l-Myc and Lin28) and Egfp genes via the plasmid vectors (pCXLE-hOCT3/4-shp53-F, pCXLE-hSK, pCXLE-hUL and pCXLE-EGFP) into HSC2 cells by electroporation in order to obtain HSC2/EGFP, HSC2/hOCT3/4-shp53-F, HSC2/hSK, HSC2/hUL, HSC2/hOCT3/4-shp53-F + hSK, HSC2/hOCT3/4-shp53-F + hUL, HSC2/hSK + hUL and HSC2/hOCT3/4-shp53-F + hSK + hUL cells. Fluorescence microscopic observation of EGFP expression in transfectant cells showed the vector transplantation efficiency was about 50 % when the pulse voltage of the electroporator was 1650 V, and that about 30 % at 1550 V (data not shown). Therefore, the optimum condition for electroporation was set as; pulse voltage: 1650 V, pulse width: 10 ms, pulse number: 3. The transfectants were cultured in D-MEM/Ham’s F-12 medium supplemented with 10 % FBS, 1 % penicillin/streptomycin. In this study, we did not use any selection methods to identify stable transfectants. The transfected cells were bigger and spindle-shaped compared to HSC2 parental cells which had cobblestone morphology (Fig. 1) Moreover, each transfectant cells showed slightly different morphology than the other (Fig. 1).

Cell proliferation ability

MTT assay was used to measure the growth rate of the transfectants. The HSC2/hOCT3/4-shp53-F + hSK + hUL had higher proliferative ability than that of parental cell (HSC2) and HSC2/EGFP. At 48 h and 72 h of culture, the growth rate of HSC2/hOCT3/4-shp53-F + hSK + hUL was significantly higher than that of HSC2 and HSC2/EGFP, while the growth rate among parental HSC2 cell, HSC2/EGFP and the other transfectants was not significantly different (Fig. 2).

Migration ability

CSCs are endowed with high migratory ability. Therefore, we measured the migration activity of the transfectants using migration assay with Boyden chamber. The HSC2/hOCT3/4-shp53-F + hSK, HSC2/hOCT3/4-shp53-F + hUL, HSC2/hSK + hUL and HSC2/hOCT3/4-shp53-F + hSK + hUL had significantly higher migration ability than that of parental cell (HSC2) and HSC2/EGFP. Especially, the HSC2/hOCT3/4-shp53-F + hSK + hUL showed highest migration ability (Fig. 3).

Wound healing ability

Wound healing assay was performed to examine the migration capability of the transfectants. HSC2/hOCT3/4-shp53-F + hSK + hUL cell showed highest would healing capacity (wound closure: 85 %) compared to HSC2/EGFP (60 %), all the other transfectants (50–80 %) and the parental HSC2 cell (35 %). Interestingly, all the transfectant cells showed higher wound healing ability than that of parental HSC2 (Fig. 4a, b). HSC2/hUL + hSK cells showed second highest migration and wound healing capacity (80 %) which suggests that Sox2, Klf4, l-Myc, Lin28 factors are essential to increase the migration capacity of the transfectants.

Sphere formation

The transfectants, HSC2/EGFP and HSC2 cells were cultured in tumor sphere-culture medium. All cells were able to grow and form primary spheres within 2 weeks of culture; however, the HSC2/hOCT3/4-shp53-F + hSK + hUL cells could form spheres burly and numerously. Briefly, the number of spheres was significantly higher in the HSC2/hOCT3/4-shp53-F + hSK + hUL cell than HSC2 and HSC2/EGFP cell (Fig. 5a). HSC2/hOCT3/4-shp53-F + hSK + hUL cells also developed comparatively bigger spheres than other cell types (Fig. 5b).

Expression of CSC markers

The expression pattern of CSC markers (CD44 and CD13) in the transfectants and HSC2 cells was analyzed by Western blotting. The protein expression of CD44 was significantly higher in HSC2/hOCT3/4-shp53-F + hSK + hUL than in HSC2/EGFP and HSC2, whereas CD13 expression was higher in all transfectant cells compared to HSC2 parental cell (Fig. 6).

Chemosensitivity and radiosensitivity (in vitro)

CSCs show resistance to anticancer drugs and radiation. Therefore, we evaluated the resistance ability of the transfectants to several chemotherapeutic drugs and radiation. The HSC2/hOCT3/4-shp53-F + hSK + hUL cell showed increased resistance to 5-FU, CDDP, DOC, TFT, zoledronic acid, cetuximab, bortezomib and X-ray radiation than other transfectants and HSC2 cells (Fig. 7).

