HPV16 infection regulates RASSF1A transcription mediated by p53

  • Authors:
    • Yueshan Lei
    • Chengjun Hu
    • Hongxin Xu
    • Yihao Tian
  • View Affiliations

  • Published online on: June 17, 2013     https://doi.org/10.3892/mmr.2013.1529
  • Pages: 413-418
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Human papillomavirus (HPV) 16 infection and RASSF1A expression play important roles in tumor development and progression. However, the precise mechanisms underlying their concerted function in the development of reproductive system tumors still remain to be elucidated. In the present study, we showed that HPV16-E6 selectively upregulates RASSF1A expression via degradation of p53, which interacts with the RASSF1A promoter and regulates apoptosis. Overexpression of p53 triggered a decrease in endogenous RASSF1A in SiHa cells, accompanied by apoptosis. Similarly, knockdown of endogenous HPV16-E6 in SiHa cells with RNA interference (RNAi) led to downregulation of RASSF1A mediated by p53 and the subsequent induction of apoptosis. These findings collectively suggest that HPV16 infection regulates p53-mediated RASSF1A expression and suppresses apoptosis. Moreover, RASSF1A may form an element of the negative autoregulatory feedback loops that act on the HPV16 response and are involved in p53-dependent apoptosis. Our results provide novel insights into the cellular mechanism of tumor development, and present a starting point for the development of novel strategies in cancer treatment and effective diagnosis.

Introduction

Human papillomaviruses (HPVs), particularly high-risk types, cause a variety of reproductive system lesions, including cervical neoplasia and cancer. High-risk HPV DNA has been detected in almost all types of cervical cancer, with HPV16 being the most prevalent type in the general population (1). RASSF1A is a tumor suppressor gene with a highly methylated promoter involved in tumorigenesis, development and prognosis (26). Previous studies have shown an association between HPV16 infection and RASSF1A expression, although the precise mechanism of action still remains to be elucidated. HPV infection and inactivation of RASSF1A appear to be inversely correlated in several types of cervical tumors and cell lines. The presence of HPV in cervical carcinomas has been shown to alleviate the requirement for RASSF1A inactivation, and no association with RASSF1A methylation has been observed. This suggests that the two events require other interaction mechanisms but engage the same tumorigenic pathway (7,8).

A previous study demonstrated that one of the HPV16 oncoproteins, E5, activates the vascular endothelial growth factor (VEGF) promoter and upregulates its expression via activation of the epidermal growth factor receptor (EGFR) (9). HPV16-E6 and -E7 are known crucial viral oncoproteins which have been shown to be consistently maintained after viral integration into the host cell genome. The probability of neoplasia is increased in HPV16 infections with E6 and E7 oncoprotein expression (10). To date, both p53-dependent and -independent mechanisms of oncogenesis, regulated by HPV proteins, have been described (11). E6 plays a primary role as an anti-apoptotic protein through association with p53 via interactions with E6-associated protein, and mediation of p53 ubiquitination and degradation that prevents eliciting of cellular responses to stress signals, such as DNA damage. However, underlying interaction mechanisms between HPV16 and the host factor RASSF1A still remain to be fully elucidated. Previous experiments conducted by our group showed that p53 binds to the RASSF1A promoter, leading to downregulation of RASSF1A expression (12). Accordingly, it is hypothesized that HPV16 infection, p53 and RASSF1A are closely interrelated.

The present study aimed to investigate whether HPV16 infection regulates RASSF1A expression as well as to determine the underlying mechanisms of action. Our results provide novel insights into the mechanisms of cancer cell development.

Materials and methods

Cells

The human cervical carcinoma cell line, SiHa, was obtained from the Center for Type Culture Collection (Wuhan, Hubei, China).

