OBP‑801, a novel histone deacetylase inhibitor, induces M‑phase arrest and apoptosis in rhabdomyosarcoma cells

  • Authors:
    • Chihiro Tomoyasu
    • Ken Kikuchi
    • Daisuke Kaneda
    • Shigeki Yagyu
    • Mitsuru Miyachi
    • Kunihiko Tsuchiya
    • Tomoko Iehara
    • Toshiyuki Sakai
    • Hajime Hosoi
  • View Affiliations

  • Published online on: October 22, 2018     https://doi.org/10.3892/or.2018.6813
  • Pages: 643-649
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Rhabdomyosarcoma (RMS) is an aggressive pediatric cancer of musculoskeletal origin. Despite multidisciplinary approaches, such as surgical resection, irradiation, and intensive chemotherapy, adopted for its treatment, the prognosis of patients with high‑risk RMS remains poor. Thus, molecularly targeted therapies are required to improve patient survival and minimize side effects. Histone deacetylases (HDACs) modify transcription by deacetylation of the lysine residues in chromatin histone tails and several non‑histone proteins. HDAC inhibitors, classes of compounds targeted to various HDAC proteins, are being studied for their roles in several types of cancers in a rigorous manner. This study aimed to investigate the potential of a novel HDAC inhibitor, OBP‑801, as a therapeutic agent for the treatment of RMS. We used 8 RMS cell lines in this study. Protein expression patterns, cell proliferation, cell cycle status, and apoptosis in RMS cells after OBP‑801 treatment in vitro were investigated. We also studied the antitumor activity of OBP‑801 in an in vivo xenograft mouse model. We observed cell cycle arrest at the M‑phase and apoptosis in all RMS cell lines after exposure to pharmacological levels of OBP‑801 for 24 h. Immunofluorescence staining revealed that OBP‑801 may induce mitotic catastrophe via chromosome misalignment and reduced survivin expression, ultimately leading to apoptosis. Our results demonstrated that the novel HDAC inhibitor OBP‑801 was an effective inhibitor of RMS cell line proliferation and may be a potent therapeutic option for RMS.

Introduction

Rhabdomyosarcoma (RMS) constitutes ~60% of all childhood soft tissue sarcomas. It is a small, round-cell tumor of musculoskeletal origin that exhibits various degrees of myogenic differentiation. Most types of RMS fall into 1 of 2 biological distinction subgroups defined as alveolar or embryonal type (1). Alveolar RMS (ARMS), which accounts for 41% of all RMS, is an aggressive soft tissue sarcoma in children, with a high invasion and metastasis at initial diagnosis. Despite a combination therapy, which includes surgical resection, radiation, and intensive chemotherapy, the prognosis for patients with ARMS remains poor (2). Recently, many molecular targeted drugs have been effectively used in patients with different types of tumors; however, for RMS, targeted drugs are still under investigation and have not been used clinically (35).

Histone deacetylase (HDAC) inhibitors are promising drugs for the treatment of diverse cancers due to their abilities to induce cell cycle control and apoptosis (69). Numerous of HDAC inhibitors are currently being assessed in various phases of clinical trials (10,11). The United States Food and Drug Administration (US FDA) has already approved the HDAC inhibitors belinostat, vorinostat, and romidepsin for the treatment of peripheral and cutaneous T cell lymphoma (1214). There have been a few studies on the tumor suppressive effects of RMS by HDAC inhibition in vitro (15,16). A recent study in an ARMS mouse model revealed that response to the HDAC inhibitor was different depending on the myogenic lineage of the tumor cells (17). Presently, the role of HDAC activity in RMS tumorigenesis and the essential mechanisms by which HDAC inhibitors exhibit their antitumor effects remain largely unknown.

OBP-801 (spiruchostatin A) was originally identified as an enhancer of PAI-1 gene expression and was established as a new HDAC inhibitor by a p21 promoter reporter screen (18). OBP-801 exerted the most potent HDAC-inhibitory activity in our study; it was ~50 times more effective than vorinostat (18). It is currently under clinical trials in the United States.