Tumorigenesis (in vivo)

The tumorigenic capacity of the transfectants was assessed using a mouse model. We injected each transfectant, HSC2 or HSC2/EGFP subcutaneously into BALB/c nude mice and monitored them for 4 weeks. While 1 × 104 cells of HSC2/hOCT3/4-shp53-F + hSK + hUL were enough to generate tumor in mice within 2 weeks, the same number of cells did not produce any HSC2 or HSC2/EGFP tumors. The minimum number of cells required for generating HSC2 or HSC2/EGFP tumor in mice was 1 × 106 (data not shown), which was 100-fold higher than the number of HSC2/hOCT3/4-shp53-F + hSK + hUL required for tumor seeding. In addition, HSC2/hOCT3/4-shp53-F + hSK + hUL showed higher tumorigenic potential (Fig. 8a–c). Histological analysis of these tumors showed squamous cell carcinoma tissues including from highly to poorly differentiated areas, but teratomas were not observed (Fig. 9).

Discussion

In this study, we demonstrated that the introduction of defined reprogramming factors (Oct4, shp53, Sox2, Klf4, l-Myc and Lin28) could generate cells with CSC-like properties from a tongue cancer cell line, HSC2. These artificial CSCs showed resistance to anticancer drugs, molecular target drugs and radiation. Moreover, these artificial CSCs could be maintained in a cost-effective way. D-MEM medium supplemented with 10 % FBS was sufficient to maintain these cells with CSC properties. These artificial CSCs could be handled easily and also managed to produce spheres in sphere-culture medium. Therefore, we may be able to develop a new strategy for generating and maintaining artificial CSCs.
We used HSC2 cell line in our present study, as it has neither invasive nor metastatic potential and showed low expression of the stem cell markers, ie- CD44 and CD13 (Fig. 6). In this study we also examined the expression of CD44v6, ALDH1, CD133 and podoplanin/D2-40, but their expression patterns in transfectant cells were not different compared to that of HSC2 parental cells (data not shown). After introduction of the reprogramming factors, most of the transfectants showed higher invasion and migration ability than HSC2 or HSC2/EGFP. Interestingly, HSC2/hOCT3/4-shp53-F + hSK + hUL cell containing all the reprogramming factors had acquired highest malignant properties in migration assay (Fig. 3), wound healing assay (Fig. 4) and sphere formation assay (Fig. 5) compared to the other transfectants, while demonstrating high expression of CD44. Moreover, our in vivo study showed that tumors developed from HSC2/hOCT3/4-shp53-F + hSK + hUL cells containing all the reprogramming factors (Oct3/4, shp53, Sox2, Klf4, l-Myc or Lin28) exhibited higher tumorigenic potential than parental HSC2 cells. Additionally, these tumors were not teratomas which suggest that our transfectant cells are not iPSCs.
CSCs may be derived from tissue stem cell, however, most of CSCs may be generated from non-tumorigenic differentiated epithelial cells by reprogramming. Nishi et al. reported that the introduction of defined reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) into MCF-10A nontumorigenic mammary epithelial cells, followed by partial differentiation, transforms the bulk of cells into tumorigenic cells with CSC properties [44]. Miyoshi et al. reported that introduction of defined reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) into human gastrointestinal cancer cell lines resulted improved sensitivity of the induced cells to chemotherapeutic agents and differentiation-inducing treatment [45]. Moreover, Oshima et al. reported the generation of CSCs with lineage specificity directly from colon cancer cells by introducing same defined factors (except c-Myc), not via an induced pluripotent stem cell state [46].
There are many attempts to generate iPSCs from malignant tumor cells by the ectopic expression of reprogramming factors. Carette et al. reported to generate the iPS cells derived from human chronic myeloid leukemia cells by using the reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) [47]. Similarly, Utikal et al. reported that they attempted to create iPSCs from melanoma by using the reprogramming factors [48]. However, it seems to achieve only limited success. It may be selected whether the ectopic expression of reprogramming factors in malignant tumor cell lines can induce cells with CSC-like properties or iPSCs during the reprogramming step.
CSCs are known to have high proliferation potency in some cancer types (eg - leukemia), whereas other reports describes CSCs as a slowly dividing cell population (eg- melanoma) and it has also been suggested that as CSCs divide slowly, the CSC population is responsible for tumor resistance to treatment [49]. However, our HSC2/hOCT3/4-shp53-F + hSK + hUL cell had higher proliferative activity though it showed resistance to various chemotherapeutic drugs, molecular-targeted agents, and radiation (Figs. 2 and 7). It is the general idea that, higher proliferation potency may lead to higher intake of anticancer agents inside the cancer cell. One proposed model that explains the origin of CSC’s ability to survive conventional chemotherapeutic regimens describes that, only the CSCs overexpressing ATP-binding cassette (ABC) transporters are able to repopulate the tumor after exposure to the chemotherapeutic agents [50]. The multidrug efflux pump ABCG2 has roles in cytotoxic drug efflux and has been described as one of the reason of the “side population” which helps define adult stem cells of tumors [51]. So, we examined the expression of ABCG2 between the transfectants and HSC2/EGFP or HSC2 to investigate the reason why the HSC2/hOCT3/4-shp53-F + hSK + hUL showed resistance to various chemotherapeutic drugs in spite of its higher proliferation potency. However, we could not detect any differences of ABCG2 expression between the transfectants and HSC2/EGFP or HSC2 (data not shown).
The CSC markers differ in different cancer types. Until now, a number of markers have been discovered, ie - CD44, CD44v4, CD44v6, CD34, CD133, CD166, CD90, CD13, EpCAM, ABCG2, ALDH1 etc. [8, a,b,c]. However, in this present study we reported the expression pattern of only a few. Further experimentation is necessary to understand the expression of other CSC markers in HSC2 and in the transfectants. The reprogramming factors (Oct4, Sox2, Klf4, l-Myc or Lin28) we used in this study might be a few of the key molecules that can trigger the conversion of non-CSCs into CSC in oral tumors. Therefore, it is necessary to identify the other key molecules that are required for the development of CSCs.

