Open Access

Vitamin K2 and cotylenin A synergistically induce monocytic differentiation and growth arrest along with the suppression of c-MYC expression and induction of cyclin G2 expression in human leukemia HL-60 cells

  • Authors:
    • Yasuhisa Maniwa
    • Takashi Kasukabe
    • Shunichi Kumakura
  • View Affiliations

  • Published online on: June 4, 2015     https://doi.org/10.3892/ijo.2015.3028
  • Pages: 473-480
  • Copyright: © Maniwa et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although all-trans retinoic acid (ATRA) is a standard and effective drug used for differentiation therapy in acute promyelocytic leukemia, ATRA-resistant leukemia cells ultimately emerge during this treatment. Therefore, the development of new drugs or effective combination therapy is urgently needed. We demonstrate that the combined treatment of vitamin K2 and cotylenin A synergistically induced monocytic differentiation in HL-60 cells. This combined treatment also synergistically induced NBT-reducing activity and non-specific esterase-positive cells as well as morphological changes to monocyte/macrophage-like cells. Vitamin K2 and cotylenin A cooperatively inhibited the proliferation of HL-60 cells in short-term and long-term cultures. This treatment also induced growth arrest at the G1 phase. Although 5 µg/ml cotylenin A or 5 µM vitamin K2 alone reduced c-MYC gene expression in HL-60 cells to approximately 45% or 80% that of control cells, respectively, the combined treatment almost completely suppressed c-MYC gene expression. We also demonstrated that the combined treatment of vitamin K2 and cotylenin A synergistically induced the expression of cyclin G2, which had a positive effect on the promotion and maintenance of cell cycle arrest. These results suggest that the combination of vitamin K2 and cotylenin A has therapeutic value in the treatment of acute myeloid leukemia.

Introduction

Acute myeloid leukemia (AML) is the most common type of leukemia in adults, and occurs in approximately one-third of newly diagnosed patients, and remains one of the most difficult hematological malignancies to treat (the 5-year overall survival rate is 20–30% for adult primary AML) (1,2). AML is characterized by the proliferation of clonal precursor myeloid cells with arrested differentiation (3). In contrast to the poor prognosis of most patients with AML, the use of differentiation therapy with all-trans-retinoic acid (ATRA) for acute promyelocytic leukemia (APL), a distinct type of AML, has revolutionized therapy for this disease by converting it from fatal to curable (4). However, ATRA is not effective in other AMLs. Furthermore, many APL patients treated with ATRA fail to respond or invariably relapse. Therefore, alternative or combination therapies are needed to improve the prognosis and survival of patients.

Cotylenin A (CN-A), which is a fucicoccan-diterpene glycoside with a complex sugar moiety, was originally isolated as a plant growth regulator and has been shown to affect several physiological processes in higher plants (5). We previously reported that CN-A exhibited potent differentiation-inducing activity in several human and murine myeloid leukemia cell lines and in leukemia cells that were freshly isolated from patients with AML (69). Furthermore, the administration of CN-A significantly prolonged the survival of mice with severe combined immunodeficiency that had been inoculated with the APL cells of the NB-4 cell line (10).

Previous studies reported that vitamin K2 (VK2) effectively induced apoptosis in various types of primary cultured leukemia cells and leukemia cell lines in vitro (1113) as well as in solid tumor cells (1416). On the other hand, in contrast to the induction of apoptosis in leukemia cells, VK2 has been shown to exhibit differentiation-inducing activity in AML cell lines, such as HL-60 and U937, in vitro (1719). Sada et al found that VK2 also had differentiation-promoting effects on myeloid lineage progenitors (20).

Since c-MYC is aberrantly expressed in a wide variety of human solid tumors (21) as well as in leukemia (22), it is an attractive target for cancer therapy. The downregulation of c-MYC is known to play a crucial role in ATRA-induced growth arrest and myeloid differentiation of AML (2327). In addition, previous findings, including ours, indicated that the expression of cyclin G2 was significantly upregulated during cell cycle arrest responses to diverse growth-inhibitory signals and strongly repressed by mitogens, suggesting the positive role of cyclin G2 in the promotion or maintenance of cell cycle arrest (2830). In order to identify useful new differentiation inducers and effective combination treatments for various types of AML and APL, we searched for substances capable of inducing cell differentiation and the expression of cyclin G2 as well as strongly suppressing the expression of c-MYC in HL-60 cells. In the present study, we demonstrated that the combined treatment of VK2 and CN-A synergistically induced monocytic differentiation in HL-60 cells and cooperatively inhibited cell proliferation showing G1 arrest. Furthermore, we showed that the combined treatment of VK2 and CN-A efficiently suppressed the expression of c-MYC and cooperatively induced the expression of cyclin G2.

