Next Article in Journal
Synthesis and Anti-Breast Cancer Evaluation of Novel N-(Guanidinyl)benzenesulfonamides
Next Article in Special Issue
Nipple Discharge of CA15-3, CA125, CEA and TSGF as a New Biomarker Panel for Breast Cancer
Previous Article in Journal
Molecular Dynamics Simulation on the Conformational Transition of the Mad2 Protein from the Open to the Closed State
Previous Article in Special Issue
Role of VEGF-A and Its Receptors in Sporadic and MEN2-Associated Pheochromocytoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Silencing of Ether à Go-Go 1 by shRNA Inhibits Osteosarcoma Growth and Cell Cycle Progression

1
Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou 363000, China
2
Department of Thoracic Surgery, the Affiliated Tangdu Hospital of Fourth Military Medical University, Xi'an 710038, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2014, 15(4), 5570-5581; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15045570
Submission received: 4 February 2014 / Revised: 18 March 2014 / Accepted: 20 March 2014 / Published: 1 April 2014
(This article belongs to the Special Issue Molecular Bases of Cancer Research)

Abstract

:
Recently, a member of the voltage-dependent potassium channel (Kv) family, the Ether à go-go 1 (Eag1) channel was found to be necessary for cell proliferation, cycle progression and tumorigenesis. However, the therapeutic potential of the Eag1 channel in osteosarcoma remains elusive. In the present study, a recombinant adenovirus harboring shRNA against Eag1 was constructed to silence Eag1 expression in human osteosarcoma MG-63 cells. We observed that Eag1-shRNA inhibited the proliferation and colony formation of MG-63 cells due to the induction of G1 phase arrest. Moreover, in vivo experiments showed that Eag1-shRNA inhibited osteosarcoma growth in a xenograft nude mice model. In addition, selective inhibition of Eag1 significantly decreased the expression levels of cyclin D1 and E. Taken together, our data suggest that the Eag1 channel plays a crucial role in regulating the proliferation and cell cycle of osteosarcoma cells, and represents a new and effective therapeutic target for osteosarcoma.

1. Introduction

Osteosarcoma (OS), which is the most common primary bone tumor in childhood and adolescence [1,2], often occurs in the long bones of the body, especially in distal femur and proximal tibia [3]. OS is derived from primitive bone-forming mesenchymal stem cells [4] and characterized by rapid growth and high metastatic potential. The two main sites of metastases of OS are lung and bone [5]. With the use of combination aggressive surgery and neoadjuvant chemotherapy, 5-year survival rates of patients presenting without metastatic disease have reached 60%–75% [6]. However, 40%–50% of patients will develop metastases with dismal prognosis, especially for patients who have primary pulmonary metastases [7]. Meanwhile, several adverse reactions are associated with high-dose chemotherapy. Thus, the development of new treatments for OS has become an urgent clinical need. With increasing knowledge and understanding of the molecular pathogenesis of OS, several genetic alterations have been found to be associated with OS [8], which may represent new potential targets for diagnosis and treatment.
Kv channels play key roles in both electrically excitable and nonexcitable cells. Recently, the importance of Kv channels in tumor biology has aroused interest [9]. Several Kv channels, especially the Eag1 (Kv10.1, KCNH1) channel stands out as the most attractive one due to its close relation to tumor development and progression. The Ether à go-go (Eag) gene was originally cloned from Drosophila melanogaster in 1969 [10]. Homology screening identified that the Eag channels family was formed by Eag, Erg (the eag-related gene) and Elk (the eag-like gene) subfamily. Two members of the Eag subfamily, three for Erg and two for Elk are differentially expressed in different species including rat, bovine and humans [11]. Two members of the Eag subfamily are Eag1 and Eag2 (Kv10.2, KCNH5) [12]. Interestingly, Eag1 channel shows a restricted distribution in normal tissues, it has been found almost exclusively in the brain but ectopically expressed in several tumor cell lines and more than 75% primary solid tumors [13,14]. In addition, suppression of Eag1 expression in several cancer cell lines causes a significant reduction of cell proliferation [15,16]. Furthermore, aggressive tumors were induced when cells were transfected with Eag1 and implanted into immune-depressed mice [17]. These studies show an oncogenic potential of Eag1 [17]. However, the molecular mechanisms responsible for the oncogenic role of Eag1 are not completely understood. Definitely Eag1 channel is a transmembrane protein, potassium permeation is not the only feature of its relevance to tumor progression. Furthermore, one of the most intriguing aspects of it, are the transformative properties [18]. Thus, further studies are needed to clarify exactly the nature of the mechanism.
Our previous studies provide evidence that Eag1 promotes the growth and angiogenesis of OS and the downreguation of Eag1 by siRNA or microRNA-34a exhibited antitumor effects on OS [19,20]. To further elucidate the mechanisms underlying the oncogenic role of Eag1 in OS, in this study we used OS cell line MG-63 as the experimental model and focused on the regulation of cell cycle progression by Eag1.