Conclusion

In summary, we here describe the possibility of reprogramming non-CSCs by the introduction of defined reprogramming factors and the consequent generation of artificial CSCs. These findings may provide a valuable model system for the study of CSCs, which might help in the development of new therapeutic strategies targeting CSCs in oral tumors.

Abbreviations

5-FU, 5-fluorouracil; ABC transporters, ATP-binding cassette transporters; CDDP, cisplatin; CiRA, center for iPS cell research and application; CSC, cancer stem cell; D-MEM, Dulbecco’s modified Eagle’s medium; DOC, docetaxel; EGF, epidermal growth factor; FBS, fetal bovine serum; FGF-2, fibroblast growth factor-2; HNSCC, head and neck squamous cell carcinoma; IgG, immunoglobulin G; iPSC, induced pluripotent stem cell; MTT, 3-(4, 5-dimethylthiazol- 2-yl)-2, 5- diphenyltetrazolium bromide; NSC, neural stem cell; OD: optical density; OSCC, oral squamous cell carcinoma; RIPA, radioimmunoprecipitation assay; TFT, trifluorothymidine

Acknowledgements

Not applicable.

Funding

This study was supported in part by a Grant-in-Aid from the Japanese Ministry of Education, Science and Culture (Grant no. 24593034).

Availability of data and materials

We state that we agree to share these data. The datasets supporting the conclusions of this article are included within the article.

Authors’ contributions

KH was involved in the study design, data analysis, and writing of the manuscript. TF carried out the experiments, data collection and evaluation, and assisted in manuscript writing. DC helped in data analysis. TM and EI evaluated some data. YK and HO helped in writing the manuscript. HO and YU revised and edited the manuscript. All the authors read and approved the final version of the manuscript.