Materials and methods

Reagents

VK2, nitroblue tetrazolium (NBT), all-trans retinoic acid (ATRA), 1α,25-dihydroxyvitamin D3 (VD3), and 12-O-tetradecanoylphorbol-13-acetate (TPA) were purchased from Sigma-Aldrich Inc. (St. Louis, MO, USA). CN-A was purified from a stock ethyl acetate extract obtained from the culture filtrate of Cladosporium fungus sp. 501-7 W by flash chromatography on a silica gel with >99% purity (5).

Cells and cell culture

Human AML HL-60 cells were cultured in RPMI-1640 medium (Sigma-Aldrich Inc.) supplemented with 10% heat-inactivated fetal bovine serum and 80 μg/ml gentamicin sulfate (MSD K.K, Tokyo, Japan) at 37°C in a humidified atmosphere of 5% CO2 in air.

Assay of cell growth

Cells were plated in multidishes (Falcon, Corning Inc., Corning, NY, USA) at a density of 2.5×104 cells/ml and incubated with or without the test compounds. Cell numbers were counted with a model Z1 Coulter Counter (Beckman Coulter Inc., Miami, FL, USA).

NBT reduction assay

The reduction of NBT was assayed colorimetrically as previously described (31). Briefly, cells were incubated in 1 ml of serum-free medium containing 1 mg/ml NBT and 100 ng/ml TPA at 37°C for 60 min. The reaction was stopped by adding HCl. Formazan solution at 560 nm was measured in a spectrophotometer (DU730, Beckman Coulter Inc.).

Assessment of monocytic differentiation

In order to assess monocytic differentiation, non-specific esterase staining was performed using an Esterase Staining kit (Muto Chemical Co., Tokyo, Japan).

Assessment of morphological differentiation

Morphological changes were examined in cell smears using light microscopy of cytospin preparations stained with May-Grunwald-Giemsa solution (Merck, Darmstadt, Germany).

Cell cycle analysis

Cells were plated in 60-mm plastic dishes at a density of 1×105 cells/ml and incubated with VK2 in the absence or presence of CN-A. After 96 h, the cells were washed with phosphate-buffered saline (PBS) and fixed gently in 100% ethanol at 4°C for 30 min. Cells were suspended in propidium iodide (PI)-RNase solution, which contained 50 μg/ml PI (MBL Co. Ltd., Nagoya, Japan) and 0.1 mg/ml RNase (Sigma-Aldrich Inc.) in PBS for 30 min at room temperature. The cell cycle analysis was performed by flow cytometry (BD FACSCalibur, Becton Dickinson, East Rutherford, NJ, USA).

RNA extraction and determination of mRNA levels by reverse transcriptase (RT)-quantitative polymerase chain reaction (qPCR)

RNA was extracted using an RNeasy Plus Mini kit (Qiagen, Valencia, CA, USA) according to the manufacturer’s instructions. Total RNA (1 μg) from leukemia cells was reverse transcribed with the ReverTra Ace qPCR RT kit (Toyobo Co. Ltd., Osaka, Japan). qPCR using the SYBER Green method was carried out with the Thunderbird SYBER qPCR Mix (Toyobo Co.) on a Thermal Cycler Dice Real-time PCR instrument (Takara Bio, Shiga, Japan) according to the manufacturer’s instructions. Real-time PCR results were calculated according to the following protocol: Relative expression level=2−ΔCt, where ΔCt=Ct (gene of interest) - Ct (housekeeping gene). The c-MYC primers used for qPCR were: forward, 5′-TTCGGGT AGTGGAAAACCAG-3′ and reverse, 5′-CAGCAGCTCGAA TTTCTTCC-3′. The GAPDH primers used for qPCR were: forward, 5′-GACGCTGGGGCTGGCATTG-3′ and reverse, 5′-GCTGGTGGTCCAGGGGTC-3′ (32). The cyclin G2 primers used for qPCR were: forward, 5′-ATCGTTTCAAG GCGCACAG-3′ and reverse, 5′-CAACCCCCCTCAGGTA TCG-3′ (33). The P21/CIP1 primers used for qPCR were: forward, 5′-CGATGCCAACCTCCTCAACGA-3′ and reverse, 5′-TCGCAGACCTCCAGCATCCA-3 (34).

