RBEL1 is required for osteosarcoma cell proliferation via inhibiting retinoblastoma 1

  • Authors:
    • Honghui Tang
    • Feng Ji
    • Jin Sun
    • Yue Xie
    • Yongyi Xu
    • Haitao Yue
  • View Affiliations

  • Published online on: December 10, 2015     https://doi.org/10.3892/mmr.2015.4670
  • Pages: 1275-1280
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteosarcoma is the most common type of primary malignant tumor of the bone. However, mechanisms underlying osteosarcoma cell proliferation are poorly understood. The present study shows that RBEL1, a newly identified Rab‑like GTPase, may be a key regulator of osteosarcoma cell proliferation. Knockdown of RBEL1 in osteosarcoma cells resulted in impaired colony formation and cell proliferation. Cell cycle analysis suggested that RBEL1 depletion induced G1‑S arrest in osteosarcoma cells. Furthermore, it was demonstrated that retinoblastoma 1 (Rb) was upregulated and activated following RBEL1 knockdown. In addition, Rb inhibitory downstream targets, such as cyclin A2, cyclin D1, c‑Myc and cyclin‑dependent kinase 2, were downregulated. Rb knockdown reversed RBEL1 depletion‑induced tumor suppressive effects. In conclusion, the present results suggest that RBEL1 modulates cell proliferation and G1‑S transition by inhibiting Rb in osteosarcoma. These results suggest a potential therapeutic target in osteosarcoma.

Introduction

Osteosarcoma is the most common type of malignant disease in the bone and the third most common malignant tumor in children (1). Although osteosarcoma has a low incidence rate (<10% of all tumors) (2), the disease exhibits aggressive malignant phenotypes and is associated with a high mortality rate (3). The most common location of osteosarcoma development is in the long bones of the limbs (3). Current treatment for osteosarcoma is surgery combined with chemotherapy. However, the 5-year survival rate for patients with metastasis remains <20% (4,5). Although intensive efforts have been made, little is known regarding the molecular mechanisms underling this disease.

The RAS superfamily comprises a large number of low-molecular-weight GTP-binding proteins (6). According to the degree of sequence conservation, the superfamily can be divided into five distinct families, including Ras, Rho, Rab, Sar1/Arf and Ran. Rab proteins constitute the largest subfamily, and are key regulators of membrane trafficking processes in eukaryotic cells (7). RBEL1, also termed RABL6, is a novel Rab-like GTPase of unknown function. Recent studies suggest that RBEL1 proteins are overexpressed in breast cancer cells (8) and are involved in cell growth and survival (9). However, the function of RBEL1 in the regulation of tumorigenesis and development of human osteosarcoma is unclear.

The present study investigated the biological function and underlying molecular mechanism of RBEL1 in osteosarcoma. It was demonstrated that RBEL1 participates in the regulation of osteosarcoma cell proliferation. Downregulation of RBEL1 in osteosarcoma resulted in decreased colony formation and cell proliferation rates. G1-S arrest was also observed in RBEL1-depleted cells. Furthermore, it was shown that RBEL1 knockdown activated retinoblastoma 1 (Rb) and suppressed E2F transcriptional activity. These findings demonstrate that RBEL1 is a potential oncogene and a novel Rb inhibitor in osteosarcoma.

Materials and methods

Cell culture, small interfering (si)RNA transfection and lentivirus infection

U2-OS cells and SAOS2 cells were purchased from the American Type Culture Collection (Manassas, VA, USA) and maintained in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 15% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) in a humidified atmosphere with 5% CO2 at 37°C. For siRNA transfection, siRNA targeting Rb (Cell Signaling Technology Inc., Danvers, MA, USA) and Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific Inc.) were mixed and incubated for 20 min at room temperature. Then the mixture was added to cells plated in growth medium without antibiotics. The medium was changed after 4–6 h. For lentivirus infection, lentivirus targeting two different sequences was purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China): KD1 targeting CGG CCT AAA GTA CCT TCA TAA and KD2 targeting CGG CCT AAA GTA CCT TCA TAA. Cells were plated in growth medium without antibiotics. At the time of infection, growth medium was replaced by medium containing lentivirus and polybrene (8 µg/ml; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and incubated for 12 h at 37°C.