Competing interests

HO is a paid member of the scientific advisory board of San Bio Co., Ltd. Other authors declare that they have no competing interests.
Not applicable.
All in vivo studies and experiments were approved by the Institutional Animal Care and Use Committee of Yamaguchi University (Ube, Japan).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Mannelli G, Gallo O. Cancer stem cells hypothesis and stem cells in head and neck cancers. Cancer Treat Rev. 2012;38:515–39.CrossRefPubMed Mannelli G, Gallo O. Cancer stem cells hypothesis and stem cells in head and neck cancers. Cancer Treat Rev. 2012;38:515–39.CrossRefPubMed
2.
Zurück zum Zitat Zhang Z, Filho MS, Nör JE. The biology of head and neck cancer stem cells. Oral Oncol. 2012;48:1–9.CrossRefPubMed Zhang Z, Filho MS, Nör JE. The biology of head and neck cancer stem cells. Oral Oncol. 2012;48:1–9.CrossRefPubMed
3.
Zurück zum Zitat Chiou SH, Yu CC, Huang CY, Lin SC, Liu CJ, Tsai TH, Chou SH, Chien CS, Ku HH, Lo JF. Positive correlations of Oct-4 and Nanog in oral cancer stem-like cells and high-grade oral squamous cell carcinoma. Clin Cancer Res. 2008;14:4085–95.CrossRefPubMed Chiou SH, Yu CC, Huang CY, Lin SC, Liu CJ, Tsai TH, Chou SH, Chien CS, Ku HH, Lo JF. Positive correlations of Oct-4 and Nanog in oral cancer stem-like cells and high-grade oral squamous cell carcinoma. Clin Cancer Res. 2008;14:4085–95.CrossRefPubMed
4.
Zurück zum Zitat Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8:755–68.CrossRefPubMed Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8:755–68.CrossRefPubMed
5.
Zurück zum Zitat Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–8.CrossRefPubMedPubMedCentral Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–8.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8.PubMed Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8.PubMed
7.
Zurück zum Zitat Ramdass B, Chowdhari A, Koka P. Cancer-initiating cells as target for prevention of recurring disease etiology: role of these malignant putative progenitor cells in relapse or metastasis of human cervical carcinoma. J Stem Cells. 2013;8:233–51. Review.PubMed Ramdass B, Chowdhari A, Koka P. Cancer-initiating cells as target for prevention of recurring disease etiology: role of these malignant putative progenitor cells in relapse or metastasis of human cervical carcinoma. J Stem Cells. 2013;8:233–51. Review.PubMed
8.
Zurück zum Zitat Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005;5:275–84. Review.CrossRefPubMed Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005;5:275–84. Review.CrossRefPubMed
10.
Zurück zum Zitat Krause M, Yaromina A, Eicheler W, Koch U, Baumann M. Cancer stem cells: targets and potential biomarkers for radiotherapy. Clin Cancer Res. 2011;17:7224–9.CrossRefPubMed Krause M, Yaromina A, Eicheler W, Koch U, Baumann M. Cancer stem cells: targets and potential biomarkers for radiotherapy. Clin Cancer Res. 2011;17:7224–9.CrossRefPubMed
11.
Zurück zum Zitat Nishi M, Akutsu H, Kudoh A, Kimura H, Yamamoto N, Umezawa A, Lee SW, Ryo A. Induced cancer stem-like cells as a model for biological screening and discovery of agents targeting phenotypic traits of cancer stem cell. Oncotarget. 2014;5:8665–80.CrossRefPubMedPubMedCentral Nishi M, Akutsu H, Kudoh A, Kimura H, Yamamoto N, Umezawa A, Lee SW, Ryo A. Induced cancer stem-like cells as a model for biological screening and discovery of agents targeting phenotypic traits of cancer stem cell. Oncotarget. 2014;5:8665–80.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Tirino V, Desiderio V, Paino F, De Rosa A, Papaccio F, La Noce M, Laino L, De Francesco F, Papaccio G. Cancer stem cells in solid tumors: an overview and new approaches for their isolation and characterization. FASEB J. 2013;27:13–24.CrossRefPubMed Tirino V, Desiderio V, Paino F, De Rosa A, Papaccio F, La Noce M, Laino L, De Francesco F, Papaccio G. Cancer stem cells in solid tumors: an overview and new approaches for their isolation and characterization. FASEB J. 2013;27:13–24.CrossRefPubMed