Results

Effects of the combined treatment of vitamin K2 (VK2) and cotylenin A (CN-A) on the cell proliferation of HL-60 cells

HL-60 cells (2.5×104 cells/ml) were cultured without or with VK2, CN-A, or VK2 plus CN-A for 6 days. Fig. 1A shows the time course of the combined effects of VK2 and CN-A on cell growth. The growth of HL-60 cells was moderately inhibited by VK2 (5 μM) or CN-A (5 μg/ml) alone, but was still observed until at least 6 days; however, no significant changes were observed in the cell number after 4 days of the treatment with the combination of both VK2 and CN-A (Fig. 1A). We also examined the long-term effects of the combined treatment of VK2 and CN-A on the proliferation of HL-60 cells. HL-60 cells (5×104 cells/ml) were cultured without or with 10 μM VK2, 5 μg/ml CN-A, or 10 μM VK2 plus 5 μg/ml CN-A for 20 days (Fig. 1B). The culture medium was replaced by fresh medium once every 5 days. Although the growth rate of VK2- or CN-A-treated cells was significantly lower than that of control cells under these culture conditions, the cell number markedly increased (100-fold between days 5 and 20). On the other hand, cell growth was greatly inhibited by the combined treatment of VK2 and CN-A, and the cell number was almost the same as that at day 5 (Fig. 1B).

VK2 and CN-A synergistically induced monocytic differentiation in HL-60 cells

We examined the combined effects of VK2 and CN-A on the induction of differentiation of HL-60 cells because VK2 or CN-A alone are inducers of differentiation in HL-60 cells (6,7,17,18). HL-60 cells (2.5×104 cells/ml) were cultured with CN-A in the presence or absence of VK2 for 6 days. CN-A and 10 μM VK2 synergistically induced the reduction of NBT (one of the typical myelo/monocytic differentiation markers of human leukemia cells) (Fig. 2). We then determined whether the induction of differentiation induced with VK2 plus CN-A was a granulocytic or monocytic lineage. HL-60 cells (2.5×104 cells/ml) were cultured without or with 10 μg/ml CN-A, 10 μM VK2, or 10 μM VK2 plus 10 μg/ml CN-A for 5 days (Fig. 3A). Nonspecific esterase-positive cells were counted under a microscope (Fig. 3B). Cells treated with CN-A plus VK2 synergistically became positive for nonspecific esterase (Fig. 3A-d and B), whereas those treated with CN-A or VK2 alone became weakly positive (Fig. 3A–b, A–c and B). The combined treatment of VK2 and CN-A also induced the marked morphological differentiation of HL-60 cells (Fig. 4D), whereas VK2 or CN-A alone induced the intermediate stage of differentiation (Fig. 4B and C). These results indicated that the treatment of HL-60 cells with VK2 and CN-A effectively induced monocytic differentiation.

Induction of G1 arrest in HL-60 cells with VK2 plus CN-A

In order to more clearly understand the combined effects of VK2 and CN-A on cell growth, we exposed HL-60 cells (1×105 cells/ml) to 10 μM VK2 plus 10 μg/ml CN-A, and then measured changes in cell cycle distribution after 4 days (Fig. 5). Under these culture conditions, VK2 or CN-A alone did not markedly affect the cell cycle (Fig. 5B and C). On the other hand, the percentage of cells in the G1 phase was significantly increased from 63% to 75% (Fig. 5A and D). The percentages of cells in the S phase and G2/M phase were inversely decreased in response to the combined treatment of VK2 and CN-A (Fig. 5A and D).

Combined treatment of VK2 and CN-A synergistically inhibited c-MYC gene expression in HL-60 cells

Previous studies reported that the induction of differentiation and growth arrest in HL-60 cells was associated with the suppression of c-MYC gene expression (2327); therefore, we investigated whether the combined treatment of CN-A and VK2 synergistically inhibited c-MYC gene expression in HL-60 cells. HL-60 cells (2.5×104 cells/ml) were cultured without or with VK2 plus CN-A for 6 days. Although 5 μg/ml CN-A or 5 μM VK2 alone inhibited c-MYC gene expression in HL-60 cells to approximately 45 or 80% that of control cells, respectively, the combined treatment almost completely suppressed c-MYC gene expression (>95% inhibition) (Fig. 6A). This synergistic inhibition of c-MYC gene expression in HL-60 cells was also observed when HL-60 cells were treated with CN-A and VK2 for 4 days (data not shown). As described above, the combined treatment of CN-A and VK2 more strongly inhibited cell growth than that of CN-A or VK2 alone (Fig. 6B) and clearly induced monocytic differentiation (Fig. 4).

