Open Access

Antitumor activity of dobutamine on human osteosarcoma cells

  • Authors:
    • Jun Yin
    • Qirong Dong
    • Minqian Zheng
    • Xiaozu Xu
    • Guoyou Zou
    • Guolin Ma
    • Kefeng Li
  • View Affiliations

  • Published online on: April 20, 2016     https://doi.org/10.3892/ol.2016.4479
  • Pages: 3676-3680
  • Copyright: © Yin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Dobutamine has been widely used for the treatment of heart failure and cardiogenic shock since the 1970s. Osteosarcoma is the most commonly observed malignant bone tumor in children. Currently, there are no effective drugs for the treatment of osteosarcoma. In the present study, the potential anticancer activity of dobutamine on human osteosarcoma cells was examined. Human osteosarcoma MG‑63 cells were treated with dobutamine at various concentrations and for various incubation times. The inhibition of cell growth by dobutamine was determined by 3‑(4,5‑dimethylthiazol‑2‑yl)-2,5‑diphenyltetrazolium bromide assay. Flow cytometry was utilized to evaluate the effect of dobutamine on cell apoptosis and the cell cycle. Furthermore, the expression levels of caspase‑3 and caspase‑9 were assessed by western blot analysis. The influence of dobutamine on cancer cell migration and invasion was additionally evaluated using wound‑healing assay and the Boyden Chamber migration method. Dobutamine significantly inhibited the growth of MG‑63 cells at a concentration of 10 µM or higher when incubated for 12 h or longer (P=0.023). Dobutamine augmented cell apoptosis and arrested the cell cycle in the G2/M phase. Western blot analysis revealed that dobutamine induces expression of caspase‑3 and caspase‑9. In addition, the invasiveness and migration of MG‑63 cells was inhibited by dobutamine in a concentration‑dependent manner. The results of the present study may lead to novel applications for dobutamine in the treatment of osteosarcoma.

Introduction

Osteosarcoma is the most commonly observed primary malignant cancer of the bone in children, and possesses high incidence and mortality rates (1). The tumor predominantly arises from the metaphyses of the long bones with active bone growth and repairation, such as the knee joint, lower femur and upper tibia. As osteosarcoma is considered to be a radioresistant tumor, chemotherapy is the primary approach for the treatment of osteosarcoma (2). However, the currently utilized chemotherapy regimens demonstrate low efficacy for the treatment of this tumor (3). Current chemotherapeutic drugs, including ifosfamide, cisplatin and high-dose methotrexate, have a number of side-effects and their use may result in acquired drug resistance in osteosarcoma cells (4). Furthermore, the prognosis of osteosarcoma is poor and >30% of patients succumb to pulmonary metastases within 5 years of diagnosis (5). Therefore, there is an urgent requirement for the development of novel effective therapeutic drugs for the treatment of osteosarcoma.

Yes-associated protein (YAP), a transcriptional co-activator, is a key regulator of the Hippo signaling pathway (6). When YAP is recruited to the nucleus, transcription of cell proliferation-promoting and anti-apoptotic genes is continuously activated (7,8). High expression of YAP has been observed in a number of types of tumor, including osteosarcoma, hepatocellular, colorectal, ovarian, breast and lung cancer cases, as well as gastric carcinoma, and has been reported to be correlated with a poor prognosis (912). These findings suggest that YAP may contribute to a malignant cellular phenotype and therefore may be an important target for anticancer drugs (13).

Dobutamine is a synthetic catecholamine developed by Eli Lilly and Company in the 1970s (14). It has been widely used as an inotropic drug for hemodynamic support in the treatment of congestive heart failure, as well as cardiogenic and septic shock (15). A previous study demonstrated that dobutamine is able to attenuate YAP-dependent transcription by inhibiting its nuclear translocation (16).

In the present study, the effect of dobutamine on the proliferation, apoptosis and invasiveness of the MG-63 human osteosarcoma cell line was investigated. The results of the present study demonstrated the potential effectiveness of dobutamine for the treatment of osteosarcoma.

