1α,25‑Dihydroxyvitamin D3 restrains stem cell‑like properties of ovarian cancer cells by enhancing vitamin D receptor and suppressing CD44

  • Authors:
    • Mintao Ji
    • Lizhi Liu
    • Yongfeng Hou
    • Bingyan Li
  • View Affiliations

  • Published online on: April 15, 2019     https://doi.org/10.3892/or.2019.7116
  • Pages: 3393-3403
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Scientific evidence linking vitamin D with various cancer types is growing, but the effects of vitamin D on ovarian cancer stem cell‑like cells (CSCs) are largely unknown. The present study aimed to examine whether vitamin D was able to restrain the stemness of ovarian cancer. A side population (SP) from malignant ovarian surface epithelial cells was identified as CSCs, in vitro and in vivo. Furthermore, 1α,25‑dihydroxyvitamin D3 [1α,25(OH)2D3] treatment inhibited the self‑renewal capacity of SP cells by decreasing the sphere formation rate and by suppressing the mRNA expression levels of cluster of differentiation CD44, NANOG, OCT4, SOX2, Krüppel‑like factor 4 and adenosine triphosphate binding cassette subfamily G member 2. Additionally, 1α,25(OH)2D3 treatment decreased the expression of Cyclin D1, whereas it increased the expression of β‑catenin and vitamin D receptor (VDR). Notably, immunofluorescence staining verified that 1α,25(OH)2D3 promoted the expression of β‑catenin in the cytoplasm. Furthermore, vitamin D3 delayed the onset of tumor formation derived from injection of ovarian CSCs to nude mice, by reducing CD44 and enhancing β‑catenin expressions in vivo. In conclusion, 1α,25(OH)2D3 restrains the stem cell‑like properties of ovarian cancer cells by enhancing the expression of VDR, by promoting the expression of β‑catenin in the cytoplasm, and by suppressing the expression of CD44. These findings provide a novel insight into the functions of vitamin D in diminishing the stemness of cancer CSCs.

Introduction

Ovarian cancer is the fourth most frequent gynecologic malignancy and the leading cause of tumor-associated mortality in the USA (1). Epithelial ovarian cancer, which accounts for ~90% of ovarian cancers, is generally diagnosed at an advanced stage (2). Furthermore, the prognosis and five-year survival have not improved significantly owing to the high recurrence rate (3). Therefore, it is urgent to identify the underlying mechanisms and to develop alternative therapeutic strategies for this type of cancer.

The ovarian surface epithelium (OSE) is the recognized source of epithelial ovarian cancer (2,4). Several characteristics of epithelial ovarian carcinomas imply it is a cancer stem cell-driven disease. First, the stem properties of OSE have been recognized previously (5), and the cancer-prone stem cell niche was also identified at the helium area of ovary (6). Second, epithelial ovarian cancer can differentiate into several subtypes that recapitulate the histology of other normal gynecologic tissues (2). Third, the high recurrence rate following the initial successful treatment may derive from a small number of cells within the cancer population, which are: i) Capable of repopulating the entire tumor and ii) exhibit cytoprotective mechanisms on somatic stem cells (610). Furthermore, the presence of cancer stem cell-like cells (CSCs) in patients is reported to be associated with poor survival and chemoresistance (11,12). Therefore, targeting CSCs may be a potential therapeutic approach to epithelial ovarian cancer.

Emerging evidence suggests that vitamin D deficiency is strongly associated with the risk of various human cancers, including colorectal, breast, prostate and ovarian cancer (13). 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3], the active metabolite of vitamin D3, functions by binding to vitamin D receptor (VDR). 1α,25(OH)2D3, or analogues of 1α,25(OH)2D3, may serve as anticancer agents owing to their ability to suppress proliferation, invasion, metastasis and angiogenesis, and to induce apoptosis (1417). Furthermore, 1α,25(OH)2D3 is reported to inhibit the proliferation of prostate cancer stem cells by inducing cell cycle arrest and senescence (18). Furthermore, results from in vitro and xenograft studies indicated that the vitamin D analogue (BXL0124) may decrease mammosphere growth by reducing cluster of differentiation CD44 expression levels (14,19,20). There is preliminary evidence that 1α,25(OH)2D3 could be used to cure and inhibit prostate and breast CSCs (21). Our previous study demonstrated that 1α,25(OH)2D3 inhibited the migration of human ovarian cancer cells by increasing the expression of VDR and suppressing epithelial-mesenchymal transition (17). However, the effects of vitamin D3 on ovarian CSCs remain largely unknown. Side population (SP) cells have been presented as functional markers of CSCs (2226). The present study investigated whether 1α,25(OH)2D3 was able to inhibit the stem cell-like phenotype of SP cells isolated from mouse OSE (MOSE) cells, and determined its possible underlying mechanisms. The present findings indicated that 1α,25(OH)2D3 suppressed the properties of ovarian CSCs by enhancing VDR expression and reducing CD44 level, which may provide the novel strategy for treating epithelial ovarian cancer.

Materials and methods

Cell culture and reagents

MOSE cells were isolated as described by Roby et al (27). Briefly, the 4 mice are 6–8 week old and ~20 g. All mice were housed with full of food and water under controlled conditions of temperature at 21±2°C, relative humidity at 55±5% and a 12:12 h light-dark cycle. Ovaries from female breeder mice (BALB/c) were resected and, following removal of the residual remnants of the oviducts, were incubated in 1.5 ml tubes for 30 min with trypsin in 5% CO2 at 37°C. Following incubation, tubes were removed from the incubator and inverted 10 times. The ovaries were transferred to a clean 1.5 ml tube for the second digestion with trypsin, as above. Cells were collected, pelleted by 1,000 × g/min for 5 min at 4°C, resuspended in DMEM/F12 medium containing 20 ng/ml mEGF, 20 ng/ml mouse basic fibroblast growth factor, 2 µg/ml insulin and 4 µg/ml heparin sodium and seeded onto 35 mm dishes. On the 2nd day, the culture dishes were not removed, because moving them is conducive to cell growth and therefore the stability of the microenvironment around the cells can be maintained. On the 3rd day, growth medium was changed according to the cell growth. MOSE cells were continuously passaged in vitro. MOSE cells maintained proliferation and underwent spontaneous neoplastic transformation when subcultured for >80 passages in vitro (see Supplementary data). The spontaneous malignant transformation of MOSE cells in late stage (M-L cells) were maintained in Dulbecco's modified Eagle's medium/F-12 medium (DMEM/F12) containing 10% fetal bovine serum (FBS), 1% penicillin, 1% streptomycin, 10 ng/ml mouse epithelial growth factor (mEGF) and 1% insulin-transferrin-selenium in an atmosphere of 95% humidity and 5% CO2 at 37°C. The MOSE cells have been tested for mycoplasma and human cell lines contamination by STR profiling and were demonstrated to be clean (see Figs. S1 and S2; Table SI). The reagents for cell culture were purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Vitamin D3 (40,000 IU/ml) and 1α,25(OH)2D3 were purchased from Shanghai General Pharmaceutical Company, Ltd. (www.cpshgp.com/english/; Shanghai, China) and Sigma-Aldrich (Merck KGaA, Darmstadt, Germany), respectively. In the present study, cells were treated by 10 nM 1α,25(OH)2D3 in vitro, and mice were was administrated by vitamin D3. In vivo, vitamin D3 is successively hydroxylated by the 25-hydroxylase and 1α-hydroxylase, and forms 1α,25(OH)2D3, the active form of vitamin D3.