2. Results

2.1. Ad5-Eag1-shRNA Inhibits Eag1 Expression in MG-63 Cells

The Eag1 mRNA levels in OS cell line MG-63 infected with adenoviral shRNA vectors were determined by RT-PCR. The results showed that Eag1 mRNA expression was effectively suppressed by Ad5-Eag1-shRNA, compared with uninfected (negative-control) cells, or cells infected by Ad5-Control-shRNA (Figure 1A). In parallel, Western blot analysis confirmed that Eag1-shRNA reduced Eag1 protein expression in MG-63 cells (Figure 1B).

2.2. Ad5-Eag1-shRNA Inhibits MG-63 Cell Proliferation

To characterize the oncogenic role of Eag1 in OS cells, we used loss-of-function approach. As shown in Figure 2A, cell proliferation was inhibited by 38% in Ad5-Eag1-shRNA infected MG-63 cells. In addition, colony formation was inhibited in Ad5-Eag1-shRNA infected MG-63 cells (Figure 2B,C). Collectively, these results suggest that Eag1 promotes the proliferation of OS cells in vitro.

2.3. Ad5-Eag1-shRNA Inhibits Tumor Growth in a Xenograft Model of OS

To investigate the in vivo role of Eag1 in OS, we established a xenograft model of OS in nude mice (Figure 3A). The results showed that the tumor volume was significantly smaller in the control group compared to the Ad5-Eag1-shRNA group (Figure 3B). These in vivo data complement our in vitro results and suggest that the blockade of Eag1 suppresses OS growth in nude mice effectively.

2.4. Ad5-Eag1-shRNA Inhibits Cell Cycle Progression of MG-63 Cells

To determine how Eag1 knockdown inhibits OS growth, we analyzed the DNA contents of MG-63 cells by flow cytometry. As shown in Figure 4A, Ad5-Eag1-shRNA significantly affected cell cycle progression of MG-63 cells. Exponentially growing cells in the negative-control group had a distribution of 38.97% ± 2.64% in the G1 phase, 53.58% ± 3.37% in the S phase, and 7.46% ± 1.16% in the G2 phase (n = 3, Figure 4B), while the distribution of different phase in the Ad5-Control-shRNA group was 41.33% ± 3.21% in the G1 phase, 46.23% ± 2.16% in the S phase, and 9.77% ± 1.43% in the G2 phase (n = 3, Figure 4C). In contrast, in Ad5-Eag1-shRNA treated cells we observed a significant increase in the proportion of G1 phase (70.15% ± 3.36%, n = 3, p < 0.05), and a significant decrease in the proportion of S phase (21.73% ± 1.28%, n = 3, p < 0.05) and G2 phase (10.25% ± 1.20%, n = 3) (Figure 4D).

2.5. Eag1 Regulates the Expression of Cell Cycle Related Proteins in MG-63 Cells

In order to explain the detailed mechanisms of G1–S transition inhibition by specific blockade of Eag1, we examined the relative expression level of cyclin D1 and cyclin E using Western blot analysis. As shown in Figure 5, the expression level of cyclin D1 and cyclin E protein was significantly decreased by 0.75-fold (n = 3) and 0.79-fold (n = 3) respectively in the Ad5-Eag1-shRNA group. These results indicate that selective inhibition of Eag1 expression causes G1–S cell cycle progression of cultured MG-63 cells.