Quantitative PCR (qPCR)

Total RNAs were prepared with the TRIzol kit (Invitrogen Life Technologies, Carlsbad, CA, USA) according to the manufacturer’s instructions. All of the RNAs were digested with RNase-free DNase I and purified according to the protocol provided by the manufacturer. In total, ~3 μg RNA was employed as the template for reverse transcription using 0.5 μg oligo(dt) and 200 units of M-MLV reverse transcriptase (Promega, Madison, WI, USA). qPCR was employed for the quantification of gene expression using the multichannel Rotor-Gene 3000 (Corbett Research, Mortlake, Australia) according to the manufacturer’s protocol. PCR cycling conditions were as follows: 5 min at 94°C, followed by 40 cycles of 30 sec at 94°C, 30 sec at 60°C and 30 sec at 72°C in a 25-μl reaction mix containing 1X SYBR-Green I. The primers used were: 5′-TGGAACAACATTAGAACAGC-3′ and 5′-CTGCAA CAAGACATACATCG-3′ for HPV16-E6; 5′-CCTGCT GGATTACATCAAAGCACT-3′ and 5′-GTCAAGGGCA TATCCTACAACAAA-3′ for HPRT. Simultaneous detection of the HPRT gene was performed to normalize HPV16-E6 expression. Similar amplification procedures were employed for HPRT and HPV16-E6. To address robustness issues, each sample was amplified at least in triplicate. Data were analyzed with Rotor-Gene version 6 software and subsequently plotted in Microsoft Excel.

Plasmid constructs
RNA interference (RNAi) clones

siRNA employed for analysis was constructed using the Ambion online siRNA design tool (www.ambion.com/techlib/misc/siRNA_design.html; Ambion, Austin, TX, USA). Hairpin DNA sequences were synthesized as two complementary oligonucleotides, annealed, and ligated between the BbsI and XbaI sites to replace the enhanced green fluorescent protein (EGFP) coding sequence of the pmU6pro vector (kindly provided by Dr David Turner, University of Michigan, USA) for generating the interference vectors, HPV16-E6-RNAi and RASSF1A-RNAi. The sequences were the following: HPV16-E6-RNAi sense, 5′-TTTGAATGTGTGTACTGC AAGCATGGCTTGCAGTACACACATTCTTTTT-3′ and antisense, 3′-TTACACACATGACGTTCGTACCGAACGT CATGTGTGTAAGAAAAAGATC-5′; RASSF1A-RNAi sense, 5′-TTTGACCTCTGTGGCGACTTCAATGTGA AGTCGCCACAGAGGTCTTTTT-3′ and antisense, 3′-TGG AGACACCGCTGAAGTTACACTTCAGCGGTGTCTCCAG AAAAAGATC-5′.

RASSF1A-pcDNA clone

To construct the RASSF1A-pcDNA vector, full-length RASSF1A was amplified from the vector donated by Dr Rongjia Zhou using PCR, and subcloned into the BamHI and EcoRI sites of the pcDNA3.0 mammalian expression vector (BD Biosciences Clontech, Palo Alto, CA, USA). PCR cycling conditions were as follows: 5 min at 94°C, followed by 35 cycles of 1 min at 94°C, 1 min at 65°C and 1 min at 72°C in a 20-μl reaction mix. The sense and antisense primers used for amplification were: 5′-AACG GATCCATGTCGGGGGAGCCTGAGC-3′ and 5′-TACGA ATTCTCACCCAAGGGGGCAGGCG-3′, respectively.

Cell preparation and transfection analysis

The human HPV16-positive cervical cancer cell line SiHa, was obtained from the Center for Type Culture Collection (Wuhan, Hubei, China). The cells were regularly maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 20% fetal bovine serum (FBS; HyClone, Logan, UT, USA) at 37°C with 5% CO2. The cells were passaged every 3 days and seeded onto 24-well plates 12 h prior to transfection. The transfection procedure was performed with Lipofectamine™ 2000 (Invitrogen Life Technologies) according to the manufacturer’s instructions.