Isolation of SP cells

M-L cells (1×106 cells/ml) were re-suspended in DMEM/F12 medium containing 2% FBS and 4% penicillin-streptomycin, and stained with 10 µg/ml Hoechst 33342 (Sigma-Aldrich; Merck KGaA) in the presence or absence of 50 µg/ml verapamil (Sigma-Aldrich; Merck KGaA) at 37°C for 90 min with intermittent shaking every 10 min. Following incubation, the cells were centrifuged by 1,000 × g/min for 5 min at 4°C and washed with cold phosphate-buffered saline (PBS). The SP and non-side population (NSP) cells were isolated and collected by MoFloAstrios EQ fluorescence activated cell sorting (Summit 6.2; FACS, www.beckmancoulter.cn/ls-discovery/flow/researchflow/moflo-astrios.html; Beckman Coulter, Inc., Brea, CA, USA). The Hoechst dye was excited with a UV laser at 346 nm and its fluorescence emissions were measured with 630/22 (Hoechst 33342 Red) and 424/44 filters (Hoechst Blue). For purification, isolated SP cells were cultured in serum-free DMEM/F12 medium containing 20 ng/ml mEGF, 20 ng/ml mouse basic fibroblast growth factor, 1:50 B27, 2 µg/ml insulin, 4 µg/ml heparin sodium and 6 mg/ml glucose for 10 days and were subsequently re-suspended in StemPro Accutase Cell Dissociation Reagent at 37°C for 10 min. The NSP cells were cultured in DMEM/F12 medium containing 20 ng/ml mEGF, 20 ng/ml mouse basic fibroblast growth factor, 2 µg/ml insulin and 4 µg/ml heparin sodium. The reagents in serum-free medium were purchased from Thermo Fisher Scientific, Inc. Subsequently, the cells were centrifuged by 1,000 × g/min for 5 min at 4°C and maintained in the fresh serum-free medium until further use.

Detection of CD44 and CD117

The SP and NSP cells were washed, re-suspended and separately incubated with fluorescence-conjugated antibodies against CD44 (1:1,000; cat. no. 555478), CD117 (1:1,000; cat. no. 555714) and IgG (1:1,000; cat. nos. 555749 and 555742; BD Biosciences, San Jose, CA, USA) in 4°C for 10 min. The appropriate concentrations of each antibody were recommended by the manufacturer. The cells were washed and analyzed by Cytomics™ FC 500 from Beckman Coulter, Inc. In brief, unstained cells, CD44+, CD117+ and double-stained control cells were used to mark the four quadrants in a dot-plot for unstained, CD44+, CD117+ and double-positive populations. The software used for the analysis is CXP Analysis, which is the part of FC500 flow cytometer (Beckman Coulter, Inc.).

Sphere-formation assay

The SP cells were re-suspended in StemPro Accutase Cell Dissociation Reagent at 37°C for 10 min, terminated by the serum-free DMEM/F12 medium. Subsequently, 1,000 cells/well were plated in the 96-well plates and cultured in serum-free medium for 10 days. The spheres with a minimum size of 50 µm were counted under a brightfield microscope (CKX41F; Olympus Corporation, Tokyo, Japan) equipped with a digital camera. The sphere-formation rate was expressed as the following formula: (Number of spheres formed/number of plated cells) × 100.

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

Total RNA extractions from 1×106 cells SP or NSP cells were performed using TRIzol® reagent (Life Technologies; Thermo Fisher Scientific, Inc.). For the synthesis of first-strand cDNA, total RNA was reverse-transcribed by the Transcriptor First Strand cDNA Synthesis Kit (Roche Diagnostics, Basel, Switzerland). Primer sequences are available in Table I. qPCR reactions were conducted with the SYBR-Green I Nucleic AcideGel stain (Roche Diagnostics) according to manufacturer's protocol. Thermocycling parameters were: 95°C for 10 min, followed by 40 cycles of 95°C for 10 sec, 60°C for 20 sec and 72°C for 30 sec. Relative expression levels were calculated using the 2−ΔΔCq method by Livak and Scmittgen (28). GAPDH was used as normalization control.

Table I.

Primer sequences for reverse transcription-quantitative polymerase chain reaction.

Table I.

Primer sequences for reverse transcription-quantitative polymerase chain reaction.

