Back to Journals » OncoTargets and Therapy » Volume 8

Disruption of human papillomavirus 16 E6 gene by clustered regularly interspaced short palindromic repeat/Cas system in human cervical cancer cells

Authors Yu L, Wang X, Zhu D, Ding W, Wang L, Zhang C, Jiang X, Shen H, Liao S, Ma D, Hu Z, Wang H

Received 14 March 2014

Accepted for publication 9 June 2014

Published 22 December 2014 Volume 2015:8 Pages 37—44

DOI https://doi.org/10.2147/OTT.S64092

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3



This paper has been retracted

 

Lan Yu, Xiaoli Wang, Da Zhu, Wencheng Ding, Liming Wang, Changlin Zhang, Xiaohui Jiang, Hui Shen, Shujie Liao, Ding Ma, Zheng Hu, Hui Wang

Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China

Abstract: High-risk human papillomavirus (HPV), especially HPV16, is considered a main causative agent of cervical cancer. Upon HPV infection, the viral oncoprotein E6 disrupts the host tumor-suppressor protein p53, thus promoting malignant transformation of normal cervical cells. Here, we used the newly developed programmable ribonucleic acid-guided clustered regularly interspaced short palindromic repeat (CRISPR)/Cas system to disrupt the HPV16 E6 gene. We showed that HPV16 E6 deoxyribonucleic acid was cleaved at specific sites, leading to apoptosis and growth inhibition of HPV16-positive SiHa and CaSki cells, but not HPV-negative C33A or human embryonic kidney 293 cells. We also observed downregulation of the E6 protein and restoration of the p53 protein. These data proved that the HPV16 E6 ribonucleic acid-guided CRISPR/Cas system might be an effective therapeutic agent in treating HPV infection-related cervical malignancy.

Keywords: CRISPR/Cas system, E6, p53, SiHa, CaSki, cervical cancer

Corrigendum for this paper has been published

Introduction

Cervical cancer is the second-most common cancer in women worldwide.1 Human papillomavirus (HPV) has long been considered to be a major risk factor for cervical cancer, especially HPV16.2 During the process of cervical carcinogenesis, the HPV oncogenes E6 and E7 promote and maintain the malignant phenotype of cervical cancer cells,3,4 making these genes attractive targets for cancer gene therapy.

Being able to target any site of the genome in the form of 5′-N20NGG-3′,5 the newly discovered clustered regularly interspaced short palindromic repeat (CRISPR)/Cas system has emerged as a powerful genome-editing tool in many organisms, including prokaryotes, Caenorhabditis elegans, and zebrafish.68 It consists of a Cas9 enzyme and a guidance ribonucleic acid (gRNA) for a specific deoxyribonucleic acid (DNA) sequence.5 Under the guidance of gRNAs, Cas9 enzyme will induce double-strand breaks (DSBs) at a determined DNA sequence complementary to the gRNA sequence (Figure 1). The breaks in the DNA are repaired by the mutagenic nonhomologous end joining (NHEJ) repair pathway, which leads to the disruption of the targeted gene.9,10

Figure 1 (A and B) Schematic representation of HPV16 E6 gene editing using CRISPR/Cas system.
Notes: (A) CRISPR/Cas system-mediated HPV16 E6 gene disruption can induce DSBs in the E6 gene, which will then be repaired by the NHEJ DNA-repairing pathway. Disruption of E6 DNA resulting from the NHEJ-repairing pathway can lead to apoptosis and growth inhibition in HPV16-infected cells. (B) Schematic representation of gRNAs targeting the E6 gene. The coordinates (268 bp, 404 bp, and 507 bp) refer to DSB points, counting from the start of the HPV16 genome. The DNA DSBs mediated by the CRISPR/Cas system are located 3 bp ahead of the protospacer-adjacent motif sequence upon gRNA recognition.
Abbreviations: HPV, human papillomavirus; CRISPR, clustered regularly interspaced short palindromic repeat; DSBs, double-strand breaks; DNA, deoxyribonucleic acid; gRNA, guide ribonucleic acid; NHEJ, nonhomologous end joining; URR, upstream regulatory region.