Materials and methods

The MG-63 human osteosarcoma cell line was purchased from the Shanghai Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Dulbecco's Modified Eagle's Medium (DMEM) and fetal bovine serum (FBS) were obtained from GE Healthcare Life Sciences (Logan, UT, USA). Propidium iodide (PI) and dobutamine were obtained from Sigma-Aldrich (St. Louis, MO, USA). The Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit was obtained from Beckman Coulter, Inc. (Brea, CA, USA).

Cell culture

MG-63 cells were grown in medium at 37°C in an atmosphere with 5% CO2. Culture medium supplemented with 10% FBS, 100 U/ml penicillin (Gibco; Thermo Fisher Scientific Inc., Waltham, MA, USA), 100 µg/ml streptomycin (Gibco; Thermo Fisher Scientific Inc.) and DMEM was used for MG-63 culture.

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

The influence of dobutamine treatment on cell viability was determined using an MTT assay. Cells were seeded into a 96-well plate (Corning, New York, NY, USA) overnight at 37°C and incubated with various concentrations of dobutamine (1, 5, 10, 25 and 50 µM) for 12, 24, 48 and 72 h. Following the indicated treatments, the cells were incubated with MTT (0.25 mg/ml; Sigma-Aldrich) in phosphate-buffered saline (PBS; Gibco; Thermo Fisher Scientific Inc.) for 4 h at 37°C, followed by removal of the medium and addition of 1 ml 100% dimethyl sulfoxide (Beyotime Institute of Biotechnology, Shanghai, China) to solubilize the MTT-formazan product. The absorbance at 490 nm was determined using an automatic multi-well spectrophotometer (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The inhibitory rate of cell growth was calculated as [1-treatment group/control group)]x100. The growth curve was drawn using time as the abscissa and inhibition rate as the ordinate. Each dobutamine dose was used in triplicate, and the MTT assay was repeated at least twice.

Flow cytometric analysis

The rate of apoptosis and percentage of cells in G1, S and G2/M phases was measured by flow cytometry. Following treatment of the experimental groups in MTT for 24 h, cells were harvested, trypsinized, washed twice with PBS and resuspended in binding buffer (Beyotime Institute of Biotechnology). The cells were subsequently stained with Annexin V-FITC and PI according to the manufacturer's protocol and analyzed by flow cytometry. The cell suspension was incubated with 50 µg/ml PI solution and 50 U/ml RNase (Beyotime Institute of Biotechnology) for 30 min in order to observe the cell cycle stage. Flow cytometric analysis was performed on a BD FACSCaliber™ using CellQuest software, version 5.1 (BD Biosciences, Franklin Lakes, NJ, USA).

Cell invasion analysis

The effect of treatment with dobutamine on the invasion of MG-63 cells was investigated using Transwell chambers with polycarbonate filters (pore size of 8 µm; Beyotime Institute of Biotechnology). MG-63 cells were seeded into the upper chamber at a density of 1×105 cells/ml and incubated in 0.6 ml DMEM medium containing 10% FBS and various concentrations (1, 5, 10, 25 and 50 µM) of dobutamine. The lower chamber was filled with 0.6 ml DMEM medium containing 20% FBS. Following 24 h of incubation at 37°C, cells on the upper filter that had not migrated through were removed by wiping, and the remaining cells were fixed in 4% paraformaldehyde (Beyotime Institute of Biotechnology) for a total of 1 h. Cells that had migrated through the filter were stained using hematoxylin (Beyotime Institute of Biotechnology) and visualized and counted under a microscope (Olympus IX53; Olympus, Tokyo, Japan).