GenePrimer sequence (5′→3′)
GAPDHF: TTGATGGCAACAATCTCCAC
R: CGTCCCGTAGACAAAATGGT
CD44F: AGCGGCAGGTTACATTCAAA
R: CAAGTTTTGGTGGCACACAG
NANOGF: AGGGTCTGCTACTGAGATGCTCTG
R: CAACCACTGGTTTTTCTGCCACCG
OCT4F: AGAAGGAGCTAGAACAGTTTGC
R: CGGTTACAGAACCATACTCG
CD133F: TAGAGGGAAGTCATTCGGCT
R: CCCAAGATACCTTCAATGCTG
SOX2F: AAAGCGTTAATTTGGATGGG
R: ACAAGAGAATTGGGAGGGGT
KLF4F: CAGTGGTAAGGTTTCTCGCC
R: GCCACCCACACTTGTGACTA
NOTCH1F: CTGAGGCAAGGATTGGAGTC
R: GAATGGAGGTAGGTGCGAAG
NOTCH2F: TGTGCCGTTGTGGTAGGTAA
R: TGCTGTGGCTCTGGCTGT
ABCG2F: TCGCAGAAGGAGATGTGTTGAG
R: CCAGAATAGCATTAAGGCCAGG
CD117F: CGGTCGACTCCAAGTTCTACAAG
R: GTTGCAGTTTGCCAAGTTGGAGT
β-cateninF: ATGGCTTGGAATGAGACTGC
R: CTCCATCATAGGGTCCATCC
c-MycF: CAACGTCTTGGAACGTCAGA
R: TCGTCTGCTTGAATGGACAG
Cyclin D1F: TGTTCGTGGCCTCTAAGATG
R: ACTCCAGAAGGGCTTCAATC
VDRF: TGACCCCACCTACGCTGACT
R: CCTTGGAGAATAGCTCCCTGTACT

[i] ABCG2, ATP binding cassette subfamily G member; CD, cluster of differentiation; F, forward; KLF, Krüppel-like factor; OCT, octamer-binding protein; R, reverse; SOX, Sex-determining region Y; VDR, vitamin D receptor.

Western blotting

Approximately 106 cells were scraped off with radioimmunoprecipitation buffer (cat. no. P0013; Beyotime Institute of Biotechnology, Haimen, China). Subsequently, the lysates were centrifuged at 7,500 × g for 30 min at 4°C and the supernatants were collected. Protein concentrations were quantified by the Bicinchoninic Acid assay (Beyotime Institute of Biotechnology). Proteins (30 µg/sample) were separated by 10% SDS-PAGE and were transferred to polyvinylidene fluoride membranes. Membranes were blocked with 5% non-fat milk in PBS + 1% Tween-20 and incubated with primary antibodies against VDR (1:500; cat. no. 12550), Cyclin D1 (1:1,000; cat. no. 2978), β-catenin (1:1,000; cat. no. 8480), Sex-determining region Y (SOX2; 1:1,000; cat. no. 23064), NANOG (1:1,000; cat. no. 8822), octamer-binding protein (OCT4; 1:1,000; cat. no. 2840) and GAPDH (1:1,000; cat. no. 5174) at 4°C overnight. Subsequently, the membranes were incubated for 1 h at room temperature with 1:3,000 anti-mouse IgG, HRP-linked antibody (cat. no. 7076) and anti-rabbit IgG, HRP-linked antibody (cat. no. 7074). All antibodies were purchased from Cell Signaling Technology, Inc. (CST; Danvers, MA, USA). Protein bands were visualized using Enhanced Chemiluminescence Detection (Merck KGaA); to quantify the level of protein expression, densitometric analysis was performed according to the manufacture's protocol (Gbox Chemi-XR 5; Syngene, Frederick, MD, USA). The specific/individual protein expressions (GeneSnap image acquisition software, GeneTools image analysis software; Syngene) should be normalized with their respective GAPDH loading control first. Subsequently, when drafting the histograms, the NSP or control groups should be set to ‘1’ and all other expression compared with that.

Immunofluorescence staining

Approximately 1×105 cells were plated onto coverslips until they reached an optimal density of 70–80%. The cells were fixed with 4% paraformaldehyde at 4°C for 20 min, permeabilized with 0.1% Triton X-100 for 15 min at 4°C and blocked with 1% FBS for 1 h at room temperature. Subsequently, the cells were incubated with primary antibody of β-catenin (CST; 1:100; cat. no. 8480) overnight at 4°C. Following this, the cells were labeled with anti-rabbit IgG (H+L), F(ab')2 Fragment (Alexa Fluor® 488-conjugated; CST; 1:1,000, cat. no. 4412) in the dark room for 1.5 h at room temperature and washed with PBS three times. Nuclei were stained with DAPI for 20 min at room temperature. The signals were detected using a TCS SP2 Confocal Laser Scanning Microscope (Leica Microsystems GmbH, Wetzlar, Germany).

X-ray and heavy particle resistance of SP cells

Approximately 1×105 cells were irradiated with 2, 4, 6, 8 or 10 Gy of X-rays using a Pantac HF-320S X-ray generator (Shimadzu Co., Kyoto, Japan) or carbon-ion beams accelerated by the Heavy ion medical accelerator in Chiba in the National Institute of Radiological Sciences (Chiba, Japan). Following irradiation, NSP cells were plated in triplicate in 60-mm dishes for assays of clonogenicity. After culturing for 14 days at 37°C, the colonies were fixed with 75% alcohol and stained with 0.3% methyl violet for 20 min at room temperature. Colonies containing >50 cells were counted as the survivors. Additionally, SP cells in serum-free medium were plated (1,000 cells/well) in 96-well plates for sphere-formation assay. The spheroid formation rates were calculated as the percentage of the spheres >50 µm. At least three parallel samples were scored in six replicates conducted for each aforementioned irradiation condition. Cell survival fractions were obtained from fitting the surviving fraction to a linear-quadratic model expressed by the following formula: SF = exp - (αD + βD2), where SF is the survival fraction, exp means ‘exp-function’, α and β are the constant of the fitted curve by the results, and D is the irradiation dose.

Tumorigenesis

BALB/c nude mice (age, 4–6 weeks, 18–20 g) were purchased from Soochow University Laboratory Animal Center (Suzhou, China). Mice were provided with water and food ad libitum and were housed at five animals per cage. All mice were housed under controlled conditions of temperature at 21±2°C, relative humidity at 55±5% and a 12:12 h light-dark cycle. All surgical procedures and care administered to the animals were approved by the Institutional Animal Care and Use Committee (approval number is ECSU-201800049). In the orthotopic model, 10 µl cell suspension (1×104 NSP or SP cells) mixed with Matrigel (1:1; BD Biosciences; cat. no. 356234) were injected orthotopically into the one ovary (n=2 mice/group). In the subcutaneous model, it has been reported that SP cells has stronger tumorigenicity than NSP cells (22,24). Therefore, different number of SP and NSP cells were injected into mice. One hundred µl cell suspension containing 1×104 SP cells or 1×106 NSP cells mixed with Matrigel were injected subcutaneously into flanks of the mice (n=4 mice/group), respectively. The subcutaneous model was used to evaluate the effect of vitamin D3 on tumorgenicity of SP cells. The mice in the vitamin D3-treatment group were injected with a single dose of vitamin D3 (1,000 IU/week) intramuscularly. The animals were sacrificed at the indicated time intervals (4–12 weeks) when tumor nodules were identified on their body surfaces. The anesthesia of the mice used was 1% pentobarbital sodium (50 mg/kg), prior to the injection of cells to their ovary. The euthanasia/sacrifice of the mice was performed by 3% pentobarbital sodium (150 mg/kg), followed by cervical dislocation to ensure the death of mice. Tumor growth was monitored by measuring 2 perpendicular diameters. Tumor volumes were calculated according to the formula: 0.5 × a × b2, where a and b are the largest and smallest diameter, respectively.