The aim of this study was to evaluate the effect of the new HDAC inhibitor, OBP-801, on the cell cycle control and on the viability of RMS cells in vitro and in vivo using a mouse tumor model. We also assessed the mechanism of action of OBP-801.

Materials and methods

Cell lines and reagents

We used the human ARMS cell lines SJ-Rh30 (Rh30), SJ-Rh41 (Rh41), SJ-Rh3 (Rh3), SJ-Rh4 (Rh4), SJ-Rh18 (Rh18) and SJ-Rh28 (Rh28) that were kindly provided by Peter J. Houghton M, San Antonio, TX, USA), and the embryonal RMS (ERMS) cell lines RD that was obtained from JCRB (Japanese Collection of Research Bioresources) Cell Bank and RMS-YM that was kindly provided by Naoki Kakazu M.D. (Department of Environmental and Preventive Medicine, Shimane University School of Medicine, Shimane, Japan) (5,19). They were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) at 37°C in a 5% CO2 incubator. OBP-801 (Oncolys BioPharma, Inc., Tokyo, Japan) was dissolved in dimethyl sulfoxide (DMSO) and stored as a 1-mM stock solution in 50-µl aliquots at −20°C. The percentage of DMSO in all experiments was <0.01%.

Western blot analysis

Cells were lysed in RIPA buffer (08714-04; Nacalai Tesque, Kyoto, Japan). Protein concentrations in cell lysates were determined using BCA assay. A total of 20 µg of protein were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) on NuPAGE Novex 4–12% Bis-Tris gels in NuPAGE MES SDS running buffer (Life Technologies; Thermo Fisher Scientific, Inc.). Proteins were subsequently transferred to Immobilon-P membranes in NuPAGE Transfer buffer (Life Technologies; Thermo Fisher Scientific, Inc). Membranes were blocked in phosphate-buffered saline with Tween-20 (PBST) containing 5% non-fat dry milk powder, and then incubated at 4°C overnight with primary antibodies against the following proteins: anti-histone-H4 (dilution 1:1,000; cat. no. 13919; Cell Signaling Technology, Inc., Danvers, MA, USA), anti-acetyl histone-H4 (dilution 1:1,000; cat. no. 06–866; Merck KGaA, Darmstadt, Germany), anti-Rb (dilution 1:2,000; cat. no. 554136; BD Biosciences, Franklin Lakes, NJ, USA), anti-phospho-histone-H3 (dilution 1:2,000; cat. no. 3377; Cell Signaling Technology, Inc.), anti-survivin (dilution 1:2,000; cat. no. 2808; Cell Signaling Technology, Inc.) and anti-p21 Waf1/Cip1 (1:2,000; cat. no. 2946; Cell Signaling Technology, Inc.), anti-total caspase-3 (dilution 1:2,000; cat. no. 610322; BD Biosciences), anti-cleaved caspase-3 (dilution 1:1,000; cat. no. 9664; Cell Signaling Technology, Inc.) and anti-β-actin (dilution 1:5,000; cat. no. A2228; Merck KGaA) for the reference protein. The membranes were then washed with PBST and incubated at room temperature for 1 h with sheep anti-mouse secondary antibody (dilution 1:10,000; cat. no. NA931; GE Healthcare, Little Chalfont, UK) or donkey anti-rabbit secondary antibody (dilution 1:10,000; cat. no. NA934; GE Healthcare). Antibody binding was detected using the enhanced chemiluminescence detection system (ECL and ECL prime; GE Healthcare) (20). The protein density of western blot was measured by ImageJ 1.52a (National Institutes of Health, Bethesda, MD, USA).

WST-8 cell viability assay

We performed WST-8 colorimetric assays with Cell Counting Kit-8 (Nacalai Tesque, Inc.) according to the manufacturer's instructions. We seeded the cells in 96-well plates in 100 µl culture medium for 24 h, then added various reagents. We determined cell viability by assessing the optical density (OD) at 450 nm with a microplate reader (Multiskan™ JX; Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan), as previously described (20).