Western blot analysis

The present study examined the levels of protein expression of caspase-3 and caspase-9 in MG-63 cells prior to and following treatment with dobutamine. The cells were treated with the different concentrations of dobutamine (1, 5, 10, 25 and 50 µM). The cells were then harvested in 5 ml of medium, pelleted by centrifugation (1,000 × g for 5 min at 4°C), washed twice using ice-cold PBS and lysed in ice-cold HEPES buffer (50 mmol/l; pH7.5), 10 mmol/l NaCl, 5 mmol/l MgCl2, 1 mmol/l ethylenediaminetetraacetic acid (all Beyotime Institute of Biotechnology), 110% glycerol (v/v), 1% Triton X-100 (v/v 1X complete), a cocktail of SigmaFast protease inhibitors (1X complete; Sigma-Aldrich), followed by treatment with 1 mg/l dobutamine on ice for 30 min. The cell lysates were clarified by centrifugation (15,000 × g for 10 min at 4°C), and the supernatants were analyzed immediately or stored at −80°C until required. Equivalent quantities of protein (50 µg) from total cell lysates were resolved by sodium dodecyl sulfate polyacrylamide gel electrophoresis using precast 12% BIS-TRIS gradient gels and transferred onto polyvinylidene difluoride membranes. Membranes were blocked overnight at 4°C using blocking buffer [5% skimmed dried milk (v/v), 150 mmol/l NACl, 10 mmol/l Tris (pH 8.0) and 0.05% Tween 20 (v/v); Beyotime Institute of Biotechnology]. Proteins were detected by incubation in blocking buffer overnight at 4°C with the following primary antibodies: Mouse anti-human monoclonal caspase-3 antibody (1:10,00 dilution; sc-65496), mouse anti-human monoclonal caspase-9 antibody (1:10,00 dilution; sc-56073) and mouse anti-human monoclonal GADPH antibody (1:10,00 dilution; sc-47778) (all Santa Cruz Biotechnology, Inc, Dallas, TX, USA). Unbound antibody was removed by washing with Tris-buffered saline (pH7.2) containing 0.5% Tween 20 (TBS-T; Beyotime Institute of Biotechnology). The membrane was subsequently incubated at room temperature with horseradish peroxidase-conjugated secondary antibody. Subsequent to washing with TBS-T three times, bands were visualized by enhanced chemiluminescence system (Pierce Biotechnology, Rockford, IL, USA), and the protein intensities were quantified using AlphaEaseFC 4.1.0 software (Alpha Innotech, San Leandro, CA, USA).

Statistical analysis

Statistical analyses were performed using SPSS version 15.0 (SPSS Inc., Chicago, IL, USA). Comparisons between two samples (experimental and control group) were employed by Student's t-test. P<0.05 was considered to represent a statistically significant difference.

Results and Discussion

Dobutamine inhibits the proliferation of MG-63 osteosarcoma cells

The results of the present study revealed that dobutamine significantly inhibited cell proliferation in a time- and concentration-dependent manner compared with the control group. As demonstrated by the proliferation inhibition graph (Fig. 1), treatment with 10, 25 and 50 µM dobutamine had a significant inhibitory effect on the survival of MG-63 cells (P=0.032).

Dobutamine augments cell apoptosis and arrests the cell cycle

Annexin V/PI staining was used to measure dobutamine-induced apoptosis. Compared with the control group, dobutamine induced a significant increase in apoptotic death, following pretreatment of MG-63 cells with 5, 10, 25 and 50 µM for 24 h (P=0.028; Table I). The percentage of MG-63 cells in G2/M phases was significantly increased at dobutamine concentrations of 25 and 50 µM (P=0.007 and P=0.003, respectively), and the percentage of cells in S-phase was significantly decreased (P=0.039) compared with the control group (Fig. 2).

Table I.

Cell cycle phase distribution and apoptosis of MG-63 cells.

Table I.

Cell cycle phase distribution and apoptosis of MG-63 cells.

Cell cycle phase

GroupG0/G1SG2/MApoptosis, %
Control54.12±6.5325.60±5.3320.28±1.792.2±0.4
1 µM dobutamine55.19±3.1625.37±3.6619.44±5.012.5±1.1
5 µM dobutamine53.96±4.5823.18±6.3422.86±4.272.9±1.7
10 µM dobutamine49.82±2.9926.78±5.9223.40±6.527.0±2.5a
25 µM dobutamine50.62±5.2721.50±4.59 27.88±4.55b11.6±4.7c
50 µM dobutamine51.21±6.5218.39±2.79 30.40±7.26d13.2±1.8e

a P=0.012

b P=0.007

c P=0.020

d P=0.003

e P=0.026 vs. control group.