Assessment of apoptosis with Annexin V/propidium iodide (PI) staining

Apoptotic cell death was measured using the FITC-conjugated Annexin V/PI assay (BioVision, Palo Alto, CA, USA), followed by flow cytometry (Becton-Dickinson, Franklin Lakes, NJ, USA). Briefly, 1×105 cells were washed with ice-cold phosphate-buffered saline (PBS), resuspended in 0.1 ml binding buffer, and stained with 10 μl FITC-conjugated Annexin V (10 mg/ml) and 10 μl PI (50 mg/ml). After incubation for 15 min at room temperature in the dark, 400 μl binding buffer was added, and the cells were subsequently analyzed with a FACScan flow cytometer (Annexin V excitation at 488 nm and emission at 515 nm; PI excitation at 488 nm and emission at 580 nm).

Western blot analysis

Proteins of freshly obtained SiHa cells were extracted with ice-cold lysis buffer and incubated on ice for 15 min. Following centrifugation for 10 min at 15,000 × g, supernatant fractions were collected, and western blot analysis was performed using routine protocols. Briefly, extracts were analyzed with 12% glycine-SDS-PAGE and transferred onto PVDF membranes with a pore size of 0.2 μm (Hybond-P; Amersham Pharmacia Biotech, Uppsala, Sweden). Nonspecific binding of antibodies was blocked with 5% low-fat milk powder in TBST for 1 h at room temperature. The membranes were incubated with human anti-p53, anti-RASSF1A, anti-HPV16-E6, anti-β-actin (1:500; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) or anti-caspase 3 (1:500; Epitomics, Inc., Burlingame, CA, USA) antibodies at 4°C overnight, followed by incubation with horseradish peroxidase (HRP)-conjugated secondary antibodies (1:50,000) for 1 h. Proteins were visualized with enhanced chemiluminescence (ECL) regent.

DNA binding assay

Previous experiments conducted by our group demonstrated that p53 binds to the RASSF1A promoter and downregulates its expression (12). Since p53 has been suggested to be the critical mediator linking HPV16 infection with RASSF1A expression, we confirmed the active binding site and used a higher dose of p53 up to 2.5 μg to investigate its regulatory effect. The experimental method used was similar to the method reported previously (12).

Immunohistochemistry

Human testicular chips (no. CC23-01) were purchased from Chao Ying Biotechnology (Xi’an, Shaanxi, China). Each chip contained 23 samples, including normal testis, seminoma, lymphoma and fibroma with three slices in the same area for each sample. The expression levels of RASSF1A and p53 proteins were analyzed with anti-RASSF1A (eBioscience, Inc., San Diego, CA, USA) and anti-p53 (Santa Cruz Biotechnology, Inc.) antibodies, respectively, using streptavidin-biotin complex (SABC) and 3,3′-diaminobenzidine (DAB) visualization methods according to the manufacturer’s instructions (Boster Biological Technology, Ltd., Wuhan, China).

Results

HPV16-E6 regulates p53 and RASSF1A levels, and suppresses apoptosis

To gain an insight into the association between HPV16-E6 and RASSF1A expression, the HPV16-E6-RNAi vector was cloned and transfected into SiHa cells containing endogenous HPV16, and RASSF1A expression was detected by western blot analysis. HPV16-E6 expression was decreased by 66% (Fig. 1) upon siRNA transfection, subsequently leading to an increase in p53 and decrease in RASSF1A levels (Fig. 2A). Further examination of the biological effect of E6 RNAi revealed a 29.85% increase in apoptosis (Fig. 2B).