Cell cycle analysis

To analyze their cell cycle distribution, we cultured the cells in the presence of OBP-801 or an equivalent volume of DMSO for 24 h. We then isolated the cells by scraping, washed them with phosphate-buffered saline (PBS), and incubated them with propidium iodide at a concentration of 50 µg/ml for 30 min to stain DNA. We determined their DNA content on a FACSCalibur™ flow cytometer (BD Biosciences). We analyzed their cell cycle status with FlowJo software 7.6.5 (Tree Star, Inc., Ashland, OR, USA), as previously described (21).

Analysis of apoptosis by flow cytometry

We analyzed cell death after Annexin V-FITC and propidium iodide staining with a TACS Annexin V-FITC Apoptosis Detection kit (R&D Systems, Inc., Minneapolis, MN, USA), according to the manufacturer's instructions. We analyzed the data with FlowJo software (Tree Star, Inc.) as previously described (21).

Immunocytochemistry

We plated the cells on Falcon® 8-Well Culture Slides (cat. no. 354118; BD Falcon; Corning, Inc., Corning, NY, USA), then fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton™ X-100, washed with PBS, and incubated with anti-survivin (dilution 1:1,000; cat. no. 2808), anti-α-tubulin (dilution 1:1,000; cat. no. 3873), anti-phospho-histone-H3 (dilution 1:1,000; cat. no. 3377), or anti-phospho-H2AX (dilution 1:1,000; cat. no. 9718; all from Cell Signaling Technology, Inc.) at 4°C overnight. We then rinsed the slides with PBS and incubated them with Alexa Fluor 488/555-conjugated anti-mouse/rabbit IgG (dilution 1:200; cat. nos. A11008, A11001 and A21422; Cell Signaling Technology, Inc.) at room temperature for 1 h. Finally, we examined the slides by fluorescence microscopy with a KEYENCE BZ-X700 instrument (Keyence Corp., Osaka, Japan).

In vivo mouse xenograft studies

Female BALB/c nu/nu nude mice (4-weeks old, total 14, 11–16 g) were purchased from Japan SLC, Inc. (Shizuoka, Japan). All experiments and procedures were conducted in accordance with the institutional animal care and use committee guidelines. The present study was also approved by the Committee for Animal Research of Kyoto Prefectural University of Medicine (permission no. M27-477). Mice were maintained at 23±2°C under a 12-h light/dark cycle (light period, 07:00-19:00 h). Food and water were available ad libitum. We subcutaneously injected 1×107 luciferase-positive Rh30 cells into the dorsal area of BALB/c nu/nu nude mice (n=3 and 4/group). We monitored tumor growth in live mice by bioluminescent detection of the luciferase activity of the Rh30 cells at days 3 and 52, as previously described (22). We assessed the tumor sizes twice per week using calipers using the formula (a × b2)/2. The mice were euthanized by barbiturate overdose.

Optical imaging for luminescence

We performed in vivo bioluminescence imaging of live mice using a Xenogen IVIS®-Illumina system (PerkinElmer, Inc., Waltham, MA, USA). The animals were maintained under inhaled anesthesia (2% isoflurane in 100% oxygen at the rate of 2.5 l/min). For imaging of the firefly luciferase reporter harbored by the tumor cells, we administered a single luciferin dose of 150 mg/kg (PerkinElmer, Inc.) via intraperitoneal injection 20 min prior to imaging. The data were acquired and analyzed using the manufacturer's proprietary Living Image software 4.4 (PerkinElmer, Inc.).

Statistical analysis

Average values were expressed as the mean ± standard deviation (SD). We used the 2-tailed Student's t-test for comparison of the means between groups. Differences with a P-value of <0.05 were considered to indicate a statistically significant difference.

Results

OBP-801 inhibits the growth of RMS cell lines

We examined the effect of the HDAC inhibitor OBP-801 on the growth of ERMS and ARMS cell lines. The mean half maximal inhibitory concentration (IC50) values were 2.6±0.2 nM for the ERMS cells and 1.8±0.8 nM for the ARMS cells (Table I). OBP-801 inhibited the growth of RD and Rh30 in a concentration- and time-dependent manner (Fig. 1A and B).

Table I.

The half maximal inhibitory concentrations of OBP-801 in the various RMS cell lines.

Table I.

The half maximal inhibitory concentrations of OBP-801 in the various RMS cell lines.