Colony formation assay

Pretreated U2-OS cells and SAOS2 cells infected with shRBEL1-1, shRBEL-2 or scramble lentivirus were plated in 6-well plates (300 cells/well) and were maintained in DMEM supplemented with 15% FBS in a humidified atmosphere of 5% CO2 at 37°C. Growth medium was changed every two days. After 12 days, cultured cells were fixed in 4% paraformaldehyde (Santa Cruz Biotechnology, Inc.) for 2 h and stained with 0.1% crystal violet (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) for 20 min. Colonies containing more than 50 individual cells were then counted with the inverted microscope (CKX41; Olympus Corporation, Beijing, China).

Bromodeoxyuridine (BrdU) incorporation assay

Cell proliferation was determined using a Cell Proliferation ELISA kit (containing BrdU labeling solution, FixDenat and anti-BrdU-POD; Roche Diagnostics GmbH, Mannheim, Germany). Briefly, pretreated U2-OS and SAOS2 cells were plated in a 96-well plate. The following day, 10 µl/well BrdU labeling solution was added and cells were incubated for 3 h. The medium was replaced by 200 µl/well FixDenat and incubated for 30 min at room temperature. Then FixDenat was removed using 100 µl/well anti-BrdU-POD. After washing twice with washing buffer, 100 µl/well substrate solution was added. The absorbance was measured at 450 nm with the ELx808 Absorbance Reader (Bio-Tek Instruments, Inc., Winooski, VT, USA).

Cell cycle analysis

Pretreated U2-OS and SAOS2 cells were harvested and washed twice in phosphate-buffered saline (PBS). Then cells were fixed in 70% ethanol at −20°C for 4 h. Cells were washed twice with PBS and stained with propidium iodide (PI)/RNase Staining Buffer (BD Biosciences, Franklin Lakes, NJ, USA) for 15 min. Cell cycle distribution was then analyzed by flow cytometry with the FACSCalibur flow cytometer (BD Biosciences).

RNA extraction, cDNA synthesis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted using TRIzol reagent (Invitrogen, Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Reverse transcription was performed using RevertAid First Strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Real-time PCRs were conducted using an ABI Prism 7500 Detection system (Applied Biosystems, Foster City, CA, USA) and SYBR Select Master mix system (Applied Biosystems) according to the manufacturer's instructions. Briefly, 10 µl 2X SYBR Select Master Mix was mixed with primers (200 nM), cDNA template (100 ng) and RNase free water made up to 20 µl. The cycling conditions werw as follows: 50°C for 2 min and 95°C for 2 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min.

Immunoblot assay

Whole-cell extracts were prepared in radioimmunoprecipitation assay buffer supplemented with 1 mM phenylmethylsulfonyl fluoride (Beijing Solarbio Science & Technology Co., Ltd.). Total protein concentration was resolved by 10% SDS-PAGE (Beijing Solarbio Science & Technology Co., Ltd.). The proteins were then transfered to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). Immunoblot analysis was conducted with incubation overnight at 4°C with the following antibodies: Anti-RBEL1 (1:1,000; ab111866) from Abcam (Cambridge, MA, USA), anti-Rb (1:200; 554144) and anti-active Rb (1:200; 554164) from (BD Biosciences), anti-c-Myc (1:100; sc-789), anti-cyclin D1 (1:200; sc-25765), anti-cyclin A2 (1:200; sc-53234) and anti-cyclin-dependent kinase 2 (CDK2; 1:200; sc-748) from Santa Cruz Biotechnology Inc., and polyclonal rabbit anti-human tubulin (1:2,000; T2200) from (Sigma-Aldrich, St. Louis, MO, USA). Membranes were then incubated with horseradish peroxidase (HRP)-conjugated anti-rabbit immunoglobulin G secondary antibodies (1:5,000; ZB-2301; Beijing Zhongshan Golden Bridge Biotechnology Co., Ltd., Beijing, China) and was detected using the Western Chemiluminescent HRP Substrate (EMD Millipore).