p53 binds to RASSF1A promoter and suppresses RASSF1A expression

According to a previous study conducted by our group, the presence of a p53 binding site in the RASSF1A promoter region was confirmed (12), and a downregulatory effect on RASSF1A was demonstrated upon p53 binding. To validate whether p53 binding is the key factor linking HPV16-E6 and RASSF1A expression, we repeated the experiment with 0–2.5 μg of p53, and examined the binding (Fig. 3A) and regulatory effects (Fig. 3B) of p53. Gel-shift assay showed that His-p53 specifically and efficiently bound to RASSF1A promoter (Fig. 3A, lane 1) which was confirmed by adding the p53 antibody and formed a supershift band (antibody/p53/RASSF1A; Fig. 3A, lane 2). However, single-stranded probes and a mutant RASSF1A probe decreased the formation of the p53/RASSF1A complex (Fig. 3A, lanes 3–7).

p53 inhibits apoptosis induction through RASSF1A regulation

In RASSF1A-expressing SiHa cells, p53 significantly inhibited RASSF1A expression. Consequently, we further investigated the impact of p53 on RASSF1A-induced apoptosis. Flow cytometry combined with Annexin V/PI staining (Fig. 4B and C) revealed that treatment of RASSF1A-expressing SiHa cells with RASSF1A siRNA inhibits apoptosis by 54%, compared to pcDNA. Upon overexpression of RASSF1A, the proportion of apoptotic cells increased from 10.1 to 19.4%, compared with pcDNA, while in the presence of p53, the proportion of apoptotic cells increased to 33.53%. Although both RASSF1A and p53 are apoptosis inducers, overexpression of the two proteins induced a significantly lower increase in the apoptotic cell percentage (35.8%) compared with the expected additive effect (52.93%; 19.4 + 33.53%), indicating that apoptosis induction by RASSF1A is at least partially inhibited by p53. Furthermore, caspase 3 appears to be involved in the apoptotic pathways of p53 and RASSF1A (Fig. 4A).

Mislocalization of p53 and RASSF1A proteins in human testicular tumors

To investigate the potential association between localization of the p53 and RASSF1A proteins and tumorigenesis, we analyzed their expression patterns in human testicular tissue chips. Immunohistochemical analysis using specific antibodies showed p53 and RASSF1A signals in samples of normal testis, spermatocytic seminoma and lymphoma, while not in fibrous tissues (Fig. 5). RASSF1A was weakly expressed in a number of spermatocytic seminoma samples. In spermatocytic seminoma and lymphoma samples, signals were observed around the nuclear membrane, and additionally in nuclei in spermatocytic seminoma cases, compared with mainly cytosolic signals in normal testis. Furthermore, localization of p53 and RASSF1A was coincident in each sample. Our results indicate co-localization of the two proteins, with altered localization patterns in human testicular tumors.

Discussion

Both oncogenes and tumor suppressor genes contribute to the genesis of cancer, which involves multiple genes, including those functioning in DNA repair, signal transduction, apoptosis and cell cycle regulation. For instance, HPV16-E6 and RASSF1A are known oncogenic and tumor suppressor genes that are critical in apoptosis regulation (1316).

The results of the present study indicate a novel function of RASSF1A in the HPV16 pathway. Treatment of cells with HPV16-E6 siRNA led to upregulation of p53 and downregulation of RASSF1A, indicating that RASSF1A acts as an element of the negative autoregulatory feedback loops activated in response to p53. Decreased expression of RASSF1A is known to be sufficient for maintaining a dynamic equilibrium of cell growth and apoptosis, and the high RASSF1A level induced by HPV16 infection could partly counteract tumorigenesis. RASSF1A may play a pivotal role in tumorigenesis, distinct from its earlier reported function as a tumor suppressor. The oncoprotein E6 promotes p53 degradation whose carcinogenic effect is suppressed by RASSF1A. In response to p53, transcriptional networks of p53-responsive genes interact with a number of transduction pathways and positive and negative autoregulatory feedback loops. In the present study, RASSF1A was identified as a novel member of the negative autoregulatory feedback loops. While RASSF1A is a known conventional tumor suppressor, the precise mechanisms by which it interacts with other oncogenes and tumor suppressors remain to be elucidated.