13.
Zurück zum Zitat Moghbeli M, Moghbeli F, Forghanifard MM, Abbaszadegan MR. Cancer stem cell detection and isolation. Med Oncol. 2014;31:69.CrossRefPubMed Moghbeli M, Moghbeli F, Forghanifard MM, Abbaszadegan MR. Cancer stem cell detection and isolation. Med Oncol. 2014;31:69.CrossRefPubMed
14.
Zurück zum Zitat Shimamura M, Nagayama Y, Matsuse M, Yamashita S, Mitsutake N. Analysis of multiple markers for cancer stem-like cells in human thyroid carcinoma cell lines. Endocr J. 2014;61:481–90.CrossRefPubMed Shimamura M, Nagayama Y, Matsuse M, Yamashita S, Mitsutake N. Analysis of multiple markers for cancer stem-like cells in human thyroid carcinoma cell lines. Endocr J. 2014;61:481–90.CrossRefPubMed
15.
Zurück zum Zitat Yan M, Yang X, Wang L, Clark D, Zuo H, Ye D, Chen W, Zhang P. Plasma membrane proteomics of tumor spheres identify CD166 as a novel marker for cancer stem-like cells in head and neck squamous cell carcinoma. Mol Cell Proteomics. 2013;12:3271–84.CrossRefPubMedPubMedCentral Yan M, Yang X, Wang L, Clark D, Zuo H, Ye D, Chen W, Zhang P. Plasma membrane proteomics of tumor spheres identify CD166 as a novel marker for cancer stem-like cells in head and neck squamous cell carcinoma. Mol Cell Proteomics. 2013;12:3271–84.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Athanassiou-Papaefthymiou M, Shkeir O, Kim D, Divi V, Matossian M, Owen JH, Czerwinski MJ, Papagerakis P, McHugh J, Bradford CR, Carey TE, Wolf GT, Prince ME, Papagerakis S. Evaluation of CD44 variant expression in oral, head and neck squamous cell carcinomas using a triple approach and its clinical significance. Int J Immunopathol Pharmacol. 2014;27:337–49.PubMedPubMedCentral Athanassiou-Papaefthymiou M, Shkeir O, Kim D, Divi V, Matossian M, Owen JH, Czerwinski MJ, Papagerakis P, McHugh J, Bradford CR, Carey TE, Wolf GT, Prince ME, Papagerakis S. Evaluation of CD44 variant expression in oral, head and neck squamous cell carcinomas using a triple approach and its clinical significance. Int J Immunopathol Pharmacol. 2014;27:337–49.PubMedPubMedCentral
17.
Zurück zum Zitat Shrivastava S, Steele R, Sowadski M, Crawford SE, Varvares M, Ray RB. Identification of molecular signature of head and neck cancer stem-like cells. Sci Rep. 2015;5:7819.CrossRefPubMedPubMedCentral Shrivastava S, Steele R, Sowadski M, Crawford SE, Varvares M, Ray RB. Identification of molecular signature of head and neck cancer stem-like cells. Sci Rep. 2015;5:7819.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Bhaijee F, Pepper DJ, Pitman KT, Bell D. Cancer stem cells in head and neck squamous cell carcinoma: a review of current knowledge and future applications. Head Neck. 2012;34:894–9.CrossRefPubMed Bhaijee F, Pepper DJ, Pitman KT, Bell D. Cancer stem cells in head and neck squamous cell carcinoma: a review of current knowledge and future applications. Head Neck. 2012;34:894–9.CrossRefPubMed
20.
Zurück zum Zitat Tabor MH, Clay MR, Owen JH, Bradford CR, Carey TE, Wolf GT, Prince ME. Head and neck cancer stem cells: the side population. Laryngoscope. 2011;121:527–33.CrossRefPubMed Tabor MH, Clay MR, Owen JH, Bradford CR, Carey TE, Wolf GT, Prince ME. Head and neck cancer stem cells: the side population. Laryngoscope. 2011;121:527–33.CrossRefPubMed
21.
Zurück zum Zitat Richard V, Nair MG, Santhosh Kumar TR, Pillai MR. Side population cells as prototype of chemoresistant, tumor-initiating cells. Biomed Res Int. 2013;2013:517237.CrossRefPubMedPubMedCentral Richard V, Nair MG, Santhosh Kumar TR, Pillai MR. Side population cells as prototype of chemoresistant, tumor-initiating cells. Biomed Res Int. 2013;2013:517237.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat O'Brien CA, Kreso A, Jamieson CH. Cancer stem cells and self-renewal. Clin Cancer Res. 2010;16:3113–20. Review.CrossRefPubMed O'Brien CA, Kreso A, Jamieson CH. Cancer stem cells and self-renewal. Clin Cancer Res. 2010;16:3113–20. Review.CrossRefPubMed