Statistical analysis

All data sets were analyzed by Student's t-test. Data are presented as the mean ± standard deviation from three independent experiments. Statistical analysis was conducted using Graphpad Prism software, version 6 (GraphPad Software, Inc., San Diego, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

RBEL1 depletion in osteosarcoma cells suppresses colony formation and cell proliferation

To explore the potential role of RBEL1 in osteosarcoma, its expression in U2-OS and SAOS2 osteosarcoma cell lines was inhibited by shRNA lentivirus targeting two different sequences. Knockdown efficiency was confirmed by RT-qPCR and immunoblot analysis (Fig. 1A and B). Then RBEL1 knockdown cells and negative control cells were subjected to a colony formation assay. As shown in Fig. 1C, colony number and size were suppressed following RBEL1 knockdown. Furthermore, a BrdU incorporation assay was performed to determine whether RBEL1 depletion also had an impact on cell proliferation. In addtion, in U2-OS and SAOS2 cells RBEL1 depletion significantly inhibited BrdU incorporation suggesting suppressed proliferation (Fig. 1D). These data suggest that RBEL1 may act as a tumor suppressor in osteosarcoma.

RBEL1 depletion suppresses G1-S transition in osteosarcoma cells

The cell cycle of eukaryotic cells is conventionally divided into four phases and requires three switch-like transitions at the onset of S phase and at entry and exit of mitosis. Among them, G1-S transition is the most deregulated cell cycle control mechanism in malignant diseases (10). Thus it was hypothesized that RBEL1 may modulate G1-S transition thereby promoting cell proliferation in osteosarcoma cells. Using flow cytometry, cell cycle distribution of U2-OS and SAOS2 cells infected with RBEL1 shRNA or negative control shRNA was examined. As shown in Fig. 2, RBEL1 depletion in U2-OS cells resulted in enhanced G1-S arrest compared with negative control cells. Similar results were also shown in SAOS2 cells. These results indicate that RBEL1 may participate in G1-S transition regulation in osteosarcoma.

RBEL1 regulates Rb activity in osteosarcoma cells

Rb is the major player in cell cycle control (11,12). A recent study indicated that RBEL1 negatively regulates Rb activation (8). Therefore, it was hypothesized that RBEL1 may regulate G1-S transition by inhibiting Rb activation. The impact of RBEL1 depletion on Rb expression was analyzed. An immunoblot assay showed that following RBEL1 knockdown, osteosarcoma cells exhibited increased expression of Rb compared with control cells (Fig. 3A). As the hypophosphorylated form of Rb is the active form that suppresses cell proliferation, it was examined whether hypophosphorylated Rb was also upregulated. As expected, RBEL1 knockdown cells expressed more hypophosphorylated Rb compared with the control (Fig. 3A). It is well-known that Rb targets the E2F transcription factor to suppress its downstream oncogene expression (11,12). To investigate whether RBEL1 depletion affects E2F transciptional activity, the expression of its downstream targets, such as cyclin A2, cyclin D1, c-Myc and CDK2, were examined. In agreement with above results, the mRNA and protein expression of these oncogenes was downregulated following RBEL1 knockdown in both cell types (Fig. 3B–D). These results indicate that RBEL1 may be a key regulator of Rb and its downstream targets.

RBEL1 exerts its oncogenic effects by inhibiting Rb

The results indicated that RBEL1 modulates cell proliferation and G1-S transition, and can inhibit Rb activity. Thus it was investigated whether RBEL1 exerts its oncogenic effects by inhibiting Rb. To test this hypothesis, Rb expression was suppressed in RBEL1 knockdown cells to examine whether RBEL1 knockdown-induced tumor suppression can be rescued. The knockdown efficiency was confirmed by RT-qPCR and an immunoblot assay (Fig. 4A and B). A flow cytometry assay showed that in U2-OS and SAOS2, RBEL1 knockdown-induced G1-S arrest was decreased after Rb suppression (Fig. 4C and D). Furthermore, Rb suppression reversed colony formation capability in cells with RBEL1-knockdown (Fig. 4E). BrdU incorporation was also increased following Rb suppression (Fig. 4F). These results suggest that RBEL1 exerts its oncogenic effects by inhibiting Rb.