Statistical analysis

Statistical analysis was performed using the paired Student's t-test for the difference between two groups. One-way analysis of variance was used where multiple comparisons were made, followed by Bonferroni's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Categorizing MOSE cells

The capacity of anchorage-independent growth was determined, which in an in vitro hallmark of neoplastic transformation of cells (29). Early (<20 passages) and intermediate (21–80 passages) MOSE cells were unable to form colonies in soft agar. Notably, late-passage (>81) MOSE cells were capable of forming >30 mm colonies. Compared with MOSE cells at earlier stages, late-passage MOSE cells exhibited an increased plating efficiency and growth rate, another proliferative parameter that is often associated with neoplastic change. Furthermore, the tumor formation rates were significantly increased in vivo (27,30). M-L cells formed tumors up to 100%, whereas M-E and M-I cells did not form tumors. Therefore, three sequential stages of transformed MOSE cells were defined as M-E (≤20 passages; early), M-I (21–80 passages; intermediate) and M-L (≥81 passages; late) cells, respectively.

Identification of ovarian cancer stem cells-like cells

Our previous study demonstrated that the acquisition of stemness was closely associated with malignant transformation of MOSE cells (unpublished data). SP cells are an important hallmark for the definition of the stem cell-like characteristics (2226); therefore, whether SP cells may be used to enrich ovarian CSCs was investigated. Malignant MOSE (M-L) cells were separated by FACS into two populations. SP cells actively pump the Hoechst 33342 dye out of the cells, while NSP cannot and therefore this is a way to isolate these two cell populations (2226). The percentage of SP cells in M-L cells reached up to 24.4% (Fig. 1A). After culturing for 3–4 days in serum-free culture medium, NSP cells gradually grew by adherence, and acquired epithelial morphology; however, SP cells formed larger spheres (Fig. 1B). Results from the sphere formation assay revealed that the sphere-forming rates of SP cells were significantly higher compared with those of the NSP cells (Fig. 1C).

To further verify the stem cell-like properties of SP cells, the expressions of CD44+ and CD117+, two well-known ovarian CSCs markers, were assessed by flow cytometry. In SP cells, the number of CD44+ (99.8%) and CD117+ (21.2%) cells were significantly higher compared with NSP cells (Fig. 1D and E). RT-qPCR results demonstrated that the mRNA levels of CD44 and CD133 in SP cells were increased, compared with the NSP cells (Fig. 1F). Notably, the mRNA expression levels of multipotent genes, such as SOX2, Krüppel-like factor (KLF)4, NOTCH1 and NOTCH2 were increased in SP cells, whereas NANOG expression was decreased, compared with NSP cells (Fig. 1G). Similarly, the protein expression levels of SOX2 and OCT4 were also significantly increased in SP compared with NSP, and the protein expression of NANOG was decreased. (Fig. 1H). In addition, the mRNA expression levels of ATP binding cassette subfamily G member (ABCG)2 and c-Myc were also significantly elevated in SP cells (Fig. 1G). Notably, the protein levels of VDR, β-catenin and Cyclin D1 in SP cells were lower compared with those in NSP cells (Fig. 1I).

Radioresistance is also a remarkable phenotype of CSCs (31,32). Thus, the survival fraction of cells irradiated by X-rays and carbon ions was determined using clonogenic and sphere-forming assay. The dose-response curves demonstrated that the survival fraction of SP cells was higher compared with the NSP cells, which indicated that SP cells exhibited resistance to both X-rays and carbon ions (Fig. 1J).

To further determine the tumorigenesis of SP and NSP cells in vivo, subcutaneous (n=4 mice/group) and orthotopic (n=2 mice/group) models of ovarian cancer were established in nude mice injected with SP or NSP. Neither of the two mice in the orthotopic NSP groups formed tumors; of the two mice that were orthotopically implanted with SP cells, one died at post-implantation, the other one grew a tumor with volume of 49.95 mm3 by 36 days post-implantation (data not shown). In the subcutaneous model, different numbers of NSP and SP cells were injected. None of the four mice injected with 1×106 NSP cells formed any tumor by 12 weeks post-injection; however, two of the four mice injected with 1×104 SP cells formed tumors within 30 days post-injection, the volumes of which was 0.018 and 48 mm3 (Fig. 1K). Taken together, these in vitro and in vivo results demonstrated that the SP cells isolated from oncogenic transformation-MOSE cells exhibited properties of CSCs with self-renewal capability, multipotency and radioresistance.

1α,25(OH)2D3 inhibits stem cell-like phenotype of SP cells

To verify whether active vitamin D3 is able to inhibit the stemness of CSCs, SP cells were treated with 10 nM 1α,25(OH)2D3. Notably, a few 1α,25(OH)2D3-treated SP cells gradually grew by adherence following the treatment for three days, compared with untreated SP cells. Furthermore, the number and diameter of spheres formed were reduced in 1α,25(OH)2D3-treated SP cells (Fig. 2A and B). Consistent with this phenotype, the mRNA expression levels of the multipotent genes NANOG, OCT4, SOX2, KLF4 and ABCG2 were downregulated in 1α,25(OH)2D3-treated SP cells, compared with the untreated SP cells (Fig. 2C and D). However, the protein levels of NANOG, OCT4 and SOX2 were not significantly reduced in SP cells treated with 1α,25(OH)2D3 (Fig. 2E). These results indicated that 1α,25(OH)2D3 may be able to suppress the self-renewal capability and expression of multipotent gene expressions including NANOG, OCT4, SOX2, KLF4 in ovarian CSCs, although this needs to be verified.