In this study, we applied the CRISPR/Cas system to target the E6 oncogene in HPV16-infected cervical cancer cell lines and investigated the feasibility of HPV16 E6-guided CRISPR/Cas as a potential intervention strategy for treatment of cervical cancer.

Materials and methods

Cell culture and transfection

SiHa, CaSki, C33A, and human embryonic kidney (HEK)-293 cells were purchased from ATCC (American Type Culture Collection, Manassas, VA, USA) and maintained in Dulbecco’s Modified Eagle’s Medium (Sigma-Aldrich Co., St Louis, MO, USA) in a humidified 5% CO2 chamber at 37°C. The cells were seeded onto 24-well and six-well plates and transfected when cellular confluence reached 80%. The transfection reagent we used was the X-tremeGene HP DNA transfection reagent (Hoffman-La Roche Ltd., Basel, Switzerland) for SiHa, C33A, and HEK293 cells. JetPEI® polymer-based DNA transfection reagent (Polyplus-transfection Inc., NY, USA) was applied to transfect CaSki cells. The total amount of DNA added to each well in 24-well and six-well plates was 1 μg and 2 μg, respectively.

Plasmids

Plasmid-encoding Cas9 was kindly provided by Addgene. As recent in vitro work has shown that the specificity of RNA-guided endonuclease is guaranteed simply by a short synthetic RNA molecule with no Cas9 restriction,11 we constructed three gRNAs targeting the HPV16 E6 gene following the protocol of Mali et al,5 and proved their effect in SiHa and CaSki cells. Sequences of the customized gRNAs are described in Table 1.

Table 1 CRISPR-gRNA recognition sequences and corresponding PAM sequences in this article
Abbreviations: CRISPR, clustered regularly interspaced short palindromic repeat; gRNA, guide ribonucleic acid; PAM, protospacer-adjacent motif.

Single-strand annealing luciferase reporter assay

Construction of the single-strand annealing (SSA) luciferase reporter pSSA Rep3-1 plasmid has been described previously.12 The pSSA Rep3-1 and control GATA zinc finger (GZF)-3-L3 + GZF1-R3 zinc-finger nuclease (ZFN) plasmids were kindly provided by Professor David Segal. Cells transfected with GZF3-L3, GZF1-R3, and pSSA Rep3-1 plasmids were used as a positive control. We then inserted a stop codon, CRISPR gRNA target sequence, and corresponding protospacer-adjacent motif sequence into the direct repeat halves of the firefly luciferase gene pSSA Rep3-1, which was then named as pSSA Rep-gRNA reporter. Cas9 plasmid (400 ng), 100 ng of each gRNA plasmid, 100 ng of pSSA Rep-gRNA reporter plasmid, and 25 ng of pRL-TK-Renilla luciferase (Promega Corporation, Fitchburg, WI, USA) were cotransfected into HEK293 cells in 24-well plates. At 48 hours’ posttransfection, cells were harvested and lysed according to the protocol of a dual-luciferase reporter assay system (Promega). Luciferase activity was determined in a microplate luminometer (BioTek).

T7 endonuclease 1 assay

The T7 endonuclease I (T7E1) assay was performed as previously described.13 Briefly, the region containing the targeting site of HPV16 E6 was polymerase chain reaction (PCR)-amplified. The primers used are described in Table 2. The PCR-amplified products were denatured by heating and reannealed to form heteroduplex DNA, and then treated with two units of T7E (New England Biolabs) per 200 ng for 15 minutes at 37°C, and finally electrophoresed using a 10% Tris/borate/ethylenediaminetetraacetic acid polyacrylamide gel.

Table 2 Primers used for PCR in this article
Abbreviations: PCR, polymerase chain reaction; gRNA, guide ribonucleic acid.

Detection of apoptosis

At 48 hours’ posttransfection, cells were trypsinized and costained with fluorescein isothiocyanate (FITC)-conjugated annexin V and propidium iodide using an annexin V-FITC apoptosis-detection kit (KeyGen Biotech) according to the manufacturer’s instructions. Double-positive cells were analyzed using a FACSCalibur™ (BD Biosciences, San Jose, CA, USA) to calculate CRISPR/Cas system-induced HPV16-positive cell death. C33A and HEK293 cells were also transfected with the same amount of plasmids. The data were analyzed using BD CellQuest™ software.