Discussion

Numerous factors have been implicated in tumorigenesis, such as mutations, chronic inflammation resulting from bacterial or viral infection, and prolonged exposure to radiation or oncogenic chemicals (13,14). However, the etiology of osteosarcoma remains largely unknown. Deregulated cell proliferation has been recognized as one of the hallmarks of cancer (15,16). Intensive efforts were made and numerous genetic or epigenetic events were attributed to proliferation deregulation (1722). The present study demonstrates a novel regulatory mechanism of cell proliferation in osteosarcoma cells. When RBEL1 was depleted, U2-OS and SAOS2 cells generated fewer and smaller colonies. Cell proliferation suppression and G1-S arrest was also observed in RBEL1-depleted cells. These results suggest RBEL1 may act as an oncogene in osteosarcoma.

The cell cycle is a tightly regulated process that ensures specific events take place in an orderly manner. Any fault in this regulatory network results in uncontrolled cell proliferation or cell death (23,24). The cell cycle is monitored by checkpoints that sense possible defects during DNA synthesis and chromosome segregation. Cell cycle arrest allows cells to properly repair these defects, thus preventing their transmission to the resulting daughter cells (25). Deregulated cell cycle arrest has been observed in osteosarcoma (26,27). However, the underlying molecular mechanism remains unclear. In the present study, RBEL1 was observed to participate in cell cycle control in osteosarcoma. G1-S transition is the most common deregulated cell cycle transition in cancer (23) and in the present study RBEL1 downregulation induced G1-S arrest in U2-OS and SAO2 cells.

Rb is a tumor suppressor protein that is dysfunctional in several types of cancer (28,29). One function of Rb is to prevent excessive cell growth by inhibiting cell cycle progression until a cell is ready to divide (11,30). RBEL1 was found to negatively regulate Rb activity. RBEL1 downregulation resulted in an increase in Rb expression and an increase in the hypophosphorylated form of the protein. Furthermore, E2F transcriptional activity was also reduced. Cyclin A2, cyclin D1, c-Myc and CDK2, the downstream target of E2F transcription factors, were upregulated in RBEL1-depleted cells. These proteins are widely accepted as oncogenes and cell cycle positive regulator in cancer cells. Thus, RBEL1 may promote cell proliferation and G1-S transition by inhibiting Rb activity.

In conclusion, these findings suggest a novel mechanism underlying osteosarcoma progression and cell cycle regulation. RBEL1 inhibits Rb activity to upregulated oncogenic factors, such as cyclin A2, cyclin D1, c-Myc and CDK2, therefore promoting G1-S transition and cell proliferation in osteosarcoma cells. These results suggest that RBEL1 may be a novel therapeutic target and potential biomarker for osteosarcoma. However, whether RBEL1 is upregulated in human osteosarcoma samples and how RBEL1 exerts its Rb inhibitory function remain unclear and require further investigation.

References

1 

Moore DD and Luu HH: Osteosarcoma. Cancer Treat Res. 162:65–92. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Kansara M, Teng MW, Smyth MJ and Thomas DM: Translational biology of osteosarcoma. Nat Rev Cancer. 14:722–735. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Nakamura T, Matsumine A, Matsubara T, Asamuma K, Niimi R, Uchida A and Sudo A: Retrospective analysis of metastatic sarcoma patients. Oncol Lett. 2:315–318. 2011.PubMed/NCBI