1α,25(OH)2D3 increases β-catenin expression but decreases Cyclin D1 expression in SP cells

We know that 1α,25(OH)2D3 exhibits its biological effects mainly through binding to VDR (13). It was determined that the protein and mRNA expression levels of VDR were increased in SP cells treated by 1α,25(OH)2D3 (Fig. 3A, D and E). Subsequently, whether 1α,25(OH)2D3 engaged in modulating VDR-mediated genes of ovarian CSCs was determined. The results of RT-qPCR demonstrated that 1α,25(OH)2D3 had the tendency to increase β-catenin but significantly decreased Cyclin D1 mRNA expression levels in SP cells compared with Control cells (Fig. 3B and C). Western blot results demonstrated that 1α,25(OH)2D3 treatment enhanced β-catenin and reduced Cyclin D1 protein expression levels in a dose-dependent manner (Fig. 3F and G). However, immunofluorescence staining indicated that the expression of β-catenin was mainly located in the nuclei of vehicle-treated SP cells, and β-catenin was blocked in cytoplasm following treatment with 1α,25(OH)2D3 (Fig. 3H). Combined with the suppression of stem cell-like properties demonstrated in Fig. 2, these results implicated that 1α,25(OH)2D3 may inhibit the stemness of SP cells through increasing VDR and cytoplasmic β-catenin and decreasing Cyclin D1 expression levels.

Vitamin D3 inhibits the tumorigenic phenotype of SP cells in vivo

To further determine whether vitamin D3 had suppressive effects on tumorigenesis of SP cells, ~1×104 SP cells were subcutaneously inoculated into nude mice. Tumor formation was monitored by palpation and direct body dissection. In two of the four vehicle-treated Control mice tumor nodules were palpable as early as 30 days post-inoculation. By contrast, two of the four mice treated with vitamin D3 exhibited signs of tumor growth at 45 days post-inoculation (Fig. 4A). To determine whether vitamin D3 also modulates the expression of stemness-associated proteins in vivo, total protein was extracted from tumor tissues and analyzed by western blotting (Fig. 4B). The results indicated that vitamin D3 treatment led to increased VDR and decreased Cyclin D1 protein expression levels, but the changes were not significant (Fig. 4C and D). Furthermore, vitamin D3 decreased the expression levels of CD44, whereas it increased the expression levels of β-catenin in vivo compared with Control mice (Fig. 4E and F). These results demonstrated that vitamin D3 may have delayed the onset of tumor formation derived from injection of ovarian CSCs by reducing CD44 and increasing β-catenin expression, but this needs to be validated further.

Discussion

Accumulating evidence demonstrates that CSCs serve crucial roles in the development of chemoresistance, tumor relapse and metastasis in patients with ovarian cancer (33,34). The present study reports that vitamin D3 not only inhibits self-renewal capability and multipotent gene expressions of ovarian CSCs in vitro, but it may also delay the onset of tumor formation. Furthermore, 1α,25(OH)2D3 treatment increased both the mRNA and protein expression levels of VDR and β-catenin, whereas it decreased the expression of Cyclin D1 and certain stemness-associated genes, including CD44, NANOG, OCT4, SOX2, KLF4 and ABCG2 in ovarian CSCs in vitro. Notably, the reduced expression of CD44 was demonstrated in vitro and in vivo.

Owing to repeating disruption and repairing with ovulation-associated remodeling, OSE with stem-like properties have been identified (5), and the junction area of hilum and oviduct contains cancer-prone stem cell niche (6). Ovarian CSCs with multipotent and self-renewal capability are usually purified using the surface markers CD44, CD117 and CD133, or functional marker such as isolation of SP and ALDH+ cells (22,35,36). It is reported that SP cells isolated from SKOV-3 cells displayed stem-like phenotype (25,37,38). In the present study, SP cells were isolated from malignant transformation MOSE cells, and demonstrated that both mRNA and protein expression levels of SOX2 and Oct4 were increased in SP cells. Additionally, they exhibited self-renewal capability and radioresistance in vitro, and an increased tumor growth in vivo. Notably, both mRNA and protein expression levels of VDR were reduced in SP cells. In general, high VDR expression is associated with reduced mortality and improved prognosis in breast and prostate tumors (39,40). 1α,25(OH)2D3 activates and represses its target genes, such as RXRA, SMAD3, CCND3, by combining with VDR. Previous studies showed that 1α,25(OH)2D3 inhibit proliferation and angiogenesis, and induce apoptosis of cancer cells in human cancers including ovarian cancer (4143). However, it is not clear whether 1α,25(OH)2D3 could reduce stemness of CSCs or promote differentiation through binding to VDR.CD44, a marker for stem cells of several cancers, serves an important role in ovarian CSCs (44,45). Additionally, a number of studies have reported that 1α,25(OH)2D3 has potent effects on prostate and breast cancer stem cells, and demonstrated that 1α,25(OH)2D3 and its analogue inhibited the proliferation of prostate and breast stem cells by inducing senescence and by decreasing CD44 expression levels (14,1820). The present study results also indicated that 1α,25(OH)2D3 suppressed the self-renewal capacity of ovarian CSCs by reducing CD44 expression levels, as well as the expressions of multipotent genes, such as Oct4, SOX2, NANOG, KLF4 and ABCG2 in vitro. Notably, 1α,25(OH)2D3 treatment decreased the mRNA expression levels of Oct4, SOX2 and NANOG, but not their protein levels. Previous studies have indicated that Oct4, SOX2 and NANOG often function in combination with each other to be involved in self-renewal and proliferation (46,47). CD44v3, a CD44 variant isoform, was reported to interact with Oct4-SOX2-NANOG (48). At the transcriptional level, Oct4, SOX2 and NANOG form a positive autoregulatory loop which is important for the maintenance of the undifferentiated state. At the post-translational level, non-coding RNAs are emerging as a key player in the control of cell proliferation and cell fate determination during differentiation (48). For example, microRNA-302 is controlled by a promoter containing Oct4-SOX2-NANOG-binding sites in human head and neck squamous cell carcinoma (48). It is worth exploring whether 1α,25(OH)2D3-decreased CD44 may weaken the interaction with Oct4-SOX2-NANOG, which may result in no changes at the protein level, even though the mRNA expression levels of Oct4, SOX2 and NANOG were significantly decreased. In addition, 1α,25(OH)2D3 increased the β-catenin expression levels and decreased Cyclin D1 expression.