23.
Zurück zum Zitat Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.CrossRefPubMed Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.CrossRefPubMed
25.
Zurück zum Zitat Marjanovic ND, Weinberg RA, Chaffer CL. Cell plasticity and heterogeneity in cancer. Clin Chem. 2013;59:168–79.CrossRefPubMed Marjanovic ND, Weinberg RA, Chaffer CL. Cell plasticity and heterogeneity in cancer. Clin Chem. 2013;59:168–79.CrossRefPubMed
26.
Zurück zum Zitat Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol. 2013, Chapter 14: Unit 14.25. doi: 10.1002/0471141755.ph1425s61. Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol. 2013, Chapter 14: Unit 14.25. doi: 10.​1002/​0471141755.​ph1425s61.
27.
Zurück zum Zitat Tsai LL, Hu FW, Lee SS, Yu CH, Yu CC, Chang YC. Oct4 mediates tumor initiating properties in oral squamous cell carcinomas through the regulation of epithelial-mesenchymal transition. PLoS One. 2014;9, e87207.CrossRefPubMedPubMedCentral Tsai LL, Hu FW, Lee SS, Yu CH, Yu CC, Chang YC. Oct4 mediates tumor initiating properties in oral squamous cell carcinomas through the regulation of epithelial-mesenchymal transition. PLoS One. 2014;9, e87207.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Qiao B, He B, Cai J, Yang W. The expression profile of Oct4 and Sox2 in the carcinogenesis of oral mucosa. Int J Clin Exp Pathol. 2013;7:28–37.PubMedPubMedCentral Qiao B, He B, Cai J, Yang W. The expression profile of Oct4 and Sox2 in the carcinogenesis of oral mucosa. Int J Clin Exp Pathol. 2013;7:28–37.PubMedPubMedCentral
29.
Zurück zum Zitat Wang J, Wang H, Li Z, Wu Q, Lathia JD, McLendon RE, Hjelmeland AB, Rich JN. c-Myc is required for maintenance of glioma cancer stem cells. PLoS One. 2008;3, e3769.CrossRefPubMedPubMedCentral Wang J, Wang H, Li Z, Wu Q, Lathia JD, McLendon RE, Hjelmeland AB, Rich JN. c-Myc is required for maintenance of glioma cancer stem cells. PLoS One. 2008;3, e3769.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Yu F, Li J, Chen H, Fu J, Ray S, Huang S, Zheng H, Ai W. Kruppel-like factor 4 (KLF4) is required for maintenance of breast cancer stem cells and for cell migration and invasion. Oncogene. 2011;30:2161–72.CrossRefPubMedPubMedCentral Yu F, Li J, Chen H, Fu J, Ray S, Huang S, Zheng H, Ai W. Kruppel-like factor 4 (KLF4) is required for maintenance of breast cancer stem cells and for cell migration and invasion. Oncogene. 2011;30:2161–72.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.CrossRefPubMed Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.CrossRefPubMed
32.
Zurück zum Zitat Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.CrossRefPubMed Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.CrossRefPubMed
33.
Zurück zum Zitat Okita K, Yamakawa T, Matsumura Y, Sato Y, Amano N, Watanabe A, Goshima N, Yamanaka S. An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells. 2013;31:458–66.CrossRefPubMed Okita K, Yamakawa T, Matsumura Y, Sato Y, Amano N, Watanabe A, Goshima N, Yamanaka S. An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells. 2013;31:458–66.CrossRefPubMed
34.
Zurück zum Zitat Okita K, Matsumura Y, Sato Y, Okada A, Morizane A, Okamoto S, Hong H, Nakagawa M, Tanabe K, Tezuka K, Shibata T, Kunisada T, Takahashi M, Takahashi J, Saji H, Yamanaka S. A more efficient method to generate integration-free human iPS cells. Nat Methods. 2011;8:409–12.CrossRefPubMed Okita K, Matsumura Y, Sato Y, Okada A, Morizane A, Okamoto S, Hong H, Nakagawa M, Tanabe K, Tezuka K, Shibata T, Kunisada T, Takahashi M, Takahashi J, Saji H, Yamanaka S. A more efficient method to generate integration-free human iPS cells. Nat Methods. 2011;8:409–12.CrossRefPubMed
35.