5 

Marina N, Gebhardt M, Teot L and Gorlick R: Biology and therapeutic advances for pediatric osteosarcoma. Oncologist. 9:422–441. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 3:11–22. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Stenmark H: Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol. 10:513–525. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Montalbano J, Jin W, Sheikh MS and Huang Y: RBEL1 is a novel gene that encodes a nucleocytoplasmic Ras superfamily GTP-binding protein and is overexpressed in breast cancer. J Biol Chem. 282:37640–37649. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Montalbano J, Lui K, Sheikh MS and Huang Y: Identification and characterization of RBEL1 subfamily of GTPases in the Ras superfamily involved in cell growth regulation. J Biol Chem. 284:18129–18142. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Hochegger H, Takeda S and Hunt T: Cyclin-dependent kinases and cell-cycle transitions: Does one fit all? Nat Rev Mol Cell Biol. 9:910–916. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Burkhart DL and Sage J: Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat Rev Cancer. 8:671–682. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Classon M and Harlow E: The retinoblastoma tumour suppressor in development and cancer. Nat Rev Cancer. 2:910–917. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Travis LB, Demark Wahnefried W, Allan JM, Wood ME and Ng AK: Aetiology, genetics and prevention of secondary neoplasms in adult cancer survivors. Nat Rev Clin Oncol. 10:289–301. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Barcellos-Hoff MH: Does microenvironment contribute to the etiology of estrogen receptor-negative breast cancer? Clin Cancer Res. 19:541–548. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Gupta SC, Kim JH, Prasad S and Aggarwal BB: Regulation of survival, proliferation, invasion, angiogenesis and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev. 29:405–434. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Fritz V and Fajas L: Metabolism and proliferation share common regulatory pathways in cancer cells. Oncogene. 29:4369–4377. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Whitfield ML, George LK, Grant GD and Perou CM: Common markers of proliferation. Nat Rev Cancer. 6:99–106. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Hwang HW and Mendell JT: MicroRNAs in cell proliferation, cell death and tumorigenesis. Br J Cancer. 94:776–780. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Deng H, Cheng Y, Guo Z, Zhang F, Lu X, Feng L, Wang X and Xu Z: Overexpression of CyclinA2 ameliorates hypoxia-impaired proliferation of cardiomyocytes. Exp Ther Med. 8:1513–1517. 2014.PubMed/NCBI

22 

Chen G, Wang H, Xie S, Ma J and Wang G: STAT1 negatively regulates hepatocellular carcinoma cell proliferation. Oncol Rep. 29:2303–2310. 2013.PubMed/NCBI

23 

Bertoli C, Skotheim JM and de Bruin RA: Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol. 14:518–528. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Tian Y, Wan H and Tan G: Cell cycle-related kinase in carcinogenesis. Oncol Lett. 4:601–606. 2012.PubMed/NCBI

25 

Malumbres M and Barbacid M: Cell cycle, CDKs and cancer: A changing paradigm. Nat Rev Cancer. 9:153–166. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Zhang L, Zhou Q, Zhang N, Li W, Ying M, Ding W, Yang B and He Q: E2F1 impairs all-trans retinoic acid-induced osteogenic differentiation of osteosarcoma via promoting ubiquitination-mediated degradation of RARα. Cell Cycle. 13:1277–1287. 2014. View Article : Google Scholar :

27 

Wang XH, Cai P, Wang MH and Wang Z: MicroRNA-25 promotes osteosarcoma cell proliferation by targeting the cell cycle inhibitor p27. Mol Med Rep. 10:855–859. 2014.PubMed/NCBI

28 

Dick FA and Rubin SM: Molecular mechanisms underlying RB protein function. Nat Rev Mol Cell Biol. 14:297–306. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Chen HZ, Tsai SY and Leone G: Emerging roles of E2Fs in cancer: An exit from cell cycle control. Nat Rev Cancer. 9:785–797. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Prior IA, Lewis PD and Mattos C: A comprehensive survey of Ras mutations in cancer. Cancer Res. 72:2457–2467. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 13 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tang H, Ji F, Sun J, Xie Y, Xu Y and Yue H: RBEL1 is required for osteosarcoma cell proliferation via inhibiting retinoblastoma 1. Mol Med Rep 13: 1275-1280, 2016
APA
Tang, H., Ji, F., Sun, J., Xie, Y., Xu, Y., & Yue, H. (2016). RBEL1 is required for osteosarcoma cell proliferation via inhibiting retinoblastoma 1. Molecular Medicine Reports, 13, 1275-1280. https://doi.org/10.3892/mmr.2015.4670
MLA
Tang, H., Ji, F., Sun, J., Xie, Y., Xu, Y., Yue, H."RBEL1 is required for osteosarcoma cell proliferation via inhibiting retinoblastoma 1". Molecular Medicine Reports 13.2 (2016): 1275-1280.
Chicago
Tang, H., Ji, F., Sun, J., Xie, Y., Xu, Y., Yue, H."RBEL1 is required for osteosarcoma cell proliferation via inhibiting retinoblastoma 1". Molecular Medicine Reports 13, no. 2 (2016): 1275-1280. https://doi.org/10.3892/mmr.2015.4670