Combined treatment of VK2 and CN-A synergistically induced cyclin G2 gene expression in HL-60 cells

Previous findings, including ours, indicated that the expression of cyclin G2 was significantly upregulated during cell cycle arrest responses to diverse growth-inhibitory signals and strongly repressed by mitogens, suggesting a positive role for cyclin G2 in the promotion or maintenance of cell cycle arrest (2830). Therefore, we determined whether the differentiation of HL-60 cells induced with VK2 and CN-A was accompanied by the induction of cyclin G2 expression. Cyclin G2 gene expression was markedly induced (>5-fold) in VK2 plus CN-A-treated HL-60 cells (Fig. 7A). The expression of cyclin G2 was approximately 2-fold higher in CN-A-treated HL-60 cells than in control cells, whereas VK2-treated cells showed only a marginal increase (Fig. 7A). Similar results were obtained when HL-60 cells were treated with VK2 plus CN-A for 4 days (data not shown).

We also examined the gene expression levels of several cell cycle regulators such as p21/CIP1, p27/KIP1, and cyclin D1. We did not observe the marked induction (>2-fold) of the expression of p21/CIP1 (Fig. 7B), p27/KIP1 (data not shown), or cyclin D1 (data not shown) in VK2-, CN-A-, or VK2 plus CN-A-treated HL-60 cells.

Effects of VK2 on the expression of c-MYC and cyclin G2 in VD3- or ATRA-treated HL-60 cells

We examined the effects of VK2 on cell growth and the expression of c-MYC and cyclin G2 in HL-60 cells treated with two typical differentiation inducers. VD3 is one of the most potent monocytic differentiation inducers identified to date (35,36). Although VD3 or VK2 alone inhibited cell growth to approximately 65 or 45% that of control cells and also suppressed the expression of c-MYC to approximately 50 or 90% that of control cells, respectively, the combined treatment of VD3 and VK2 inhibited cell growth to approximately 30% that of control cells and suppressed the expression of c-MYC to <10% that of control cells (Fig. 8A and B). Furthermore, VD3 plus VK2 cooperatively induced cyclin G2 gene expression more than that of the additive manner (Fig. 8C). On the other hand, ATRA alone, which is a standard drug used for differentiation therapy in APL, inhibited cell growth to approximately 20% that of control cells, suppressed c-MYC expression to <10% that of control cells, and markedly induced cyclin G2 gene expression (6.9-fold) (Fig. 8). Under this treatment condition using ATRA, VK2 marginally increased the effects of ATRA on cell growth, c-MYC expression, and cyclin G2 expression (Fig. 8).

Discussion

Vitamin Ks (VK) are known to act as co-factor for the γ-carboxylation of prothrombin and other VK-dependent coagulation factors (37). VK promotes osteogenesis through the γ-carboxylation of glutamate residues in osteocalcin. VK2 is a naturally-occurring and the main form of vitamin K in the tissues. A synthetic VK2 analog has been approved as an anti-osteoporotic medicine by the Ministry of Health, Labor and Welfare in Japan. The safety of the long-term administration of VK2 has been well established (38). Although the exact mechanism has not yet been elucidated in detail, VK2 and their analogs have been shown to inhibit the survival of various cancer cell lines (1416) and leukemia cells (1113). Furthermore, previous studies reported that VK2 exhibited some differentiation-inducing activity in AML cell lines in vitro (1719). Only VK2 alone induced the intermediate stage of differentiation in HL-60 cells in the present study (Fig. 4D) and, even at higher concentrations (>10 μM), VK2 could not induce mature differentiation, but induced apoptosis (data not shown). As VK2 is a naturally-occurring, safe, and clinically-utilized agent, we searched for substances that could enhance the differentiation-inducing activity of VK2. We found that CN-A, a differentiation inducer, synergistically induced the differentiation of HL-60 cells along with growth arrest, and markedly suppressed the expression of c-MYC and induction of cyclin G2 expression. This is the first study to examine the effects of VK2 plus CN-A on the induction of differentiation and expression of growth arrest-associated genes such as c-MYC and cyclin G2.