High-risk HPV types, including types 16 and 18, have been identified in ~2/3 of all cervical cancer patients worldwide (17,18). HPV16-E6 binds to and degrades the p53 tumor suppressor protein, leading to malfunction of its DNA repair mechanism (19,20). Previous studies have shown that RASSF1A inactivation and HPV infection are mutually exclusive, and highlight a possible correlation between HPV infection and RASSF1A expression, which may reflect functional interactions between RASSF1A and viral E6 (7,21). One hypothesis is that methylation underlies this correlation. However, in the present study, a novel mechanism is reported where HPV16-E6 regulates RASSF1A transcription mediated via p53 protein. Treatment of cells with HPV16-E6-siRNA led to upregulation of p53 protein, and subsequently, to a decrease in RASSF1A transcription. RASSF1A induces apoptosis and cell cycle alterations via its capability to bind and stabilize the microtubule, control mitosis and regulate genome stability. Specific effector factors include cyclin D1, p120E4F, Cdc20, PMCA4b, Bax, CNK1 and Raf1-MST2 (14,2226). Our experiments demonstrated that p53 and RASSF1A induce apoptosis through caspase 3 activation, maintaining their reported identities as tumor suppressors. However, overexpression of both proteins resulted in significantly lower apoptosis compared to the expected additive effect, indicating an additional potential role of RASSF1A in a feedback regulatory loop to balance cell survival and death.

In summary, our findings provide novel insights into the cellular mechanism of tumor development that might facilitate cancer therapy and diagnosis. Further knowledge of the molecular mechanisms downstream of RASSF1A is required to provide a reference for tumor gene therapy.

Acknowledgements

Funding was provided by the Fundamental Research Funds for Central Universities (274621). The authors thank Professor Rongjia Zhou for kindly providing RASSF1A and p53 expression plasmids.

References

1 

Doorbar J: Molecular biology of human papillomavirus infection and cervical cancer. Clin Sci (Lond). 110:525–541. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Tomizawa Y, Kohno T, Kondo H, et al: Clinicopathological significance of epigenetic inactivation of RASSF1A at 3p21.3 in stage I lung adenocarcinoma. Clin Cancer Res. 8:2362–2368. 2002.PubMed/NCBI

3 

Dammann R, Li C, Yoon JH, Chin PL, et al: Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat Genet. 25:315–319. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Lehmann U, Langer F, Feist H, et al: Quantitative assessment of promoter hypermethylation during breast cancer development. Am J Pathol. 160:605–612. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Xing M, Cohen Y, Mambo E, et al: Early occurrence of RASSF1A hypermethylation and its mutual exclusion with BRAF mutation in thyroid tumorigenesis. Cancer Res. 64:1664–1668. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Yegnasubramanian S, Kowalski J, Gonzalgo ML, et al: Hypermethylation of CpG islands in primary and metastatic human prostate cancer. Cancer Res. 64:1975–1986. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Kuzmin I, Liu L, Dammann R, et al: Inactivation of RAS association domain family 1A gene in cervical carcinomas and the role of human papillomavirus infection. Cancer Res. 63:1888–1893. 2003.PubMed/NCBI

8 

Cohen Y, Singer G, Lavie O, et al: The RASSF1A tumor suppressor gene is commonly inactivated in adenocarcinoma of the uterine cervix. Clin Cancer Res. 9:2981–2984. 2003.PubMed/NCBI

9 

Kim SH, Juhnn YS, Kang S, et al: Human papillomavirus 16 E5 up-regulates the expression of vascular endothelial growth factor through the activation of epidermal growth factor receptor, MEK/ERK1,2 and PI3K/Akt. Cell Mol Life Sci. 63:930–938. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Tan S, Hougardy BM, Meersma GJ, et al: Human papilloma virus 16 E6 RNA interference enhances cisplatin and death receptor-mediated apoptosis in human cervical carcinoma cells. Mol Pharmacol. 81:701–709. 2012. View Article : Google Scholar