3. Discussion

Increasing evidence has suggested that Eag1 is a promising target for cancer treatment. Nonspecific blockers such as the tricyclic antidepressant imipramine and antihistamine astemizole, or specific blockers such as monoclonal antibodies or small interfering RNA (siRNA) against Eag1 could reduce the proliferation of cancer cells [16,21,22]. Thus, the potential therapeutic value of Eag1 channels is high [23].
Compared with nonspecific blockers, siRNA is a more specific tool to evaluate the role of Eag1 in cancer progression. Knockdown of Eag1 by siRNA resulted in reduced proliferation of tumor cells without obvious nonspecific responses [16]. In this study, we found that Eag1-shRNA could effectively downregulate Eag1 expression with good specificity. In addition, Eag1-shRNA inhibited cell proliferation in OS. Proliferation inhibition can be a result of cell cycle arrest. By flow cytometry analysis we demonstrated that Eag1-shRNA significantly increased the percentage of cells in the G1 phase and reduced the percentage of cells in the S and G2 phase.
To analyze how Eag1 modulates OS cell proliferation and cell cycle progression, it is important to note that Eag1 contributes to tumor progression independently of its primary function as an ion channel. Previous studies have shown that potassium permeation is not the only feature of the Eag1 channel relevant to tumor progression [18]. Moreover, overexpression of Eag1 in OS cells is regulated by the p38 mitogen-activated protein kinases (p38MAPK) pathway independent of K+ influx [24]. However, our in vitro and in vivo data could not exclude that the oncogenic role of Eag1 is dependent on its K+ conducting activity. Several K+ channels are often overexpressed in tumor cells and regulate proliferation. They are needed to control specific checkpoints in the cell cycle progression. It is well accepted that the Eag1 channel is necessary for progression through the G1 phase of the cell cycle [25]. This may be due to the fact that during cell cycle progression into the G1 phase, the membrane potential becomes hyperpolarized relative to the normal resting potential. The hyperpolarization relates to Eag1 channel activation in the early G1 and activated Eag1 channel and was accompanied by an increase of some regulator expression which is key for the G1–S transition.
Cyclins are a family of proteins that control cell cycle by activating cyclin-dependent kinase (Cdk) [26]. Recently, aberrant expression of cyclins was found to correlate with tumorigenesis [27,28]. Among them, cyclin D1 and cyclin E have been areas of intense investigation. Cyclin D1 is a key regulator of the G1 phase progression of the cell cycle [29], while cyclin E is required for the transition from G1 to S phase and is necessary for the entry of cells into S phase [30]. Additionally, the Eag1 channel plays a critical role in G1 phase progression of MCF-7 cells (human breast cancer) and cyclin D1 and E are the main factors regulated by Eag1 [30]. In order to explain the detailed mechanisms of the G1–S transition inhibition by Eag1 knockdown, we examined the expression levels of cyclin D1 and cyclin E. Our results suggest that the inhibition effect of Eag1 shRNA on MG-63 cell proliferation is achieved at least partially via the inhibition of cyclin D1 and cyclin E expression and G1–S progression. Interestingly, a recent study reported that the inhibition of Eag1 inhibited the proliferation of human bone marrow-derived mesenchymal stem cells, accompanied by reduced levels of cyclin D1, cyclin E, p-ERK1/2, and p-Akt [31]. These results are consistent with our data and suggest that Eag1 may regulate the expression of cyclin D1 and E via the activation of MAPK and PI3K/Akt signaling pathways. In fact, the inhibition of MAPK and PI3K/Akt pathways has been implicated in anti-tumor effects on OS [32]. Further studies are needed to elucidate the crosstalk of Eag1 with these signaling pathways in the initiation and progression of OS.
Taken together, this study provides in vitro and in vivo evidence of anti-proliferative effects of Eag1 knockdown on human OS cells. Our data help reveal possible mechanisms by which Eag1 promotes cell proliferation (Figure 6). shRNA targeting Eag1 may be considered a novel therapeutic agent for OS treatment.

4. Materials and Methods

4.1. Cell Culture

Human OS cell line MG-63 and human embryonic kidney cell line 293 (HEK293) were obtained from the American type culture of collection (ATCC, Manassas, VA, USA). MG-63 and HEK293 cells were cultured in RPMI-1640 medium (Gibco, Rockville, MD, USA) supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 μg/mL streptomycin at 37 °C in a humidified atmosphere with 5% CO2. Cells were subcultured every 3–4 days.

4.2. Preparation of Adenoviral shRNA Vectors

The oligonucleotides targeting human Eag1 were designed and selected as the template: AGC CAT CTT GGT CCC TTA TAA, which shared no homology with other coding sequences in human by BLAST analysis. A ring sequence of nine base pairs (TTC AAG ACG) existed between the sense and antisense strands. The shRNA was synthesis by Sangon Biotech (Shanghai, China). Plasmid pGeneSil-1, which was purchased from GeneSil Biotechnology (Wuhan, China), contained the human U6 promoter inserted between the BamHI and the HindIII sites. The shRNA-expressing cassette was subcloned into pAdTrack vector between the HindIII and the XbaI sites. The recombinant plasmid was linearized by digestion with restriction endonuclease, and subsequently cotransformed into E. coli BJ5183 cells with an adenoviral backbone plasmid, pAdEasy-1 (GeneSil Biotechnology, Wuhan, China). Recombinant plasmids were selected for kanamycin resistance, and transduced into the HEK293 cells. A recombinant adenovirus expressing shRNA against Eag1 (Ad5-Eag1-shRNA) was generated. The recombinant adenovirus control (Ad5-Control-shRNA), which carries a CTA GGT GTT CTA GTC TGG ACT and did not target any known human genes, was generated as control. All viruses were propagated and purified on a CsCl gradient using standard methods. The viruses were titrated for viral particles using standard methods based on spectrophotometry at 260 nm. Functional titer (plaque forming units) was determined with a plaque assay on HEK293 cells according to the method developed by Quantum Biotechnology (Taichuang, Taiwan, China).