The proto-oncogene c-MYC has been shown to play an important role in cellular metabolism, apoptosis, differentiation, cell cycle progress and tumorigenesis (3643). The expression of c-MYC in particular was found to contribute to leukemogenesis and promote the progression of leukemia (43). The downregulation of c-MYC is critical for the ATRA-induced growth arrest and myeloid differentiation of AML (2327). The inhibition of c-MYC was also shown to suppress the proliferation and induce the differentiation of primary AML cells (27). Furthermore, the overexpression of c-MYC in an APL cell line inversely inhibited ATRA-induced cell differentiation (27). These findings indicate that c-MYC is an attractive target for differentiation therapy.

In the present study, we found that VK2 markedly enhanced the downregulation of c-MYC gene expression induced by differentiation inducers, whereas VK2 alone at the doses used weakly suppressed gene expression (Figs. 6A and 8B). The combined treatment of VK2 and CN-A exhibited the most potent suppressive effects on c-MYC gene expression among the inducers or their combinations tested. This combined treatment reduced the expression of c-MYC to approximately one fortieth that of control levels (Fig. 6A), and synergistically induced differentiation (Figs. 3 and 4) and growth arrest (Figs. 1 and 5). Although VK2 also effectively enhanced the suppressive effects of c-MYC expression induced by VD3 (Fig. 8B), VK2 plus VD3 reduced the expression of c-MYC less than that of VK2 plus CN-A (Figs. 6A and 8B). Furthermore, no derivative of active vitamin D3 has so far been used clinically as an anticancer agent because of the side effect of hypercalcemia (44). These results suggest that the combination of VK2 and CN-A has therapeutic value in the treatment of AML. Furthermore, since we previously found that CN-A was also capable of stimulating the functional and morphological differentiation of ATRA-resistant APL cells (10), the combined treatment of VK2 plus CN-A may be useful for differentiation therapy in retinoid-resistant leukemia.

The expression of cyclin G2 was shown to be significantly upregulated during cell cycle arrest responses to diverse growth-inhibitory signals and strongly repressed by mitogens, suggesting the positive role of cyclin G2 in the promotion or maintenance of cell cycle arrest (2830). We previously reported that the combination of differentiation inducers including CN-A effectively inhibited the proliferation of several human breast cancer cell lines as well as leukemia cells (27,43). This treatment induced growth arrest in cells at the G1 phase rather than apoptosis and also rapidly and markedly induced cyclin G2 gene expression (29,45).

Cyclin G2 knockdown induced by cyclin G2 small interfering RNA markedly reduced the potency of CN-A plus other inducers to induce growth inhibition. Ectopically inducible cyclin G2 expression was shown to potently inhibit the proliferation of breast cancer cells (29). Therefore, for more effective differentiation therapy in AML, it is necessary to find new differentiation inducers or combination therapies that can induce cell differentiation and growth arrest. We have searched for substances that are capable of inducing cell differentiation and the expression of cyclin G2, and that can also strongly suppress the expression of c-MYC in HL-60 cells. We showed that the treatment with VK2 plus CN-A induced functional and morphological differentiation as well as growth arrest in HL-60 AML cells. Furthermore, this treatment almost completely suppressed the expression of c-MYC and markedly induced the expression of cyclin G2. Therefore, our results suggest an attractive combination for effective differentiation therapy in human myeloid leukemia. More detailed studies on the mechanisms underlying this effective combined treatment of VK2 plus CN-A are required.

Acknowledgements

This study was supported partly by a grant from the Ministry of Education, Culture, Sports, Science and Technology of Japan and by a grant from Shimane University ‘SUIGAN’ project.

References

1 

Guerrouahen BS, Futami M, Vaklavas C, Kanerva J, Whichard ZL, Nwawka K, Blanchard EG, Lee FY, Robinson LJ, Arceci R, et al: Dasatinib inhibits the growth of molecularly heterogeneous myeloid leukemias. Clin Cancer Res. 16:1149–1158. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Robak T and Wierzbowska A: Current and emerging therapies for acute myeloid leukemia. Clin Ther. 31:2349–2370. 2009. View Article : Google Scholar

3 

McCulloch EA: Stem cells in normal and leukemic hemopoiesis (Henry Stratton Lecture, 1982). Blood. 62:1–13. 1983.PubMed/NCBI