Cell lineTumor typeIC50 (nM)
RDERMS2.5±1.4
RMS-YMERMS2.7±0.3
Rh30ARMS1.5±0.8
Rh41ARMS2.5±0.9
Rh3ARMS0.7±0.4
Rh4ARMS2.2±1.0
Rh18ARMS2.6±0.8
Rh28ARMS0.9±0.7

[i] The experiment was performed in triplicate and the half maximal inhibitory concentrations (IC50) are shown as the mean ± SD (n=3). RMS, rhabdomyosarcoma.

OBP-801 induces cell cycle arrest and apoptosis in RMS cell lines

To evaluate the extent of HDAC inhibition in RMS cells, we performed immunoblot analyses using anti-acetylated histone antibodies. After 24 h, OBP-801 induced the accumulation of acetylated histone H4 in a concentration-dependent fashion (Fig. 2A). OBP-801 also induced p21waf1/Cip1 in a concentration-dependent manner, and hypophosphorylation of Rb in ARMS, but not in ERMS cells (Fig. 2A). We observed that OBP-801 (10 nM, 24 h) induced arrest in the G1 and G2/M-phases in ARMS cells and in the G2/M-phase in ERMS cells (Fig. 2B). In addition, OBP-801 induced cell death in RMS cells (Fig. 1B). OBP-801 (10 nM) induced early and late apoptosis in RMS cells 48 h after the treatment, as indicated by Annexin V staining assessed by flow cytometry (Fig. 2C). Treatment with OBP-801 also led to the expression of cleaved caspase-3 in a concentration-dependent manner (Fig. 2D).

OBP-801 causes RMS cell death via mitotic catastrophe

To examine if OBP-801 caused G2- or M-phase arrest, we immunoblotted with an antibody against phospho-histone H3, a marker of mitosis. Cells treated with OBP-801 had higher phosphorylation levels of histone H3 than control cells 24 h after the treatment; the effect was concentration-dependent (Fig. 3A). We next analyzed the nuclear morphology of RMS cells with OBP-801 treatment. We found that control cells exhibited normal chromosome distribution in metaphase, with correctly formed mitotic spindles. However, upon treatment with OBP-801, dividing RMS cells exhibited aberrant metaphase morphologies: their chromosomes were not aligned at the metaphase plate, they had an abnormal mitotic spindle distribution, survivin was not recruited to the centromeres, and exhibited lower levels of survivin than that in control cells (Fig. 3B and C); OBP-801 affected the abundance of survivin in a dose-dependent manner (Fig. 3A). In addition, OBP-801-treated RMS cells were stained with the γH2AX antibody, which indicated that they entered mitosis with damaged DNA (Fig. 3C).

OBP-801 inhibits ARMS tumor growth in vivo and improves the survival of tumor-bearing mice

We measured the bioluminescence from Rh30 cells in nude mice at days 3 and 52 after injection and found that the mice that had received OBP-801 had lower tumor-related bioluminescence intensities at day 52 (Fig. 4A). The mice in the drug-treated group had smaller tumors and survived longer than those in the control group (Fig. 4C and D). We did not observe statistically significant differences between the body weights of the mice in the drug-treated and control groups, indicating that the drug did not cause toxicity (Fig. 4B).

Discussion

We used a new HDAC inhibitor, OBP-801, as a potential therapeutic drug for treating RMS. HDAC inhibitor-induced cell death is tumor-selective, as previously described (23,24). The IC50 values of OBP-801 that we observed for ERMS cells were very low compared with the concentration required to induce notable toxic effects in normal human fibroblasts (30 µM) (18). The lack of apparent cytotoxic effects in our mouse tumor model supports the tumor-selectivity of OBP-801.

The cyclin-dependent kinase inhibitor, p21, a regulator of cellular proliferation via G1 arrest, is a crucial target for HDAC inhibitors (25,26). In this study, we confirmed that OBP-801 increased the protein level of p21 Waf1/Cip1 in a concentration-dependent manner in RMS cells, possibly via the induction of p21 mRNA. Notably, however, M-phase arrest was predominant than G1- and G2-phase arrest. These findings indicated that OBP-801 may induce arrest of M-phase followed by cell death in RMS cells.