4.3. Adenovirus Infection

MG-63 cells (1 × 105) in serum-free RPMI-1640 were infected with Ad5-Eag1-shRNA or Ad5-Control-shRNA at 5 MOI (multiplicity of infection, calculated as PFU/cell numbers) in a humidified atmosphere of 5% CO2 at 37 °C. Virus-containing medium was removed 8 h later and replaced with fresh RPMI-1640 medium containing 10% (v/v) FBS. Cells were incubated for an additional 48 h.

4.4. RT-PCR

Total RNA was isolated from the cultured cells by Trizol reagent (Invitrogen, Rockville, MD, USA). RNA purity and integrity was examined by running an aliquot on a denaturing 1% (w/v) agarose gel. cDNA was then synthesized from 1 μg of total RNA using 200 U reverse transcriptase (Takara, Tokyo, Japan), plus 200 μM dNTPs and 2.5 μM oligo-dT primer, in a 20 μL reaction volume, for 10 min at 30 °C then 60 min at 42 °C and finally at 80 °C for 5 min. One μL of cDNA was then amplified by PCR in 25 μL reaction containing 2.5 U DNA polymerase and 200 μM dNTPs. Sequences of forward and backward primers, amplified fragment sizes, annealing temperatures were as follows: Eag1, 5′-GCT TTT GAG AAC GTG GAT GAG-3′, 5′-CGA AGA TGG TGG CAT AGA GAA-3′, 475 bp, 56 °C. β-actin, 5′-TCC ACC TTC CAG CAG ATG TG-3′, 5′-GCA TTT GCG GTG GAC GAT-3′, 75 bp, 54 °C. Samples of PCR products were run on a 2% (w/v) agarose gel and the bands were visualized by ethidium bromide staining on a UV trans illuminator. Each experiment was repeated three times. Some of the PCR products were sequenced to check the PCR specificity.

4.5. Western Blot Analysis

A number of 5 × 107–6 × 107 cells were collected and lysed in ice-cold lysis buffer containing 50 mmol/L Tris-Cl (pH 7.5), 150 mmol/L NaCl, 0.2 mmol/L EDTA, 1 mmol/L PMSF and 1% (v/v) Nonidet-P40 for 30 min. The lysates were centrifuged at 13,200 rpm for 10 min at 4 °C and the supernatants were collected. Twenty five μg protein was resolved by a 12% SDS-PAGE and blotted on nitrocellulose membranes (Bio-Rad, Richmond, CA, USA). Membranes were blocked with 10% (w/v) nonfat milk powder at room temperature for 1 h, and then incubated with antibodies against Eag1 (Abcam, Cambridge, MA, USA), cyclin D1 (Cell Signaling Technology®, Danvers, MA, USA), cyclin E and GAPDH (Santa Cruz Biotechnology, Santa Cruz, CA, USA) overnight, followed by incubation with horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA). Then the membranes were developed with chemiluminescent detection kit (Zhongshan Biotechnology, Beijing, China) and exposed to X-ray films. Experiments were performed at least three times with representative data presented.

4.6. Cell Proliferation Assay

The cell proliferation was analyzed by using Cell Counting Assay Kit-8 (CCK-8; Dojindo Molecular Technologies, Gaithersburg, MD, USA) according to the manufacturer’s protocol. In brief, 1 × 104 cells were starved in a serum-free medium for 12 h and then the cells were transduced. After 48 h, cells were harvested. Ten microliters of Cell Counting Assay Kit-8 solution was added to each well, the cells were incubated for another 1 h, and the absorbance (A) at 490 nm was measured by using a spectrophotometer (Bio-Rad, Richmond, CA, USA). Experiments were performed at least three times with representative data presented.

4.7. Colony Formation Assay

For the colony formation assay, 0.5% agar (Sigma, St. Louis, MO, USA) was added in 60 mm dishes. The treated MG-63 cells were mixed with 0.3% soft agar and added to the bottom agar with 300 cells per dish. The cells were incubated for up to two weeks. The assay was stopped when the colonies were clearly visible even without looking under the microscope.