Cell-proliferation assay

Cell-proliferation assays were performed using the Cell Counting Kit-8 (CCK-8; Beyotime). SiHa, CaSki, C33A, and HEK293 cells were seeded onto 96-well plates at a density of 5×103 cells per well at 24 hours’ posttransfection. All experiments were performed three times. At 0, 24, 48, 72, and 96 hours after being seeded onto 96-well plates, CCK-8 solution was applied at 10 μL per well, followed by 3 hours’ incubation at 37°C. Absorbance values were measured at 490 nm in a microplate reader (Bio-Rad Laboratories Inc., Hercules, CA, USA).

Western blot analysis

The protein levels of E6 and p53 in SiHa and CaSki Cells were determined by Western blot analysis. We seeded SiHa and CaSki cells onto six-well plates. SiHa and CaSki cells were cotransfected with Cas9/gRNA plasmid. At 48 hours’ posttransfection, cells were collected and lysed on ice for 30 minutes with a lysis buffer containing 150 mM NaCl, 50 mM Tris (pH 7.4), 1% Triton X-100, 1% sodium deoxycholate, 0.1% sodium dodecyl sulfate, and a protease-inhibitor cocktail. Cellular proteins were transferred onto a polyvinylidene difluoride membrane. Nonspecific binding sites of protein were blocked by 5% skim milk in Tris-buffered saline–Tween 20. The final results were detected by horseradish peroxidase-conjugated antirabbit immunoglobulin secondary antibody using a SuperSignal™ West Pico kit (Thermo Fisher Scientific, Waltham, MA, USA). Antibodies used for Western blot were: anti-glyceraldehyde 3-phosphate dehydrogenase (GAPDH; 10494-1-AP; Proteintech), anti-p53 (10442-1-AP; Proteintech), and anti-HPV16 E6 (orb10837; Biorbyt, Cambridge, UK). GAPDH was used as an internal control.

Statistical analysis

Statistical analysis among different groups was conducted using one-way analysis of variance. All analyses were performed with SPSS 12.0 (SPSS Inc., Chicago, IL, USA). Data are presented as means ± standard deviation. P<0.05 was considered statistically significant.

Results

HPV16 E6 gRNAs induced DSBs at specific sites of HPV16 E6 gene

We first synthesized three gRNAs targeting the HPV16 E6 gene (Figure 1). We tested whether the three synthesized gRNAs could induce DSBs at specific sites by using a mammalian cell-based SSA assay in HEK293 cells. When gRNA-induced DSBs occurred, SSA homologous recombination led to the formation of an active luciferase gene. The Renilla luciferase plasmid was used as a control to monitor CRISPR-induced cytotoxicity. Compared with the negative control, which was transfected with only Cas9 plasmid (Figure 2A), the gRNA-2/Cas9 and gRNA-3/Cas9 group showed higher activity, while the gRNA-1/Cas9 group did not. Meanwhile, measurement of the cotransfected Renilla luciferase revealed no significant change of signal (Figure 2B), indicating that the gRNAs used in this study had minimal cytotoxicity.

Figure 2 (AD) CRISPR gRNA/Cas9-mediated HPV16 E6 DNA cleavage verified by dual-luciferase reporter assay and T7E1 assay.
Notes: Firefly luciferase (A) and Renilla luciferase (B) activity were measured at 48 hours after transfection. Negative cells were transfected with a Cas9 plasmid. (C and D) The T7E1 assay was applied to detect mutations of the HPV16 E6 gene caused by the CRISPR/Cas system. Black arrows indicate the expected positions of PCR products cleaved by T7E1 in SiHa (C) cells and in CaSki (D) cells. M represents the 100 bp DNA marker. Blank cells were treated with only Cas9 plasmid. Indel represents the Cas9-mediated cleavage efficiency which was calculated according to the fraction of cleaved DNA of HPV16 E6 gene. **P<0.01, n=4. One-way analysis of variance.
Abbreviations: CRISPR, clustered regularly interspaced short palindromic repeat; gRNA, guide ribonucleic acid; HPV, human papillomavirus; DNA, deoxyribonucleic acid; T7E1, T7 endonuclease 1; PCR, polymerase chain reaction; RLU, relative light unit.