Based on our observation that OBP-801 induced cell arrest of M-phase followed by apoptosis in RMS cells, we investigated whether apoptosis was associated with mitotic catastrophe. Mitotic catastrophe is a regulated antitumor mechanism that disrupts the survival and/or proliferation of cells that are unable to undergo complete mitosis due to unrecoverable DNA damage and that have mitotic machinery problems, and/or have a failure at mitotic checkpoints (2729). Recently, relationships were reported between HDACs and mitotic arrest/catastrophe via DNA damage (30) and between Aurora B kinase activity (31) and Eg5 acetylation (32). Mitotic catastrophe is assessed based on unique morphological nuclear changes, such as multinucleation, macronucleation, and micronucleation. We found that OBP-801 treatment of dividing RMS cells induced γH2AX-positive aberrant metaphase morphologies, concurrent with lower levels and misalignment of survivin. Recently, Aljaberi et al reported that the activity of survivin in mitosis was regulated by cyclical acetylation and deacetylation during mitosis (33), which may explain how an HDAC inhibitor could induce mitotic arrest via survivin in a manner consistent with our results. These results indicated that OBP-801 may induce apoptosis in RMS cells via mitotic catastrophe.

In conclusion, to the best of our knowledge, this is the first study revealing that treatment of RMS cells with the novel HDAC inhibitor OBP-801 induced M-phase arrest followed by apoptosis via mitotic catastrophe. These results indicated that OBP-801 may be promising for the treatment of RMS. We believe that this drug should be chosen for future clinical trials.

Acknowledgements

The authors acknowledge Peter J. Houghton M.D. (Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio) for the cell lines (Rh30, Rh41, Rh3, Rh4, Rh18 and Rh28) and Naoki Kakazu M.D. (Department of Environmental and Preventive Medicine, Shimane University School of Medicine, Shimane, Japan) for cell line RMS-YM.

Funding

The present study was supported by JSPS KAKENHI grant no. JP26461591, JSPS KAKENHI grant no. JP25253095, and the Health Labour Sciences Research grant no. 17ck0106333 h.

Availability of data and materials

All the data supporting the conclusions of this article are included in the article.

Authors' contributions

CT, KK, TI and HH conceived and designed this study. Experimental methodology was developed by KK, SY, MM, KT, TI, TS and HH. Experimental procedures were carried out by CT, DK and KK. All authors participated in the analysis and interpretation of data. CT and KK performed the statistical analysis and wrote the 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

The present study was approved by the Committee for Animal Research of Kyoto Prefectural University of Medicine (permission no. M27-477).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

RMS

rhabdomyosarcoma

ARMS

alveolar RMS

HDAC

histone deacetylase

ERMS

embryonal RMS

DMSO

dimethyl sulfoxide

PBS

phosphate-buffered saline

SD

standard deviation

IC50

half maximal inhibitory concentration

References

1 

Merlino G and Helman LJ: Rhabdomyosarcoma-working out the pathways. Oncogene. 18:5340–5348. 1999. View Article : Google Scholar : PubMed/NCBI

2 

Davicioni E, Anderson MJ, Finckenstein FG, Lynch JC, Qualman SJ, Shimada H, Schofield DE, Buckley JD, Meyer WH, Sorensen PH, et al: Molecular classification of rhabdomyosarcoma-genotypic and phenotypic determinants of diagnosis: A report from the Children's Oncology Group. Am J Pathol. 174:550–564. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Kikuchi K, Soundararajan A, Zarzabal LA, Weems CR, Nelon LD, Hampton ST, Michalek JE, Rubin BP, Fields AP and Keller C: Protein kinase C iota as a therapeutic target in alveolar rhabdomyosarcoma. Oncogene. 32:286–295. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Miyachi M, Kakazu N, Yagyu S, Katsumi Y, Tsubai-Shimizu S, Kikuchi K, Tsuchiya K, Iehara T and Hosoi H: Restoration of p53 pathway by nutlin-3 induces cell cycle arrest and apoptosis in human rhabdomyosarcoma cells. Clin Cancer Res. 15:4077–4084. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Sokolowski E, Turina CB, Kikuchi K, Langenau DM and Keller C: Proof-of-concept rare cancers in drug development: The case for rhabdomyosarcoma. Oncogene. 33:18772014. View Article : Google Scholar : PubMed/NCBI