4 

Petrie K, Zelent A and Waxman S: Differentiation therapy of acute myeloid leukemia: Past, present and future. Curr Opin Hematol. 16:84–91. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Sassa T, Tojyo T and Munakata K: Isolation of a new plant growth substance with cytokinin-like activity. Nature. 227:3791970. View Article : Google Scholar : PubMed/NCBI

6 

Asahi K, Honma Y, Hazeki K, Sassa T, Kubohara Y, Sakurai A and Takahashi N: Cotylenin A, a plant-growth regulator, induces the differentiation in murine and human myeloid leukemia cells. Biochem Biophys Res Commun. 238:758–763. 1997. View Article : Google Scholar : PubMed/NCBI

7 

Yamamoto-Yamaguchi Y, Yamada K, Ishii Y, Asahi KI, Tomoyasu S and Honma Y: Induction of the monocytic differentiation of myeloid leukaemia cells by cotylenin A, a plant growth regulator. Br J Haematol. 112:697–705. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Yamada K, Honma Y, Asahi KI, Sassa T, Hino KI and Tomoyasu S: Differentiation of human acute myeloid leukaemia cells in primary culture in response to cotylenin A, a plant growth regulator. Br J Haematol. 114:814–821. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Honma Y: Cotylenin A - a plant growth regulator as a differentiation-inducing agent against myeloid leukemia. Leuk Lymphoma. 43:1169–1178. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Honma Y, Ishii Y, Sassa T and Asahi K: Treatment of human promyelocytic leukemia in the SCID mouse model with cotylenin A, an inducer of myelomonocytic differentiation of leukemia cells. Leuk Res. 27:1019–1025. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Yaguchi M, Miyazawa K, Katagiri T, Nishimaki J, Kizaki M, Tohyama K and Toyama K: Vitamin K2 and its derivatives induce apoptosis in leukemia cells and enhance the effect of all-trans retinoic acid. Leukemia. 11:779–787. 1997. View Article : Google Scholar : PubMed/NCBI

12 

Yokoyama T, Miyazawa K, Naito M, Toyotake J, Tauchi T, Itoh M, Yuo A, Hayashi Y, Georgescu MM, Kondo Y, et al: Vitamin K2 induces autophagy and apoptosis simultaneously in leukemia cells. Autophagy. 4:629–640. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Kitagawa J, Hara T, Tsurumi H, Ninomiya S, Ogawa K, Adachi S, Kanemura N, Kasahara S, Shimizu M and Moriwaki H: Synergistic growth inhibition in HL-60 cells by the combination of acyclic retinoid and vitamin K2. J Cancer Res Clin Oncol. 137:779–787. 2011. View Article : Google Scholar

14 

Mizuta T and Ozaki I: Hepatocellular carcinoma and vitamin K. Vitam Horm. 78:435–442. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Matsumoto K, Okano J, Nagahara T and Murawaki Y: Apoptosis of liver cancer cells by vitamin K2 and enhancement by MEK inhibition. Int J Oncol. 29:1501–1508. 2006.PubMed/NCBI

16 

Showalter SL, Wang Z, Costantino CL, Witkiewicz AK, Yeo CJ, Brody JR and Carr BI: Naturally occurring K vitamins inhibit pancreatic cancer cell survival through a caspase-dependent pathway. J Gastroenterol Hepatol. 25:738–744. 2010. View Article : Google Scholar

17 

Sakai I, Hashimoto S, Yoda M, Hida T, Ohsawa S, Nakajo S and Nakaya K: Novel role of vitamin K2: A potent inducer of differentiation of various human myeloid leukemia cell lines. Biochem Biophys Res Commun. 205:1305–1310. 1994. View Article : Google Scholar : PubMed/NCBI

18 

Miyazawa K, Yaguchi M, Funato K, Gotoh A, Kawanishi Y, Nishizawa Y, Yuo A and Ohyashiki K: Apoptosis/differentiation-inducing effects of vitamin K2 on HL-60 cells: Dichotomous nature of vitamin K2 in leukemia cells. Leukemia. 15:1111–1117. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Funato K, Miyazawa K, Yaguchi M, Gotoh A and Ohyashiki K: Combination of 22-oxa-1,25-dihydroxyvitamin D(3), a vitamin D(3) derivative, with vitamin K(2) (VK2) synergistically enhances cell differentiation but suppresses VK2-inducing apoptosis in HL-60 cells. Leukemia. 16:1519–1527. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Sada E, Abe Y, Ohba R, Tachikawa Y, Nagasawa E, Shiratsuchi M and Takayanagi R: Vitamin K2 modulates differentiation and apoptosis of both myeloid and erythroid lineages. Eur J Haematol. 85:538–548. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Field JK and Spandidos DA: The role of ras and myc oncogenes in human solid tumours and their relevance in diagnosis and prognosis (Review). Anticancer Res. 10:1–22. 1990.PubMed/NCBI