Dobutamine reduces the migration and invasion of MG-63 cells

To investigate whether dobutamine treatment affects osteosarcoma cell movement, the migratory rate of the MG-63 cells was observed. Fig. 3 demonstrates that dobutamine significantly decreased cell migration from the edge of the wound (P=0.041). Similarly, the cell invasion/Transwell assay showed that a large number of cells passed through the filter in the control group, whereas the cells passing through the filter were markedly reduced following dobutamine treatment. Furthermore, treatment with dobutamine reduced the number of invasive cells in a concentration-dependent manner (Fig. 4). The number of invasive cells in the dobutamine groups was significantly reduced compared with that in the control group (P=0.039 for 10 µM, P=0.015 for 25 µM and P=0.011 for 50 µM) (Fig. 4).

Dobutamine induces expression of caspase-3 and caspase-9

Caspases are a family of endoproteases that provide crucial links in cell regulatory networks that control inflammation and cell death (17). Caspase-3 and 9 are crucial mediators in apoptosis signaling pathways (18). Western blot analysis was used to investigate the expression of caspase-3 and caspase-9 in MG-63 cells following dobutamine treatment. Protein expression analysis indicated that caspase-3 levels were increased following treatment with dobutamine at the concentrations of 10 and 50 µM for 72 h (P=0.011 and P=0.013, respectively), and caspase-9 levels were increased following treatment with dobutamine at a concentration of 50 µM for 72 h (P=0.031) (Fig. 5). These findings indicated that dobutamine may induce cancer cell apoptosis and cell death.

Recent reports have demonstrated that YAP is highly expressed in human osteosarcoma MG-63 cells (19). The results of the present study indicate that the inhibitory effect of dobutamine may be associated with the inhibition of YAP translocation. Silencing of the YAP gene by RNA interference led to a similar effect to that caused by dobutamine (20). In addition, the present study found that dobutamine arrests the cell cycle at the G2/M transition stage and augments cell apoptosis. Previous studies have demonstrated that YAP activates cell apoptosis in response to DNA damage via interaction with p73 in several cancer cell lines (21). The findings of the present study may result in a novel application for dobutamine in the treatment of cancer.

In conclusion, the results of the present study demonstrated that dobutamine was able to significantly suppress osteosarcoma cell growth by inhibiting cell proliferation, inducing cell apoptosis and redistributing cell cycle phases. These findings indicate that dobutamine may become a novel therapeutic agent for the treatment of osteosarcoma. However, additional in vivo studies are required in order to confirm the effectiveness and safety of dobutamine in the treatment of osteosarcoma.

Acknowledgements

This study was supported by grants from the National Science and Technology Support Program (no. 2012BAI10B02) and the National Science Foundation of China (no. 81571641), and an internal grant from China-Japan Friendship Hospital, Beijing, China (no. 2014-3-MS-18).

References

1 

Basu-Roy U, Basilico C and Mansukhani A: Perspectives on cancer stem cells in osteosarcoma. Cancer Lett. 338:158–167. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Ando K, Heymann MF, Stresing V, Mori K, Rédini F and Heymann D: Current therapeutic strategies and novel approaches in osteosarcoma. Cancers (Basel). 5:591–616. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Lamplot JD, Denduluri S, Qin J, Li R, Liu X, Zhang H, Chen X, Wang N, Pratt A, Shui W, et al: The current and future therapies for human osteosarcoma. Curr Cancer Ther Rev. 9:55–77. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Luetke A, Meyers PA, Lewis I and Juergens H: Osteosarcoma treatment - where do we stand? A state of the art review. Cancer Treat Rev. 40:523–532. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Loh AH, Navid F, Wang C, Bahrami A, Wu J, Neel MD and Rao BN: Management of local recurrence of pediatric osteosarcoma following limb-sparing surgery. Ann Surg Oncol. 21:1948–1955. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Huang J, Wu S, Barrera J, Matthews K and Pan D: The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila homolog of YAP. Cell. 122:421–434. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Zhao B, Wei X, Li W, Udan RS, Yang Q, Kim J, Xie J, Ikenoue T, Yu J, Li L, et al: Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 21:2747–2761. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Steinhardt AA, Gayyed MF, Klein AP, Dong J, Maitra A, Pan D, Montgomery EA and Anders RA: Expression of Yes-associated protein in common solid tumors. Hum Pathol. 39:1582–1589. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Zender L, Spector MS, Xue W, Flemming P, Cordon-Cardo C, Silke J, Fan ST, Luk JM, Wigler M, Hannon GJ, et al: Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell. 125:1253–1267. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Overholtzer M, Zhang J, Smolen GA, Muir B, Li W, Sgroi DC, Deng CX, Brugge JS and Haber DA: Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc Natl Acad Sci USA. 103:12405–12410. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Roden R and Wu TC: How will HPV vaccines affect cervical cancer? Nat Rev Cancer. 6:753–763. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Castle PE, Dockter J, Giachetti C, Garcia FA, McCormick MK, Mitchell AL, Holladay EB and Kolk DP: A cross-sectional study of a prototype carcinogenic human papillomavirus E6/E7 messenger RNA assay for detection of cervical precancer and cancer. Clin Cancer Res. 13:2599–2605. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Molden T, Kraus I, Karlsen F, Skomedal H and Hagmar B: Human papillomavirus E6/E7 mRNA expression in women younger than 30 years of age. Gynecol Oncol. 100:95–100. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Tuttle RR and Mills J: Dobutamine: Development of a new catecholamine to selectively increase cardiac contractility. Circ Res. 36:185–196. 1975. View Article : Google Scholar : PubMed/NCBI