6 

Minucci S and Pelicci PG: Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer. 6:38–51. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Bolden JE, Peart MJ and Johnstone RW: Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 5:769–784. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Khabele D, Son DS, Parl AK, Goldberg GL, Augenlicht LH, Mariadason JM and Montgomery RV: Drug-induced inactivation or gene silencing of class I histone deacetylases suppresses ovarian cancer cell growth: implications for therapy. Cancer Biol Ther. 6:795–801. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Strait KA, Warnick CT, Ford CD, Dabbas B, Hammond EH and Ilstrup SJ: Histone deacetylase inhibitors induce G2-checkpoint arrest and apoptosis in cisplatinum-resistant ovarian cancer cells associated with overexpression of the Bcl-2-related protein Bad. Mol Cancer Ther. 4:603–611. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Khan O and La Thangue NB: HDAC inhibitors in cancer biology: Emerging mechanisms and clinical applications. Immunol Cell Biol. 90:85–94. 2012. View Article : Google Scholar : PubMed/NCBI

11 

West AC and Johnstone RW: New and emerging HDAC inhibitors for cancer treatment. J Clin Invest. 124:30–39. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Porcu P and Wong HK: We should have a dream: Unlocking the workings of the genome in cutaneous T-cell lymphomas. Clin Lymphoma Myeloma. 9:409–411. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Mann BS, Johnson JR, Cohen MH, Justice R and Pazdur R: FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 12:1247–1252. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Jain S and Zain J: Romidepsin in the treatment of cutaneous T-cell lymphoma. J Blood Med. 2:37–47. 2011.PubMed/NCBI

15 

Blattmann C, Oertel S, Ehemann V, Thiemann M, Huber PE, Bischof M, Witt O, Deubzer HE, Kulozik AE, Debus J, et al: Enhancement of radiation response in osteosarcoma and rhabdomyosarcoma cell lines by histone deacetylase inhibition. Int J Radiat Oncol Biol Phys. 78:237–245. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Kutko MC, Glick RD, Butler LM, Coffey DC, Rifkind RA, Marks PA, Richon VM and LaQuaglia MP: Histone deacetylase inhibitors induce growth suppression and cell death in human rhabdomyosarcoma in vitro. Clin Cancer Res. 9:5749–5755. 2003.PubMed/NCBI

17 

Abraham J, Nuñez-Álvarez Y, Hettmer S, Carrió E, Chen HI, Nishijo K, Huang ET, Prajapati SI, Walker RL, Davis S, et al: Lineage of origin in rhabdomyosarcoma informs pharmacological response. Genes Dev. 28:1578–1591. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Shindoh N, Mori M, Terada Y, Oda K, Amino N, Kita A, Taniguchi M, Sohda KY, Nagai K, Sowa Y, et al: YM753, a novel histone deacetylase inhibitor, exhibits antitumor activity with selective, sustained accumulation of acetylated histones in tumors in the WiDr xenograft model. Int J Oncol. 32:545–555. 2008.PubMed/NCBI

19 

Kubo K, Naoe T, Utsumi KR, Ishiguro Y, Ueda K, Shiku H and Yamada K: Cytogenetic and cellular characteristics of a human embryonal rhabdomyosarcoma cell line, RMS-YM. Br J Cancer. 63:879–884. 1991. View Article : Google Scholar : PubMed/NCBI

20 

Kikuchi K, Tsuchiya K, Otabe O, Gotoh T, Tamura S, Katsumi Y, Yagyu S, Tsubai-Shimizu S, Miyachi M, Iehara T, et al: Effects of PAX3-FKHR on malignant phenotypes in alveolar rhabdomyosarcoma. Biochem Biophys Res Commun. 365:568–574. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Kikuchi K, Hettmer S, Aslam MI, Michalek JE, Laub W, Wilky BA, Loeb DM, Rubin BP, Wagers AJ and Keller C: Cell-cycle dependent expression of a translocation-mediated fusion oncogene mediates checkpoint adaptation in rhabdomyosarcoma. PLoS Genet. 10:e10041072014. View Article : Google Scholar : PubMed/NCBI