22 

Delgado MD, Albajar M, Gomez-Casares MT, Batlle A and León J: MYC oncogene in myeloid neoplasias. Clin Transl Oncol. 15:87–94. 2013. View Article : Google Scholar

23 

Kumakura S, Ishikura H, Tsumura H, Hayashi H, Endo J and Tsunematsu T: c-myc protein expression during cell cycle phases in differentiating HL-60 cells. Leuk Lymphoma. 14:171–180. 1994. View Article : Google Scholar : PubMed/NCBI

24 

Kumakura S, Ishikura H, Tsumura H, Iwata Y, Endo J and Kobayashi S: C-Myc and Bcl-2 protein expression during the induction of apoptosis and differentiation in TNF alpha-treated HL-60 cells. Leuk Lymphoma. 23:383–394. 1996. View Article : Google Scholar : PubMed/NCBI

25 

Jiang G, Albihn A, Tang T, Tian Z and Henriksson M: Role of Myc in differentiation and apoptosis in HL60 cells after exposure to arsenic trioxide or all-trans retinoic acid. Leuk Res. 32:297–307. 2008. View Article : Google Scholar

26 

Cheng YC, Lin H, Huang MJ, Chow JM, Lin S and Liu HE: Downregulation of c-Myc is critical for valproic acid-induced growth arrest and myeloid differentiation of acute myeloid leukemia. Leuk Res. 31:1403–1411. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Pan XN, Chen JJ, Wang LX, Xiao RZ, Liu LL, Fang ZG, Liu Q, Long ZJ and Lin DJ: Inhibition of c-Myc overcomes cytotoxic drug resistance in acute myeloid leukemia cells by promoting differentiation. PLoS One. 9:e1053812014. View Article : Google Scholar : PubMed/NCBI

28 

Horne MC, Donaldson KL, Goolsby GL, Tran D, Mulheisen M, Hell JW and Wahl AF: Cyclin G2 is up-regulated during growth inhibition and B cell antigen receptor-mediated cell cycle arrest. J Biol Chem. 272:12650–12661. 1997. View Article : Google Scholar : PubMed/NCBI

29 

Kasukabe T, Okabe-Kado J and Honma Y: Cotylenin A, a new differentiation inducer, and rapamycin cooperatively inhibit growth of cancer cells through induction of cyclin G2. Cancer Sci. 99:1693–1698. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Zimmermann M, Arachchige-Don AS, Donaldson MS, Dallapiazza RF, Cowan CE and Horne MC: Elevated cyclin G2 expression intersects with DNA damage checkpoint signaling and is required for a potent G2/M checkpoint arrest response to doxorubicin. J Biol Chem. 287:22838–22853. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Kasukabe T, Honma Y, Hozumi M and Nomura H: Inhibition of proliferation and induction of differentiation of human and mouse myeloid leukemia cells by new ethyleneglycol-type nonphosphorus alkyl ether lipids. Jpn J Cancer Res. 81:807–812. 1990. View Article : Google Scholar : PubMed/NCBI

32 

Marzi I, Cipolleschi MG, D’Amico M, Stivarou T, Rovida E, Vinci MC, Pandolfi S, Dello Sbarba P, Stecca B and Olivotto M: The involvement of a Nanog, Klf4 and c-Myc transcriptional circuitry in the intertwining between neoplastic progression and reprogramming. Cell Cycle. 12:353–364. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Stossi F, Likhite VS, Katzenellenbogen JA and Katzenellenbogen BS: Estrogen-occupied estrogen receptor represses cyclin G2 gene expression and recruits a repressor complex at the cyclin G2 promoter. J Biol Chem. 281:16272–16278. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Liang X-H, Li L-L, Wu G-G, Xie Y-C, Zhang G-X, Chen W, Yang HF, Liu QL, Li WH, He WG, et al: Upregulation of CPE promotes cell proliferation and tumorigenicity in colorectal cancer. BMC Cancer. 13:4122013. View Article : Google Scholar : PubMed/NCBI