Zurück zum Zitat Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, Weinberg, R.A. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008;40:499–507.CrossRefPubMedPubMedCentral Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, Weinberg, R.A. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008;40:499–507.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Okano H, Nakamura M, Yoshida K, Okada Y, Tsuji O, Nori S, Ikeda E, Yamanaka S, Miura K. Step toward safe cell therapy using induced pluripotent stem cell. Circ Res. 2013;112:523–33. Review.CrossRefPubMed Okano H, Nakamura M, Yoshida K, Okada Y, Tsuji O, Nori S, Ikeda E, Yamanaka S, Miura K. Step toward safe cell therapy using induced pluripotent stem cell. Circ Res. 2013;112:523–33. Review.CrossRefPubMed
37.
Zurück zum Zitat Ohnishi K, Semi K, Yamamoto T, Shimizu M, Tanaka A, Mitsunaga K, Okita K, Osafune K, Arioka Y, Maeda T, Soejima H, Moriwaki H, Yamanaka S, Woltjen K, Yamada Y. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell. 2014;156:663–77.CrossRefPubMed Ohnishi K, Semi K, Yamamoto T, Shimizu M, Tanaka A, Mitsunaga K, Okita K, Osafune K, Arioka Y, Maeda T, Soejima H, Moriwaki H, Yamanaka S, Woltjen K, Yamada Y. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell. 2014;156:663–77.CrossRefPubMed
38.
Zurück zum Zitat Matsui T, Takano M, Yoshida K, Ono S, Fujisaki C, Matsuzaki Y, Toyama Y, Nakamura M, Okano H, Akamatsu W. Neural stem cells directly differentiated from partially reprogrammed fibroblasts rapidly acquire gliogenic competency. Stem Cells. 2012;30:1109–19.CrossRefPubMed Matsui T, Takano M, Yoshida K, Ono S, Fujisaki C, Matsuzaki Y, Toyama Y, Nakamura M, Okano H, Akamatsu W. Neural stem cells directly differentiated from partially reprogrammed fibroblasts rapidly acquire gliogenic competency. Stem Cells. 2012;30:1109–19.CrossRefPubMed
39.
Zurück zum Zitat Kim J, Efe JA, Zhu S, Talantova M, Yuan X, Wang S, Lipton SA, Zhang K, Ding S. Direct reprogramming of mouse fibroblasts to neural progenitors. Proc Natl Acad Sci U S A. 2011;108:7838–43.CrossRefPubMedPubMedCentral Kim J, Efe JA, Zhu S, Talantova M, Yuan X, Wang S, Lipton SA, Zhang K, Ding S. Direct reprogramming of mouse fibroblasts to neural progenitors. Proc Natl Acad Sci U S A. 2011;108:7838–43.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Wang T, Choi E, Monaco MC, Major EO, Medynets M, Nath A. Direct induction of human neural stem cells from peripheral blood hematopoietic progenitor cells. J Vis Exp. 2015;95:52298.PubMed Wang T, Choi E, Monaco MC, Major EO, Medynets M, Nath A. Direct induction of human neural stem cells from peripheral blood hematopoietic progenitor cells. J Vis Exp. 2015;95:52298.PubMed
41.
Zurück zum Zitat Szabo E, Rampalli S, Risueño RM, Schnerch A, Mitchell R, Fiebig-Comyn A, Levadoux-Martin M, Bhatia M. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature. 2010;468:521–6.CrossRefPubMed Szabo E, Rampalli S, Risueño RM, Schnerch A, Mitchell R, Fiebig-Comyn A, Levadoux-Martin M, Bhatia M. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature. 2010;468:521–6.CrossRefPubMed
43.
Zurück zum Zitat Muto J, Imai T, Ogawa D, Nishimoto Y, Okada Y, Mabuchi Y, Kawase T, Iwanami A, Mischel PS, Saya H, Yoshida K, Matsuzaki Y, Okano H. RNA-binding protein Musashi1 modulates glioma cell growth through the post-transcriptional regulation of Notch and PI3 kinase/Akt signaling pathways. PLoS One. 2012;7, e33431.CrossRefPubMedPubMedCentral Muto J, Imai T, Ogawa D, Nishimoto Y, Okada Y, Mabuchi Y, Kawase T, Iwanami A, Mischel PS, Saya H, Yoshida K, Matsuzaki Y, Okano H. RNA-binding protein Musashi1 modulates glioma cell growth through the post-transcriptional regulation of Notch and PI3 kinase/Akt signaling pathways. PLoS One. 2012;7, e33431.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Nishi M, Sakai Y, Akutsu H, Nagashima Y, Quinn G, Masui S, Kimura H, Perrem K, Umezawa A, Yamamoto N, Lee SW, Ryo A. Induction of cells with cancer stem cell properties from nontumorigenic human mammary epithelial cells by defined reprogramming factors. Oncogene. 2014;33:643–52.CrossRefPubMed Nishi M, Sakai Y, Akutsu H, Nagashima Y, Quinn G, Masui S, Kimura H, Perrem K, Umezawa A, Yamamoto N, Lee SW, Ryo A. Induction of cells with cancer stem cell properties from nontumorigenic human mammary epithelial cells by defined reprogramming factors. Oncogene. 2014;33:643–52.CrossRefPubMed