We performed the T7E1 assay to further confirm the activity of the three gRNAs. Each of the gRNA/Cas9 plasmids was introduced into SiHa and CaSki cells. Cellular DNA of gRNA/Cas9-transfected cells were PCR-amplified and treated with T7E1. In SiHa and CaSki cells, after the T7E1 treatment of heteroduplex DNA, the gRNA-2/Cas9 and gRNA-3/Cas9 groups gave rise to cleaved products at about 200 and 300 bp, while the products of the gRNA-1/Cas9 group were uncleaved. The CRISPR/Cas system induced mutations at the HPV16 E6 site with frequencies up to 22% (gRNA-2/Cas9 group) and 21% (gRNA-3/Cas9 group) in SiHa cells, and 17% (gRNA-2/Cas9 group) and 19% (gRNA-3/Cas9 group) in CaSki cells (Figure 2C and D). Therefore, gRNA-1/Cas9 was not effective, and the gRNA-2/Cas9 and gRNA-3/Cas9 groups demonstrating cleavage activities were selected for further study. Taken together, these results indicate that the gRNA-guided CRISPR/Cas system can effectively induce DSBs in the E6 oncogene.

Effects of E6 gRNAs on apoptosis

To determine whether gRNA-guided CRISPR could specifically induce apoptosis of HPV16-positive human cervical cancer cells, we treated SiHa, CaSki, C33A, and HEK293 cells with E6-specific gRNA/Cas9 and calculated the apoptotic rates of these cells by flow-cytometry analysis. Compared with controls, HPV16-positive SiHa and CaSki cells showed dramatically increased apoptotic rates when treated with gRNA-2/Cas9 and gRNA-3/Cas9 (Figure 3A and B). On the other hand, HPV-negative C33A and HEK293 cells demonstrated no apparent apoptosis on receiving the same gRNA/Cas9 (Figure 3C and D). The corresponding apoptotic rates of these four cell lines that were treated with gRNA-2/Cas9 and gRNA-3/Cas9 are shown in Figure 3E and F.

Figure 3 (AF) CRISPR/Cas system-induced apoptosis.
Notes: The apoptosis rate was monitored by flow cytometry after 48 hours of transfection. SiHa (A), CaSki (B), C33A (C), and HEK293 (D) cells were costained with propidium iodide and annexin V-FITC. Following gRNA/Cas9 treatment, apoptosis of SiHa and CaSki cells increased significantly compared to untreated and gRNA-1/Cas9-treated cells, while C33A and HEK293 cells did not (E and F). **P<0.01; n=3. One-way analysis of variance. “Blank” represents nontransfected cells.
Abbreviations: CRISPR, clustered regularly interspaced short palindromic repeat; HEK, human embryonic kidney; FITC, fluorescein isothiocyanate; gRNA, guide ribonucleic acid.

Cellular proliferation affected by E6 disruption

The inhibitory effects of CRISPR gRNA/Cas9 on cell proliferation were detected by CCK-8 assay. At 72 and 96 hours posttransfection, the absorbance values of SiHa and CaSki cells treated with gRNA-2/Cas9 and gRNA-3/Cas9 were significantly lower than those of gRNA-1/Cas9-transfected cells and the untreated cells (Figure 4A and B). However, in HPV16-negative C33A and HEK293 cells, the absorbance rate showed no significant difference between transfected and nontransfected wells, indicating that E6 gRNA/Cas9 inhibited cellular proliferation only in HPV16-positive cell lines (Figure 4C and D).

Figure 4 (AD) gRNA/Cas9-mediated inhibition of cellular proliferation. Viability of SiHa (A), CaSki (B), C33A (C), and HEK293 (D) cells was measured by CCK-8 assay. “Blank” represents nontransfected cells.
Notes: *P<0.05; **P<0.01; n=3. One-way analysis of variance.
Abbreviations: gRNA, guide ribonucleic acid; HEK, human embryonic kidney; CCK, cell counting kit; OD, optical density.