β-Catenin is a dual function protein, involved in both stemness and contribution to metastasis. On the one hand, β-catenin, as an important molecule of the Wnt signaling pathway, sustains stem cell renewal ability by maintaining multipotency in certain cell types (49). On the other hand, β-catenin, as a proto-oncogene, drives metastasis formation by translocation to the nucleus. Alterations in the localization and expression levels of β-catenin are associated with many cancers, including hepatocellular, colorectal, lung, breast, ovarian and endometrial cancer (5055). Furthermore, the level of β-catenin can be modulated by VDR in colon cancer cells (56). In addition, 1α,25(OH)2D3 suppresses the expression of Cyclin D1 in epidermal carcinoma (57). These studies indicated that β-catenin and Cyclin D1 are both downstream target genes of 1α,25(OH)2D3. In the present study, 1α,25(OH)2D3 increased the expression of VDR and decreased the expression of Cyclin D1 at mRNA and protein levels. Notably, 1α,25(OH)2D3 significantly increased the expression of β-catenin, which was inconsistent with previous studies. However, immunofluorescence staining verified that 1α,25(OH)2D3 only increased the expression of β-catenin in cytoplasm, consistent with Pálmer's study (58), which may result in the decrease of Cyclin D1. These results demonstrated that 1α,25(OH)2D3 inhibited the stemness of CSCs through blocking the localization of β-catenin in cytoplasm.

However, there are several limitations in the present study. The number of mice used in the present study was not sufficient, which may have reduced the statistical power. Since the previous studies showed that SP cells exhibited more tumorigenicity than NSP cells in subcutaneous model in vivo (22,24), in the present study, 1×106 NSP cells were subcutaneously injected into mice. Two of the four mice injected with 1×104 SP cells formed tumors within 30 days post-injection, but none of the four mice injected with 1×106 NSP cells formed any tumor by 12 weeks. Additionally, whether SP cells would display more tumorgenicity than NSP cells in the orthotopic model need to be investigated. Unfortunately, the successful rate of the orthotopic operation was low and could barely get enough orthotopic mice for further study. In addition, the direct or indirect interaction between VDR and β-catenin, which is also very interesting and may be a novel underlying mechanism, was not investigated.

In conclusion, the present study demonstrated that 1α,25(OH)2D3 restrained stem cell-like properties of ovarian cancer cells by enhancing VDR and had the tendency to promote cytoplasmic β-catenin, and reducing CD44 expression levels. These results may provide a novel strategy for vitamin D3 in diminishing the stemness of CSCs. Future studies utilizing human ovarian cancer tissues to examine the role of vitamin D3 in stem cell-like cell self-renewal may extend our understanding of ovarian cancer biology, which may lead to chemotherapeutic agents that may suppress stemness and improve the clinical outcome for patients with epithelial ovarian cancer.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Miss Hemei Zhang (Wenzhou Center for Disease Control and Prevention), Dr Fei Jiang (Soochow University), Dr Jianmei Wan (Soochow University), Miss Ping Wang (Fuzhou General Hospital), Professor Guangming Zhou (Soochow University) and Professor Zengli Zhang (Soochow University) for contributing materials and writing the manuscript.

Funding

The present study was supported by The National Natural Scientific Funding of China (grant nos. 81673151, 81372979, 8137298 and 111335011), in part, by The State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions.

Availability of data and materials

The data sets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

BL and LL conceived and designed the experiments. LL and MJ performed the experiments. LL, MJ and YH analyzed the data. YH contributed to the material and analysis tools. MJ and BL drafted and revised the manuscript. All authors have read and approved the final manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

All surgical procedures and care administered to the animals were approved by the Institutional Animal Care and Use Committee.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Bast RC Jr, Hennessy B and Mills GB: The biology of ovarian cancer: New opportunities for translation. Nat Rev Cancer. 9:415–428. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Kurman RJ and Shih Ie M: The origin and pathogenesis of epithelial ovarian cancer: A proposed unifying theory. Am J Surg Pathol. 34:433–443. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Chang HL, MacLaughlin DT and Donahoe PK: Somatic stem cells of the ovary and their relationship to human ovarian cancers. StemBook; Cambridge MA: Harvard Stem Cell Institute: 2008–2009

5 

Szotek PP, Chang HL, Brennand K, Fujino A, Pieretti-Vanmarcke R, Lo Celso C, Dombkowski D, Preffer F, Cohen KS, Teixeira J, et al: Normal ovarian surface epithelial label-retaining cells exhibit stem/progenitor cell characteristics. Proc Natl Acad Sci USA. 105:12469–12473. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Flesken-Nikitin A, Hwang CI, Cheng CY, Michurina TV, Enikolopov G and Nikitin AY: Ovarian surface epithelium at the junction area contains a cancer-prone stem cell niche. Nature. 495:241–245. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Aguilar-Gallardo C, Rutledge EC, Martínez-Arroyo AM, Hidalgo JJ, Domingo S and Simón C: Overcoming challenges of ovarian cancer stem cells: Novel therapeutic approaches. Stem Cell Rev. 8:994–1010. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Zhao J: Cancer stem cells and chemoresistance: The smartest survives the raid. Pharmacol Ther. 160:145–158. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Zhang S, Cui B, Lai H, Liu G, Ghia EM, Widhopf GF II, Zhang Z, Wu CC, Chen L, Wu R, et al: Ovarian cancer stem cells express ROR1, which can be targeted for anti-cancer-stem-cell therapy. Proc Natl Acad Sci USA. 111:17266–17271. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Wang Y, Cardenas H, Fang F, Condello S, Taverna P, Segar M, Liu Y, Nephew KP and Matei D: Epigenetic targeting of ovarian cancer stem cells. Cancer Res. 74:4922–4936. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Chau WK, Ip CK, Mak AS, Lai HC and Wong AS: c-Kit mediates chemoresistance and tumor-initiating capacity of ovarian cancer cells through activation of Wnt/β-catenin-ATP-binding cassette G2 signaling. Oncogene. 32:2767–2781. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Abubaker K, Latifi A, Luwor R, Nazaretian S, Zhu H, Quinn MA, Thompson EW, Findlay JK and Ahmed N: Short-term single treatment of chemotherapy results in the enrichment of ovarian cancer stem cell-like cells leading to an increased tumor burden. Mol Cancer. 12:242013. View Article : Google Scholar : PubMed/NCBI