35 

Tanaka H, Abe E, Miyaura C, Kuribayashi T, Konno K, Nishii Y and Suda T: 1 alpha,25-Dihydroxycholecalciferol and a human myeloid leukaemia cell line (HL-60). Biochem J. 204:713–719. 1982.PubMed/NCBI

36 

Kasukabe T, Honma Y, Hozumi M, Suda T and Nishii Y: Control of proliferating potential of myeloid leukemia cells during long-term treatment with vitamin D3 analogues and other differentiation inducers in combination with antileukemic drugs: In vitro and in vivo studies. Cancer Res. 47:567–572. 1987.PubMed/NCBI

37 

Vermeer C and Schurgers LJ: A comprehensive review of vitamin K and vitamin K antagonists. Hematol Oncol Clin North Am. 14:339–353. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Sasaki N, Kusano E, Takahashi H, Ando Y, Yano K, Tsuda E and Asano Y: Vitamin K2 inhibits glucocorticoid-induced bone loss partly by preventing the reduction of osteoprotegerin (OPG). J Bone Miner Metab. 23:41–47. 2005. View Article : Google Scholar

39 

Masui K, Tanaka K, Akhavan D, Babic I, Gini B, Matsutani T, Iwanami A, Liu F, Villa GR, Gu Y, et al: mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab. 18:726–739. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Sheth A, Escobar-Alvarez S, Gardner J, Ran L, Heaney ML and Scheinberg DA: Inhibition of human mitochondrial peptide deformylase causes apoptosis in c-myc-overexpressing hematopoietic cancers. Cell Death Dis. 5:e11522014. View Article : Google Scholar : PubMed/NCBI

41 

Gómez-Casares MT, García-Alegria E, López-Jorge CE, Ferrándiz N, Blanco R, Alvarez S, Vaqué JP, Bretones G, Caraballo JM, Sánchez-Bailón P, et al: MYC antagonizes the differentiation induced by imatinib in chronic myeloid leukemia cells through downregulation of p27(KIP1). Oncogene. 32:2239–2246. 2013. View Article : Google Scholar

42 

Singh AM and Dalton S: The cell cycle and Myc intersect with mechanisms that regulate pluripotency and reprogramming. Cell Stem Cell. 5:141–149. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Hoffman B, Amanullah A, Shafarenko M and Liebermann DA: The proto-oncogene c-myc in hematopoietic development and leukemogenesis. Oncogene. 21:3414–3421. 2002. View Article : Google Scholar : PubMed/NCBI

44 

DeLuca HF: Evolution of our understanding of vitamin D. Nutr Rev. 66(Suppl 2): S73–S87. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Kasukabe T, Okabe-Kado J, Kato N, Sassa T and Honma Y: Effects of combined treatment with rapamycin and cotylenin A, a novel differentiation-inducing agent, on human breast carcinoma MCF-7 cells and xenografts. Breast Cancer Res. 7:R1097–R1110. 2005. View Article : Google Scholar

Related Articles

Journal Cover

August-2015
Volume 47 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Maniwa Y, Kasukabe T and Kumakura S: Vitamin K2 and cotylenin A synergistically induce monocytic differentiation and growth arrest along with the suppression of c-MYC expression and induction of cyclin G2 expression in human leukemia HL-60 cells. Int J Oncol 47: 473-480, 2015
APA
Maniwa, Y., Kasukabe, T., & Kumakura, S. (2015). Vitamin K2 and cotylenin A synergistically induce monocytic differentiation and growth arrest along with the suppression of c-MYC expression and induction of cyclin G2 expression in human leukemia HL-60 cells. International Journal of Oncology, 47, 473-480. https://doi.org/10.3892/ijo.2015.3028
MLA
Maniwa, Y., Kasukabe, T., Kumakura, S."Vitamin K2 and cotylenin A synergistically induce monocytic differentiation and growth arrest along with the suppression of c-MYC expression and induction of cyclin G2 expression in human leukemia HL-60 cells". International Journal of Oncology 47.2 (2015): 473-480.
Chicago
Maniwa, Y., Kasukabe, T., Kumakura, S."Vitamin K2 and cotylenin A synergistically induce monocytic differentiation and growth arrest along with the suppression of c-MYC expression and induction of cyclin G2 expression in human leukemia HL-60 cells". International Journal of Oncology 47, no. 2 (2015): 473-480. https://doi.org/10.3892/ijo.2015.3028