45.
Zurück zum Zitat Miyoshi N, Ishii H, Nagai K, Hoshino H, Mimori K, Tanaka F, Nagano H, Sekimoto M, Doki Y, Mori M. Defined factors induced reprogramming of gastrointestinal cancer cells. Proc Natl Acad Sci U S A. 2010;107:40–5.CrossRefPubMed Miyoshi N, Ishii H, Nagai K, Hoshino H, Mimori K, Tanaka F, Nagano H, Sekimoto M, Doki Y, Mori M. Defined factors induced reprogramming of gastrointestinal cancer cells. Proc Natl Acad Sci U S A. 2010;107:40–5.CrossRefPubMed
46.
Zurück zum Zitat Oshima N, Yamada Y, Nagayama S, Kawada K, Hasegawa S, Okabe H, Sakai Y, Aoi T. Induction of cancer stem cell properties in colon cancer cells by defined factors. PLoS One. 2014;9, e101735.CrossRefPubMedPubMedCentral Oshima N, Yamada Y, Nagayama S, Kawada K, Hasegawa S, Okabe H, Sakai Y, Aoi T. Induction of cancer stem cell properties in colon cancer cells by defined factors. PLoS One. 2014;9, e101735.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Carette JE, Pruszak J, Varadarajan M, Blomen VA, Gokhale S, Camargo FD, Wernig M, Jaenisch R, Brummelkamp TR. Generation of iPSCs from cultured human malignant cells. Blood. 2010;115:4039–42.CrossRefPubMedPubMedCentral Carette JE, Pruszak J, Varadarajan M, Blomen VA, Gokhale S, Camargo FD, Wernig M, Jaenisch R, Brummelkamp TR. Generation of iPSCs from cultured human malignant cells. Blood. 2010;115:4039–42.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Utikal J, Maherali N, Kulalert W, Hochedlinger K. Sox2 is dispensable for the reprogramming of melanocytes and melanoma cells into induced pluripotent stem cells. J Cell Sci. 2009;122:3502–10.CrossRefPubMedPubMedCentral Utikal J, Maherali N, Kulalert W, Hochedlinger K. Sox2 is dispensable for the reprogramming of melanocytes and melanoma cells into induced pluripotent stem cells. J Cell Sci. 2009;122:3502–10.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Ciurea ME, Georgescu AM, Purcaru SO, Artene SA, Emami GH, Boldeanu MV, Tache DE, Dricu A. Cancer stem cells: biological functions and therapeutically targeting. Int J Mol Sci. 2014;15:8169–85.CrossRefPubMedPubMedCentral Ciurea ME, Georgescu AM, Purcaru SO, Artene SA, Emami GH, Boldeanu MV, Tache DE, Dricu A. Cancer stem cells: biological functions and therapeutically targeting. Int J Mol Sci. 2014;15:8169–85.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Alisi A, Cho WC, Locatelli F, Fruci D. Multidrug resistance and cancer stem cells in neuroblastoma and hepatoblastoma. Int J Mol Sci. 2013;14:24706–25.CrossRefPubMedPubMedCentral Alisi A, Cho WC, Locatelli F, Fruci D. Multidrug resistance and cancer stem cells in neuroblastoma and hepatoblastoma. Int J Mol Sci. 2013;14:24706–25.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Padmanabhan R, Chen KG, Gillet JP, Handley M, Mallon BS, Hamilton RS, Park K, Varma S, Mehaffey MG, Robey PG, McKay RD, Gottesman MM. Regulation and expression of the ATP-binding cassette transporter ABCG2 in human embryonic stem cells. Stem Cells. 2012;30:2175–87.CrossRefPubMedPubMedCentral Padmanabhan R, Chen KG, Gillet JP, Handley M, Mallon BS, Hamilton RS, Park K, Varma S, Mehaffey MG, Robey PG, McKay RD, Gottesman MM. Regulation and expression of the ATP-binding cassette transporter ABCG2 in human embryonic stem cells. Stem Cells. 2012;30:2175–87.CrossRefPubMedPubMedCentral
Metadaten
Titel
Induction of artificial cancer stem cells from tongue cancer cells by defined reprogramming factors
verfasst von
Koji Harada
Tarannum Ferdous
Dan Cui
Yasuhiro Kuramitsu
Takuya Matsumoto
Eiji Ikeda
Hideyuki Okano
Yoshiya Ueyama
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2016
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2416-9

Weitere Artikel der Ausgabe 1/2016

BMC Cancer 1/2016 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.