4.8. Tumorigenicity Assay in Nude Mice

Thymus-null BALB/c nude mice (female, age 6–8 weeks) were obtained from the Animal Center of the Chinese Academy of Medical Sciences (Beijing, China). All animal procedures were performed according to approved protocols and in accordance with recommendations for the proper use and care of laboratory animals. OS xenografts were established in nude mice according to our previous reports [20]. In brief, a total of 1.5 × 105 MG-63 cells in 150 μL Phosphate-buffered Saline (PBS) were subcutaneously injected in the hind right leg. One week later, the tumors grew to visible size. The OS-bearing mice were randomly divided into three groups (six in each group). Group 1 received intra-tumor injections with Ad5-Eag1-shRNA (10 MOI) every 2 days (6 injections totally). Group 2 received intra-tumor injections of Ad5-Control-shRNA (10 MOI) every 2 days (6 injections totally). Group 3 received normal saline injection as controls. Tumor volume (mm3) was determined based on the following formula: ab2/2 where a was the length and b was the width of the tumor [20].

4.9. Flow Cytometry Analysis

The cells were collected and washed twice with cold PBS following the infection with shRNA for 48 h, and then resuspended at 1 × 106 cells/mL and fixed in 70% cold ethanol overnight at 4 °C. A fluorochrome solution containing 50 μg/mL PI, 3.4 mmol/L sodium citration, 20 μg/mL RNase A and 1% Triton X-100 was added and the mixture was incubated in the dark at room temperature for 30 min. The distribution of the cell cycle was measured using flow cytometry (FCM; Partec, Münster, Germany). FCM analysis was performed using the Cell Quest software (Beckton Dickinson, Franklin Lakes, NJ, USA).

4.10. Statistical Analysis

All data were presented as mean ± standard deviation (SD). Statistical significance was determined using t-test or analysis of variance (ANOVA) using the SPSS18.0 program (SPSS Company, Chicago, IL, USA). p < 0.05 was considered as significant difference.

5. Conclusions

Our findings suggest that the Eag1 channel plays a crucial role in regulating the proliferation and cell cycle of osteosarcoma cells, and represents a new and effective therapeutic target for osteosarcoma.