Effects of E6 gRNAs on protein levels of E6 and p53

The effects of gRNA-guided CRISPR on E6 and p53 protein expression were detected by Western blot analysis. HPV16 E6 protein levels were downregulated in both cell lines, and p53 protein levels were upregulated correspondingly (Figure 5). As p53 is a tumor-suppressor gene, increasing its expression may contribute to reversal of the malignant phenotype of HPV16-positive cell lines.

Discussion

HPV oncoprotein E6 plays important roles in maintaining the malignant phenotype of cervical cancer cells.3 Through forming E6/E6AP complex in HPV-infected cells, E6 oncoprotein consequently leads to proteosomal degradation of tumor suppressor p53.14,15 We speculated that when oncoprotein E6 is disrupted, the function of p53 protein will be recovered, leading to cellular apoptosis (Figure 3) and growth inhibition (Figure 4) in HPV16-positive cells.16 Therefore, cancer gene therapy targeting the E6 gene may increase the expression level of the p53 protein and contribute to reversal of the malignant phenotype in cervical cancer cells.

Previous studies have shown that small interfering RNA (siRNA) targeting HPV E6 messenger RNA can effectively knock down E6 expression and induce apoptosis in HPV-positive cervical cancer cells.16 However, the effect of siRNA is transient and only lasts for a few days.17 Moreover, it was also reported that human cells can counteract long-term silencing of critical genes disrupted by siRNA.18 Therefore, in this study, instead of targeting E6 RNA, we applied CRISPR endonuclease to directly disrupt viral E6 DNA (Figure 1). We showed that gRNA-2 and gRNA-3 can efficiently cleave viral E6 DNA and cause DSBs in corresponding areas (Figure 2). These DSBs are then repaired by the error-prone NHEJ repair pathway, creating point mutations or frameshift mutations in the E6 gene.19 Compared to the transient effects of siRNA, one advantage of the CRISPR/Cas system-induced DNA mutation is that it is permanent and could be passed down to the next generation.20 In addition, another advantage of CRISPR-based antiviral therapy is that gRNAs are easy to construct, making this technology easily adopted by less equipped laboratories or companies.

One important concern of CRISPR-based gene therapy for cervical cancer is its specificity toward the HPV DNA sequence. Although the CRISPR/Cas9 system can recognize sequences in the form of 5′-N20NGG-3′,5 the targeting specificity is mainly determined by the 3′-end of the recognition sites, while mismatches at the 5'-end are tolerable.21 In our study, we observed that the activities of HPV16 E6-targeted gRNA/Cas9 were specific enough to induce apoptosis only in HPV16-positive SiHa and CaSki cell lines, but not in HPV16-negative C33A and HEK293 cells (Figure 3). Disruption of the E6 oncogene in these cells led directly to restoration of the tumor suppressor p53, as expected (Figure 5). These data suggested that at least to some extent, our CRISPR/Cas system targeting HPV16 E6 gene is feasible for application as antiviral therapy. We speculate that paired gRNA-guided double nicking of the CRISPR system might further improve the specificity of the HPV16 E6-directed CRISPR/Cas system.22

Figure 5 (AD) Western blot analysis. HPV16 E6 protein and p53 protein in SiHa (A) and CaSki (B) cells were subjected to Western blot at 48 hours after transfection. HPV16 E6:GAPDH and p53:GAPDH ratios in SiHa (C) and CaSki (D) cells were calculated by ImageJ software, and are shown as bar graphs.
Notes: *P<0.05; **P<0.01. One-way analysis of variance. Data represent the means ± standard deviation of two individual experiments. “Blank” represents nontransfected cells.
Abbreviations: HPV, human papillomavirus; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; gRNA, guide ribonucleic acid.

In conclusion, our study demonstrated for the first time that the customized CRISPR gRNA/Cas9 could specifically cleave the E6 oncogene, reverse the malignant phenotype, and increase the expression of p53 in HPV16-positive cervical cancer SiHa and CaSki cell lines. Therefore, the HPV16 E6-targeted CRISPR/Cas system might be used as a valuable strategy for the treatment of HPV-related cervical cancer.