13 

Feldman D, Krishnan AV, Swami S, Giovannucci E and Feldman BJ: The role of vitamin D in reducing cancer risk and progression. Nat Rev Cancer. 14:342–357. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Pervin S, Hewison M, Braga M, Tran L, Chun R, Karam A, Chaudhuri G, Norris K and Singh R: Down-regulation of vitamin D receptor in mammospheres: Implications for vitamin D resistance in breast cancer and potential for combination therapy. PLoS One. 8:e532872013. View Article : Google Scholar : PubMed/NCBI

15 

Pereira F, Larriba MJ and Muñoz A: Vitamin D and colon cancer. Endocr Relat Cancer. 19:R51–R71. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Larriba MJ and Muñoz A: SNAIL vs. vitamin D receptor expression in colon cancer: Therapeutics implications. Br J Cancer. 92:985–989. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Hou YF, Gao SH, Wang P, Zhang HM, Liu LZ, Ye MX, Zhou GM, Zhang ZL and Li BY: 1α,25(OH)2D3 suppresses the migration of ovarian cancer SKOV-3 cells through the inhibition of epithelial-mesenchymal transition. Int J Mol Sci. 17:12852016. View Article : Google Scholar :

18 

Maund SL, Barclay WW, Hover LD, Axanova LS, Sui G, Hipp JD, Fleet JC, Thorburn A and Cramer SD: Interleukin-1α mediates the antiproliferative effects of 1,25-dihydroxyvitamin D3 in prostate progenitor/stem cells. Cancer Res. 71:5276–5286. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Choudhury S, Almendro V, Merino VF, Wu Z, Maruyama R, Su Y, Martins FC, Fackler MJ, Bessarabova M, Kowalczyk A, et al: Molecular profiling of human mammary gland links breast cancer risk to a p27+ cell population with progenitor characteristics. Cell Stem Cell. 13:117–130. 2013. View Article : Google Scholar : PubMed/NCBI

20 

So JY, Lee HJ, Smolarek AK, Paul S, Wang CX, Maehr H, Uskokovic M, Zheng X, Conney AH, Cai L, et al: A novel Gemini vitamin D analog represses the expression of a stem cell marker CD44 in breast cancer. Mol Pharmacol. 79:360–367. 2011. View Article : Google Scholar : PubMed/NCBI

21 

So JY, Wahler J, Das Gupta S, Salerno DM, Maehr H, Uskokovic M and Suh N: HES1-mediated inhibition of Notch1 signaling by a Gemini vitamin D analog leads to decreased CD44+/CD24−/low tumor-initiating subpopulation in basal-like breast cancer. J Steroid Biochem Mol Biol. 148:111–121. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Yasuda K, Torigoe T, Morita R, Kuroda T, Takahashi A, Matsuzaki J, Kochin V, Asanuma H, Hasegawa T, Saito T, et al: Ovarian cancer stem cells are enriched in side population and aldehyde dehydrogenase bright overlapping population. PLoS One. 8:e681872013. View Article : Google Scholar : PubMed/NCBI

23 

Szotek PP, Pieretti-Vanmarcke R, Masiakos PT, Dinulescu DM, Connolly D, Foster R, Dombkowski D, Preffer F, Maclaughlin DT and Donahoe PK: Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness. Proc Natl Acad Sci USA. 103:11154–11159. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Hu L, McArthur C and Jaffe RB: Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br J Cancer. 102:1276–1283. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Rizzo S, Hersey JM, Mellor P, Dai W, Santos-Silva A, Liber D, Luk L, Titley I, Carden CP, Box G, et al: Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2. Mol Cancer Ther. 10:325–335. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Sales-Pardo I, Avendaño A, Martinez-Muñoz V, García-Escarp M, Celis R, Whittle P, Barquinero J, Domingo JC, Marin P and Petriz J: Flow cytometry of the side population: Tips & tricks. Cell Oncol. 28:37–53. 2006.PubMed/NCBI

27 

Roby KF, Taylor CC, Sweetwood JP, Cheng Y, Pace JL, Tawfik O, Persons DL, Smith PG and Terranova PF: Development of a syngeneic mouse model for events related to ovarian cancer. Carcinogenesis. 21:585–591. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

29 

de la Mare JA, Jurgens T and Edkins AL: Extracellular Hsp90 and TGFβ regulate adhesion, migration and anchorage independent growth in a paired colon cancer cell line model. BMC cancer. 17:2022017. View Article : Google Scholar : PubMed/NCBI

30 

McCloskey CW, Goldberg RL, Carter LE, Gamwell LF, Al-Hujaily EM, Collins O, Macdonald EA, Garson K, Daneshmand M, Carmona E, et al: A new spontaneously transformed syngeneic model of high-grade serous ovarian cancer with a tumor-initiating cell population. Front Oncol. 4:532014. View Article : Google Scholar : PubMed/NCBI

31 

Lobo NA, Shimono Y, Qian D and Clarke MF: The biology of cancer stem cells. Annu Rev Cell Dev Biol. 23:675–699. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Gerweck LE and Wakimoto H: At the crossroads of cancer stem cells, radiation biology, and radiation oncology. Cancer Res. 76:994–998. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Steg AD, Bevis KS, Katre AA, Ziebarth A, Dobbin ZC, Alvarez RD, Zhang K, Conner M and Landen CN: Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin Cancer Res. 18:869–881. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Nuti SV, Mor G, Li P and Yin G: TWIST and ovarian cancer stem cells: Implications for chemoresistance and metastasis. Oncotarget. 5:7260–7271. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Huang R, Wu D, Yuan Y, Li X, Holm R, Trope CG, Nesland JM and Suo Z: CD117 expression in fibroblasts-like stromal cells indicates unfavorable clinical outcomes in ovarian carcinoma patients. PLoS One. 9:e1122092014. View Article : Google Scholar : PubMed/NCBI

36 

Chen J, Wang J, Chen D, Yang J, Yang C, Zhang Y, Zhang H and Dou J: Evaluation of characteristics of CD44+CD117+ ovarian cancer stem cells in three dimensional basement membrane extract scaffold versus two dimensional monocultures. BMC Cell Biol. 14:72013. View Article : Google Scholar : PubMed/NCBI