Acknowledgments

This study was supported by the Scientific and Technologial Innovation Programs of the Nanjing military region, China (No. 10MA077).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Damron, T.A.; Ward, W.G.; Stewart, A. Osteosarcoma, chondrosarcoma, and Ewing’s sarcoma: National cancer data base report. Clin. Orthop. Relat. Res 2007, 459, 40–47. [Google Scholar]
  2. Sluga, M.; Windhager, R.; Pfeiffer, M.; Ofner, P.; Lang, S.; Dominkus, M.; Nehrer, S.; Zoubek, A.; Kotz, R. Osteosarcoma and Ewing’s sarcoma-The most frequent malignant bone tumors in children-therapy and outcome. Z. Orthop. Ihre Grenzgeb 2002, 140, 652–655. [Google Scholar]
  3. Klein, M.J.; Siegal, G.P. Osteosarcoma: Anatomic and histologic variants. Am. J. Clin. Pathol 2006, 125, 555–581. [Google Scholar]
  4. Benayahu, D.; Shur, I.; Marom, R.; Meller, I.; Issakov, J. Cellular and molecular properties associated with osteosarcoma cells. J. Cell Biochem 2001, 84, 108–114. [Google Scholar]
  5. Panizo-Santos, A.; Sola, I.; Lozano, M.; de Alava, E.; Pardo, J. Metastatic osteosarcoma presenting as a small bowel polyp: A case report and review of the literature. Arch. Pathol. Lab. Med 2000, 124, 1682–1684. [Google Scholar]
  6. Hawkins, D.S.; Arndt, C.A. Pattern of disease recurrence and prognostic factors in patients with osteosarcoma treated with contemporary chemotherapy. Cancer 2003, 98, 2447–2456. [Google Scholar]
  7. Wu, P.K.; Chen, W.M.; Chen, C.F.; Lee, O.K.; Haung, C.K.; Chen, T.H. Primary osteogenic sarcoma with pulmonary metastasis: Clinical results and prognostic factors in 91 patients. Jpn. J. Clin. Oncol 2009, 39, 514–522. [Google Scholar]
  8. Tan, M.L.; Choong, P.F.; Dass, C.R. Osteosarcoma: Conventional treatment vs. gene therapy. Cancer Biol. Ther 2009, 8, 106–117. [Google Scholar]
  9. Pardo, L.A.; Contreras-Jurado, C.; Zientkowska, M.; Alves, F.; Stuhmer, W. Role of voltage-gated potassium channels in cancer. J. Membr. Biol 2005, 205, 115–124. [Google Scholar]
  10. Kaplan, W.D.; Trout, W.E., 3rd. The behavior of four neurological mutants ofDrosophila. Genetics 1969, 61, 399–409. [Google Scholar]
  11. Warmke, J.W.; Ganetzky, B. A family of potassium channel genes related to eag in Drosophila and mammals. Proc. Natl. Acad. Sci. USA 1994, 91, 3438–3442. [Google Scholar]
  12. Bauer, C.K.; Schwarz, J.R. Physiology of EAG K+ channels. J. Membr. Biol 2001, 182, 1–15. [Google Scholar]
  13. Camacho, J. Ether à go-go potassium channels and cancer. Cancer Lett 2006, 233, 1–9. [Google Scholar]
  14. Hemmerlein, B.; Weseloh, R.M.; de Queiroz, F.M.; Knötgen, H.; Sánchez, A.; Rubio, M.E.; Martin, S.; Schliephacke, T.; Jenke, M.; Radzun, H.J.; et al. Overexpression of Eag1 potassium channels in clinical tumours. Mol. Cancer 2006, 5, 41. [Google Scholar]
  15. De Queiroz, F.M.; Suarez-Kurtz, G.; Stühmer, W.; Pardo, L.A. Ether à go-go potassium channel expression in soft tissue sarcoma patients. Mol. Cancer 2006, 5, 42. [Google Scholar]
  16. Weber, C.; de Queiroz, F.M.; Downie, B.R.; Suckow, A.; Stühmer, W.; Pardo, L.A. Silencing the activity and proliferative properties of the human EagI potassium channel by RNA interference. J. Biol. Chem 2006, 281, 13030–13037. [Google Scholar]
  17. Pardo, L.A.; del Camino, D.; Sánchez, A.; Alves, F.; Brüggemann, A.; Beckh, S.; Stühmer, W. Oncogenic potential of EAG K+ channels. EMBO J 1999, 18, 5540–5547. [Google Scholar]
  18. Downie, B.R.; Sánchez, A.; Knötgen, H.; Contreras-Jurado, C.; Gymnopoulos, M.; Weber, C.; Stühmer, W.; Pardo, L.A. Eag1 expression interferes with hypoxia homeostasis and induces angiogenesis in tumors. J. Biol. Chem 2008, 283, 36234–36240. [Google Scholar]
  19. Wu, J.; Wu, X.; Zhong, D.; Zhai, W.; Ding, Z.; Zhou, Y. Short hairpin RNA (shRNA) ether à go-go 1 (Eag1) inhibition of human osteosarcoma angiogenesis via VEGF/PI3K/AKT signaling. Int. J. Mol. Sci 2012, 13, 12573–12583. [Google Scholar]
  20. Wu, X.; Zhong, D.; Gao, Q.; Zhai, W.; Ding, Z.; Wu, J. MicroRNA-34a inhibits human osteosarcoma proliferation by downregulating ether à go-go 1 expression. Int. J. Med. Sci 2013, 10, 676–682. [Google Scholar]
  21. García-Ferreiro, R.E.; Kerschensteiner, D.; Major, F.; Monje, F.; Stühmer, W.; Pardo, L.A. Mechanism of block of hEag1 K+ channels by imipramine and astemizole. J. Gen. Physiol 2004, 124, 301–317. [Google Scholar]