Acknowledgments

This work was supported by funds from the National Development Program (973) for Key Basic Research of China (2009CB521808 and 2013CB911304) and National Natural Science Funding of China (81230038, 81372805, 81301126, 81172466, 81302266, 81402158, and 2013M531699). The authors thank Professor David Segal for pSSA Rep3-1, GZF3-L3, and GZF1-R3 ZFN plasmids.

Disclosure

The authors report no conflicts of interest in this work.


References

1.

Armstrong EP. Prophylaxis of cervical cancer and related cervical disease: a review of the cost-effectiveness of vaccination against oncogenic HPV types. J Manag Care Pharm. 2010;16:217–230.

2.

Clifford GM, Smith JS, Plummer M, Muñoz N, Franceschi S. Human papillomavirus types in invasive cervical cancer worldwide: a meta-analysis. Br J Cancer. 2003;88:63–73.

3.

zur Hausen H. Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer. 2002;2:342–350.

4.

Talora C, Sgroi DC, Crum CP, Dotto GP. Specific down-modulation of Notch1 signaling in cervical cancer cells is required for sustained HPV-E6/E7 expression and late steps of malignant transformation. Genes Dev. 2002;16:2252–2263.

5.

Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science. 2013;339:823–826.

6.

Barrangou R, Fremaux C, Deveau H, et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science. 2007;315:1709–1712.

7.

Katic I, Großhans H. Targeted heritable mutation and gene conversion by Cas9-CRISPR in Caenorhabditis elegans. Genetics. 2013;195:1173–1176.

8.

Hwang WY, Fu Y, Reyon D, et al. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotechnol. 2013;31:227–229.

9.

Rassool FV. DNA double strand breaks (DSB) and non-homologous end joining (NHEJ) pathways in human leukemia. Cancer Lett. 2003;193:1–9.

10.

Santiago Y, Chan E, Liu PQ, et al. Targeted gene knockout in mammalian cells by using engineered zinc-finger nucleases. Proc Natl Acad Sci U S A. 2008;105:5809–5814.

11.

Cho SW, Kim S, Kim JM, Kim JS. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol. 2013;31:230–232.

12.

Bhakta MS, Segal DJ. The generation of zinc finger proteins by modular assembly. Methods Mol Biol. 2010;649:3–30.

13.

Kim HJ, Lee HJ, Kim H, Cho SW, Kim JS. Targeted genome editing in human cells with zinc finger nucleases constructed via modular assembly. Genome Res. 2009;19:1279–1288.

14.

Zanier K, Charbonnier S, Baltzinger M, Nominé Y, Altschuh D, Travé G. Kinetic analysis of the interactions of human papillomavirus E6 oncoproteins with the ubiquitin ligase E6AP using surface plasmon resonance. J Mol Biol. 2005;349:401–412.

15.

Talis AL, Huibregtse JM, Howley PM. The role of E6AP in the regulation of p53 protein levels in human papillomavirus (HPV)-positive and HPV-negative cells. J Biol Chem. 1998;273:6439–6445.

16.

Jonson AL, Rogers LM, Ramakrishnan S, Downs LS Jr. Gene silencing with siRNA targeting E6/E7 as a therapeutic intervention in a mouse model of cervical cancer. Gynecol Oncol. 2008;111:356–364.

17.

Castanotto D, Rossi JJ, The promises and pitfalls of RNA-interference-based therapeutics. Nature. 2009;457:426–433.

18.

Tang S, Tao M, McCoy JP Jr, Zheng ZM. Short-term induction and long-term suppression of HPV16 oncogene silencing by RNA interference in cervical cancer cells. Oncogene. 2006;25:2094–2104.

19.

Heidenreich E, Novotny R, Kneidinger B, Holzmann V, Wintersberger U. Non-homologous end joining as an important mutagenic process in cell cycle-arrested cells. EMBO J. 2003;22:2274–2283.

20.

Li D, Qiu Z, Shao Y, et al. Heritable gene targeting in the mouse and rat using a CRISPR-Cas system. Nat Biotechnol. 2013;31:681–683.

21.

Mussolino C, Cathomen T. RNA guides genome engineering. Nat Biotechnol. 2013;31:208–209.

22.

Ran FA, Hsu PD, Lin CY, et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell. 2013;154:1380–1389.

Creative Commons License © 2014 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.