22 

Nishijo K, Hosoyama T, Bjornson CR, Schaffer BS, Prajapati SI, Bahadur AN, Hansen MS, Blandford MC, McCleish AT, Rubin BP, et al: Biomarker system for studying muscle, stem cells, and cancer in vivo. FASEB J. 23:2681–2690. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Marks PA, Richon VM, Miller T and Kelly WK: Histone deacetylase inhibitors. Adv Cancer Res. 91:137–168. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Ungerstedt JS, Sowa Y, Xu WS, Shao Y, Dokmanovic M, Perez G, Ngo L, Holmgren A, Jiang X and Marks PA: Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc Natl Acad Sci USA. 102:673–678. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Zupkovitz G, Grausenburger R, Brunmeir R, Senese S, Tischler J, Jurkin J, Rembold M, Meunier D, Egger G, Lagger S, et al: The cyclin-dependent kinase inhibitor p21 is a crucial target for histone deacetylase 1 as a regulator of cellular proliferation. Mol Cell Biol. 30:1171–1181. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Bose P, Dai Y and Grant S: Histone deacetylase inhibitor (HDACI) mechanisms of action: Emerging insights. Pharmacol Ther. 143:323–336. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25:486–551. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R and Kroemer G: Cell death by mitotic catastrophe: A molecular definition. Oncogene. 23:2825–2837. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Vitale I, Galluzzi L, Castedo M and Kroemer G: Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol. 12:385–392. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Cornago M, Garcia-Alberich C, Blasco-Angulo N, Vall-Llaura N, Nager M, Herreros J, Comella JX, Sanchis D and Llovera M: Histone deacetylase inhibitors promote glioma cell death by G2 checkpoint abrogation leading to mitotic catastrophe. Cell Death Dis. 5:e14352014. View Article : Google Scholar : PubMed/NCBI

31 

Li Y, Kao GD, Garcia BA, Shabanowitz J, Hunt DF, Qin J, Phelan C and Lazar MA: A novel histone deacetylase pathway regulates mitosis by modulating Aurora B kinase activity. Genes Dev. 20:2566–2579. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Nalawansha DA, Gomes ID, Wambua MK and Pflum MKH: HDAC inhibitor-induced mitotic arrest ss mediated by Eg5/KIF11 acetylation. Cell Chem Biol. 24:481–492. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Aljaberi AM, Webster JR and Wheatley SP: Mitotic activity of survivin is regulated by acetylation at K129. Cell Cycle. 14:1738–1747. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 41 Issue 1

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
Tomoyasu C, Kikuchi K, Kaneda D, Yagyu S, Miyachi M, Tsuchiya K, Iehara T, Sakai T and Hosoi H: OBP‑801, a novel histone deacetylase inhibitor, induces M‑phase arrest and apoptosis in rhabdomyosarcoma cells. Oncol Rep 41: 643-649, 2019
APA
Tomoyasu, C., Kikuchi, K., Kaneda, D., Yagyu, S., Miyachi, M., Tsuchiya, K. ... Hosoi, H. (2019). OBP‑801, a novel histone deacetylase inhibitor, induces M‑phase arrest and apoptosis in rhabdomyosarcoma cells. Oncology Reports, 41, 643-649. https://doi.org/10.3892/or.2018.6813
MLA
Tomoyasu, C., Kikuchi, K., Kaneda, D., Yagyu, S., Miyachi, M., Tsuchiya, K., Iehara, T., Sakai, T., Hosoi, H."OBP‑801, a novel histone deacetylase inhibitor, induces M‑phase arrest and apoptosis in rhabdomyosarcoma cells". Oncology Reports 41.1 (2019): 643-649.
Chicago
Tomoyasu, C., Kikuchi, K., Kaneda, D., Yagyu, S., Miyachi, M., Tsuchiya, K., Iehara, T., Sakai, T., Hosoi, H."OBP‑801, a novel histone deacetylase inhibitor, induces M‑phase arrest and apoptosis in rhabdomyosarcoma cells". Oncology Reports 41, no. 1 (2019): 643-649. https://doi.org/10.3892/or.2018.6813