  22. Gómez-Varela, D.; Zwick-Wallasch, E.; Knötgen, H.; Sánchez, A.; Hettmann, T.; Ossipov, D.; Weseloh, R.; Contreras-Jurado, C.; Rothe, M.; Stühmer, W.; et al. Monoclonal antibody blockade of the human Eag1 potassium channel function exerts antitumor activity. Cancer Res 2007, 67, 7343–7349. [Google Scholar]
  23. Pardo, L.A.; Stühmer, W. Eag1 as a cancer target. Expert Opin. Ther. Targets 2008, 12, 837–843. [Google Scholar]
  24. Wu, X.; Zhong, D.; Lin, B.; Zhai, W.; Ding, Z.; Wu, J. p38 MAPK regulates the expression of ether à go-go potassium channel in human osteosarcoma cells. Radiol. Oncol 2013, 47, 42–49. [Google Scholar]
  25. Pardo, L.A.; Brüggemann, A.; Camacho, J.; Stühmer, W. Cell cycle-related changes in the conducting properties of r-eag K+ channels. J. Cell Biol 1998, 143, 767–775. [Google Scholar]
  26. Galderisi, U.; Jori, F.P.; Giordano, A. Cell cycle regulation and neural differentiation. Oncogene 2003, 22, 5208–5219. [Google Scholar]
  27. Deshpande, A.; Sicinski, P.; Hinds, P.W. Cyclins and cdks in development and cancer: A perspective. Oncogene 2005, 24, 2909–2915. [Google Scholar]
  28. Malumbres, M. Cyclins and related kinases in cancer cells. J. BUON 2007, 12, S45–S52. [Google Scholar]
  29. Drobnjak, M.; Osman, I.; Scher, H.I.; Fazzari, M.; Cordon-Cardo, C. Overexpression of cyclin D1 is associated with metastatic prostate cancer to bone. Clin. Cancer Res 2000, 6, 1891–1895. [Google Scholar]
  30. Borowiec, A.S.; Hague, F.; Gouilleux-Gruart, V.; Lassoued, K.; Ouadid-Ahidouch, H. Regulation of IGF-1-dependent cyclin D1 and E expression by hEag1 channels in MCF-7 cells: The critical role of hEag1 channels in G1 phase progression. Biochim. Biophys. Acta 2011, 1813, 723–730. [Google Scholar]
  31. Zhang, Y.Y.; Yue, J.; Che, H.; Sun, H.Y.; Tse, H.F.; Li, G.R. BKCa and hEag1 channels regulate cell proliferation and differentiation in human bone marrow-derived mesenchymal stem cells. J. Cell. Physiol 2014, 229, 202–212. [Google Scholar]
  32. Miwa, S.; Sugimoto, N.; Yamamoto, N.; Shirai, T.; Nishida, H.; Hayashi, K.; Kimura, H.; Takeuchi, A.; Igarashi, K.; Yachie, A.; et al. Caffeine induces apoptosis of osteosarcoma cells by inhibiting AKT/mTOR/S6K, NF-κB and MAPK pathways. Anticancer Res 2012, 32, 3643–3649. [Google Scholar]
Figure 1. shRNA mediated knockdown of Ether à go-go 1 (Eag1) in MG-63 cells. (A) RT-PCR analysis of Eag1 mRNA level in MG-63 cells treated with Eag1-shRNA. β-actin was internal control; (B) Western blot analysis of Eag1 protein level in MG-63 cells treated with Eag1-shRNA; (C) Densitometry analysis of Rag1 protein level. GAPDH was loading control. The results were expressed as mean ± SD (n = 3). ** p < 0.01.
Figure 1. shRNA mediated knockdown of Ether à go-go 1 (Eag1) in MG-63 cells. (A) RT-PCR analysis of Eag1 mRNA level in MG-63 cells treated with Eag1-shRNA. β-actin was internal control; (B) Western blot analysis of Eag1 protein level in MG-63 cells treated with Eag1-shRNA; (C) Densitometry analysis of Rag1 protein level. GAPDH was loading control. The results were expressed as mean ± SD (n = 3). ** p < 0.01.
Ijms 15 05570f1
Figure 2. Eag1 blockage inhibits the proliferation and tumorigenicity of OS cells. (A) The proliferation of MG-63 cells was determined by CCK-8 assay. The proliferation of MG-63 cells was significantly reduced after treatment with Eag1-shRNA. Data were presented as mean ± SD (n = 6). ** p < 0.01; (B,C) The tumorigenicity of MG-63 cells was determined by colony formation assay. The tumorigenicity of MG-63 cells was significantly reduced after treatment with Eag1-shRNA. Data were presented as mean ± SD (n = 3). ** p < 0.01.
Figure 2. Eag1 blockage inhibits the proliferation and tumorigenicity of OS cells. (A) The proliferation of MG-63 cells was determined by CCK-8 assay. The proliferation of MG-63 cells was significantly reduced after treatment with Eag1-shRNA. Data were presented as mean ± SD (n = 6). ** p < 0.01; (B,C) The tumorigenicity of MG-63 cells was determined by colony formation assay. The tumorigenicity of MG-63 cells was significantly reduced after treatment with Eag1-shRNA. Data were presented as mean ± SD (n = 3). ** p < 0.01.
Ijms 15 05570f2
Figure 3. Ad5-Eag1-shRNA inhibits OS growth in vivo. (A) Schematic representation of the experiments with OS xenograft tumors and different treatments. Xenograft tumors were established by subcutaneous injection of 1 × 105 MG-63 cells. After one week, the tumors grew to visible size. Ad5-Eag1-shRNA treatment was given by intra-tumor injection every two days (blue arrows). Mice in the control group were injected with saline or Ad5-Control-shRNA; (B) The length and width of tumors were measured every two days after inoculation and the volume of tumor was calculated. After 10 days, the tumor volume growth curve was drafted. ** p < 0.01 vs. saline or Ad5-Control-shRNA group.