37 

Yanamoto S, Kawasaki G, Yamada S, Yoshitomi I, Kawano T, Yonezawa H, Rokutanda S, Naruse T and Umeda M: Isolation and characterization of cancer stem-like side population cells in human oral cancer cells. Oral Oncol. 47:855–860. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Ruan Z, Liu J and Kuang Y: Isolation and characterization of side population cells from the human ovarian cancer cell line SK-OV-3. Exp Ther Med. 10:2071–2078. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Hendrickson WK, Flavin R, Kasperzyk JL, Fiorentino M, Fang F, Lis R, Fiore C, Penney KL, Ma J, Kantoff PW, et al: Vitamin D receptor protein expression in tumor tissue and prostate cancer progression. J Clin Oncol. 29:2378–2385. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Ditsch N, Toth B, Mayr D, Lenhard M, Gallwas J, Weissenbacher T, Dannecker C, Friese K and Jeschke U: The association between vitamin D receptor expression and prolonged overall survival in breast cancer. J Histochem Cytochem. 60:121–129. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Jiang F, Li P, Fornace AJ Jr, Nicosia SV and Bai W: G2/M arrest by 1,25-dihydroxyvitamin D3 in ovarian cancer cells mediated through the induction of GADD45 via an exonic enhancer. J Biol Chem. 278:48030–48040. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Jiang F, Bao J, Li P, Nicosia SV and Bai W: Induction of ovarian cancer cell apoptosis by 1,25-dihydroxyvitamin D3 through the down-regulation of telomerase. J Biol Chem. 279:53213–53221. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Zhang X, Jiang F, Li P, Li C, Ma Q, Nicosia SV and Bai W: Growth suppression of ovarian cancer xenografts in nude mice by vitamin D analogue EB1089. Clin Cancer Res. 11:323–328. 2005.PubMed/NCBI

44 

Hiraga T, Ito S and Nakamura H: Cancer stem-like cell marker CD44 promotes bone metastases by enhancing tumorigenicity, cell motility, and hyaluronan production. Cancer Res. 73:4112–4122. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, et al: The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 17:211–215. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Kashyap V, Rezende NC, Scotland KB, Shaffer SM, Persson JL, Gudas LJ and Mongan NP: Regulation of stem cell pluripotency and differentiation involves a mutual regulatory circuit of the NANOG, OCT4, and SOX2 pluripotency transcription factors with polycomb repressive complexes and stem cell microRNAs. Stem Cells Dev. 18:1093–1108. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, Murray HL, Jenner RG, et al: Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 122:947–956. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Bourguignon LY, Wong G, Earle C and Chen L: Hyaluronan-CD44v3 interaction with Oct4-Sox2-Nanog promotes miR-302 expression leading to self-renewal, clonal formation, and cisplatin resistance in cancer stem cells from head and neck squamous cell carcinoma. J Biol Chem. 287:32800–32824. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Schneider S, Steinbeisser H, Warga RM and Hausen P: Beta-catenin translocation into nuclei demarcates the dorsalizing centers in frog and fish embryos. Mech Dev. 57:191–198. 1996. View Article : Google Scholar : PubMed/NCBI

50 

Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, Hein K, Vogt R and Kemler R: Wnt/β-catenin signaling regulates telomerase in stem cells and cancer cells. Science. 336:1549–1554. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Lee E, Madar A, David G, Garabedian MJ, Dasgupta R and Logan SK: Inhibition of androgen receptor and beta-catenin activity in prostate cancer. Proc Natl Acad Sci USA. 110:15710–15715. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Li S, Li S, Sun Y and Li L: The expression of beta-catenin in different subtypes of breast cancer and its clinical significance. Tumour Biol. 35:7693–7698. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Wang H, Wang H, Makki MS, Wen J, Dai Y, Shi Q, Liu Q, Zhou X and Wang J: Overexpression of β-catenin and cyclinD1 predicts a poor prognosis in ovarian serous carcinomas. Int J Clin Exp Pathol. 7:264–271. 2014.PubMed/NCBI

54 

Clevers H and Nusse R: Wnt/β-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Sebio A, Kahn M and Lenz HJ: The potential of targeting Wnt/β-catenin in colon cancer. Expert Opin Ther Targets. 18:611–615. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Larriba MJ, Ordóñez-Moran P, Chicote I, Martín-Fernández G, Puig I, Muñoz A and Pálmer HG: Vitamin D receptor deficiency enhances Wnt/beta-catenin signaling and tumor burden in colon cancer. PLoS One. 6:e235242011. View Article : Google Scholar : PubMed/NCBI

57 

Jiang YJ, Teichert AE, Fong F, Oda Y and Bikle DD: 1α,25(OH)2-dihydroxyvitamin D3/VDR protects the skin from UVB-induced tumor formation by interacting with the beta-catenin pathway. J Steroid Biochem Mol Biol. 136:229–232. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Pálmer HG, González-Sancho JM, Espada J, Berciano MT, Puig I, Baulida J, Quintanilla M, Cano A, de Herreros AG, Lafarga M and Muñoz A: Vitamin D3 promotes the differentiation of colon carcinoma cells by the induction of E-cadherin and the inhibition of beta-catenin signaling. J Cell Biol. 154:369–387. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2019
Volume 41 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ji M, Liu L, Hou Y and Li B: 1α,25‑Dihydroxyvitamin D3 restrains stem cell‑like properties of ovarian cancer cells by enhancing vitamin D receptor and suppressing CD44. Oncol Rep 41: 3393-3403, 2019
APA
Ji, M., Liu, L., Hou, Y., & Li, B. (2019). 1α,25‑Dihydroxyvitamin D3 restrains stem cell‑like properties of ovarian cancer cells by enhancing vitamin D receptor and suppressing CD44. Oncology Reports, 41, 3393-3403. https://doi.org/10.3892/or.2019.7116
MLA
Ji, M., Liu, L., Hou, Y., Li, B."1α,25‑Dihydroxyvitamin D3 restrains stem cell‑like properties of ovarian cancer cells by enhancing vitamin D receptor and suppressing CD44". Oncology Reports 41.6 (2019): 3393-3403.
Chicago
Ji, M., Liu, L., Hou, Y., Li, B."1α,25‑Dihydroxyvitamin D3 restrains stem cell‑like properties of ovarian cancer cells by enhancing vitamin D receptor and suppressing CD44". Oncology Reports 41, no. 6 (2019): 3393-3403. https://doi.org/10.3892/or.2019.7116