15 

Roesslein M, Froehlich C, Jans F, Piegeler T, Goebel U and Loop T: Dobutamine mediates cytoprotection by induction of heat shock protein 70 in vitro. Life Sci. 98:88–95. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Bao Y, Nakagawa K, Yang Z, Ikeda M, Withanage K, Ishigami-Yuasa M, Okuno Y, Hata S, Nishina H and Hata Y: A cell-based assay to screen stimulators of the Hippo pathway reveals the inhibitory effect of dobutamine on the YAP-dependent gene transcription. J Biochem. 150:199–208. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Brentnall M, Rodriguez-Menocal L, De Guevara RL, Cepero E and Boise LH: Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 14:322013. View Article : Google Scholar : PubMed/NCBI

18 

Fujita E, Egashira J, Urase K, Kuida K and Momoi T: Caspase-9 processing by caspase-3 via a feedback amplification loop in vivo. Cell Death Differ. 8:335–344. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Zhang YH, Li B, Shen L, Shen Y and Chen XD: The role and clinical significance of YES-associated protein 1 in human osteosarcoma. Int J Immunopathol Pharmacol. 26:157–167. 2013.PubMed/NCBI

20 

Zhou Z, Zhu JS and Xu ZP: RNA interference mediated YAP gene silencing inhibits invasion and metastasis of human gastric cancer cell line SGC-7901. Hepatogastroenterology. 58:2156–2161. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Lapi E, Di Agostino S, Donzelli S, Gal H, Domany E, Rechavi G, Pandolfi PP, Givol D, Strano S, Lu X and Blandino G: PML, YAP, and p73 are components of a proapoptotic autoregulatory feedback loop. Mol Cell. 32:803–814. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 11 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yin J, Dong Q, Zheng M, Xu X, Zou G, Ma G and Li K: Antitumor activity of dobutamine on human osteosarcoma cells. Oncol Lett 11: 3676-3680, 2016
APA
Yin, J., Dong, Q., Zheng, M., Xu, X., Zou, G., Ma, G., & Li, K. (2016). Antitumor activity of dobutamine on human osteosarcoma cells. Oncology Letters, 11, 3676-3680. https://doi.org/10.3892/ol.2016.4479
MLA
Yin, J., Dong, Q., Zheng, M., Xu, X., Zou, G., Ma, G., Li, K."Antitumor activity of dobutamine on human osteosarcoma cells". Oncology Letters 11.6 (2016): 3676-3680.
Chicago
Yin, J., Dong, Q., Zheng, M., Xu, X., Zou, G., Ma, G., Li, K."Antitumor activity of dobutamine on human osteosarcoma cells". Oncology Letters 11, no. 6 (2016): 3676-3680. https://doi.org/10.3892/ol.2016.4479