Figure 3. Ad5-Eag1-shRNA inhibits OS growth in vivo. (A) Schematic representation of the experiments with OS xenograft tumors and different treatments. Xenograft tumors were established by subcutaneous injection of 1 × 105 MG-63 cells. After one week, the tumors grew to visible size. Ad5-Eag1-shRNA treatment was given by intra-tumor injection every two days (blue arrows). Mice in the control group were injected with saline or Ad5-Control-shRNA; (B) The length and width of tumors were measured every two days after inoculation and the volume of tumor was calculated. After 10 days, the tumor volume growth curve was drafted. ** p < 0.01 vs. saline or Ad5-Control-shRNA group.
Ijms 15 05570f3
Figure 4. The effects of Eag1-shRNA on cell cycle progression of MG-63 cells. (A) Cells were infected with shRNA for 48 h. Eag1 knockdown induced a significant increase in cells arrested in the G1 phase (p < 0.05) and a decrease in cells arrested in the S phase (p < 0.05); (BD) Representative flow cytometry images of MG-63 cells in negative-control (38.97% ± 2.64% in the G1 phase, 53.58% ± 3.37% in the S phase, and 7.46% ± 1.16% in the G2 phase), Ad5-Control-shRNA (41.33% ± 3.21% in the G1 phase, 46.23% ± 2.16% in the S phase, and 9.77% ± 1.43% in the G2 phase), and Ad5-Eag1-shRNA groups (70.15% ± 3.36% in the G1 phase, 21.73% ± 1.28% in the S phase, and 10.25% ± 1.20% in the G2 phase).
Figure 4. The effects of Eag1-shRNA on cell cycle progression of MG-63 cells. (A) Cells were infected with shRNA for 48 h. Eag1 knockdown induced a significant increase in cells arrested in the G1 phase (p < 0.05) and a decrease in cells arrested in the S phase (p < 0.05); (BD) Representative flow cytometry images of MG-63 cells in negative-control (38.97% ± 2.64% in the G1 phase, 53.58% ± 3.37% in the S phase, and 7.46% ± 1.16% in the G2 phase), Ad5-Control-shRNA (41.33% ± 3.21% in the G1 phase, 46.23% ± 2.16% in the S phase, and 9.77% ± 1.43% in the G2 phase), and Ad5-Eag1-shRNA groups (70.15% ± 3.36% in the G1 phase, 21.73% ± 1.28% in the S phase, and 10.25% ± 1.20% in the G2 phase).
Ijms 15 05570f4
Figure 5. Regulation of cyclin D1 and cyclin E expression by Eag1-shRNA in MG-63 cells. (A) Representative blots showing significantly decreased expression of cyclin E (n = 3) and cyclin D1 (n = 3); (B) Densitometry analysis of cyclin E and cyclin D1 protein levels in MG-63 cells. GAPDH was the loading control. All data were normalized to the negative-control and presented as mean ± SD (n = 3). *** p < 0.001.
Figure 5. Regulation of cyclin D1 and cyclin E expression by Eag1-shRNA in MG-63 cells. (A) Representative blots showing significantly decreased expression of cyclin E (n = 3) and cyclin D1 (n = 3); (B) Densitometry analysis of cyclin E and cyclin D1 protein levels in MG-63 cells. GAPDH was the loading control. All data were normalized to the negative-control and presented as mean ± SD (n = 3). *** p < 0.001.
Ijms 15 05570f5
Figure 6. Schematic illustration of the role of Eag1 channel during OS cell cycle progression and proliferation.
Figure 6. Schematic illustration of the role of Eag1 channel during OS cell cycle progression and proliferation.
Ijms 15 05570f6

Share and Cite

MDPI and ACS Style

Wu, J.; Zhong, D.; Fu, X.; Liu, Q.; Kang, L.; Ding, Z. Silencing of Ether à Go-Go 1 by shRNA Inhibits Osteosarcoma Growth and Cell Cycle Progression. Int. J. Mol. Sci. 2014, 15, 5570-5581. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15045570

AMA Style

Wu J, Zhong D, Fu X, Liu Q, Kang L, Ding Z. Silencing of Ether à Go-Go 1 by shRNA Inhibits Osteosarcoma Growth and Cell Cycle Progression. International Journal of Molecular Sciences. 2014; 15(4):5570-5581. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15045570

Chicago/Turabian Style

Wu, Jin, Daixing Zhong, Xijin Fu, Qingjun Liu, Liangqi Kang, and Zhenqi Ding. 2014. "Silencing of Ether à Go-Go 1 by shRNA Inhibits Osteosarcoma Growth and Cell Cycle Progression" International Journal of Molecular Sciences 15, no. 4: 5570-5581. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms15045570

Article Metrics

Back to TopTop