11 

Vassallo J, Derchain SF, Pinto GA, Martinez EZ, Syrjänen KJ and Andrade LA: High risk HPV and p53 protein expression in cervical intraepithelial neoplasia. Int J Gynaecol Obstet. 71:45–48. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Tian Y, Hou Y, Zhou X, et al: Tumor suppressor RASSF1A promoter: p53 binding and methylation. PLoS One. 6:e170172011. View Article : Google Scholar : PubMed/NCBI

13 

Preethy CP, Padmapriya R, Periasamy VS, et al: Antiproliferative property of n-hexane and chloroform extracts of Anisomeles malabarica (L). R Br in HPV16-positive human cervical cancer cells. J Pharmacol Pharmacother. 3:26–34. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Matallanas D, Romano D, Yee K, et al: RASSF1A elicits apoptosis through an MST2 pathway directing proapoptotic transcription by the p73 tumor suppressor protein. Mol Cell. 27:962–975. 2007. View Article : Google Scholar

15 

Shivakumar L, Minna J, Sakamaki T, et al: The RASSF1A tumor suppressor blocks cell cycle progression and inhibits cyclin D1 accumulation. Mol Cell Biol. 22:4309–4318. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Song MS, Song SJ, Ayad NG, et al: The tumour suppressor RASSF1A regulates mitosis by inhibiting the APC-Cdc20 complex. Nat Cell Biol. 6:129–137. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Fen J, Yoshinouchi M, Nakamura K, et al: Eradication of HPV post-surgical treatments, its correlation with specific types, types of surgery and the physical status. Oncol Rep. 12:375–379. 2004.PubMed/NCBI

18 

Bosch FX, Manos MM, Muñoz N, et al: Prevalence of human papillomavirus in cervical cancer: a worldwide perspective. International Biological Study on Cervical Cancer (IBSCC) Study Group. J Natl Cancer Inst. 87:796–802. 1995. View Article : Google Scholar

19 

Pöllänen R, Soini Y, Vähäkangas K, et al: Aberrant p53 protein expression in cervical intra-epithelial neoplasia. Histopathology. 23:471–474. 1993.

20 

Boulet G, Horvath C, Vanden Broeck D, et al: Human papilloma virus: E6 and E7 oncogenes. Int J Biochem Cell Biol. 39:2006–2011. 2007. View Article : Google Scholar

21 

Kuzmin I, Gillespie JW, Protopopov A, et al: The RASSF1A tumor suppressor gene is inactivated in prostate tumors and suppresses growth of prostate carcinoma cells. Cancer Res. 62:3498–3502. 2002.PubMed/NCBI

22 

Rong R, Jiang LY, Sheikh MS, et al: Mitotic kinase Aurora-A phosphorylates RASSF1A and modulates RASSF1A-mediated microtubule interaction and M-phase cell cycle regulation. Oncogene. 26:7700–7708. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Ahmed-Choudhury J, Agathanggelou A, Fenton SL, et al: Transcriptional regulation of cyclin A2 by RASSF1A through the enhanced binding of p120E4F to the cyclin A2 promoter. Cancer Res. 65:2690–2697. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Song SJ, Song MS, Kim SJ, et al: Aurora A regulates prome-taphase progression by inhibiting the ability of RASSF1A to suppress APC-Cdc20 activity. Cancer Res. 69:2314–2323. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Baksh S, Tommasi S, Fenton S, et al: The tumor suppressor RASSF1A and MAP-1 link death receptor signaling to Bax conformational change and cell death. Mol Cell. 18:637–650. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Rabizadeh S, Xavier RJ, Ishiguro K, et al: The scaffold protein CNK1 interacts with the tumor suppressor RASSF1A and augments RASSF1A-induced cell death. J Biol Chem. 279:29247–29254. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August 2013
Volume 8 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
Lei Y, Hu C, Xu H and Tian Y: HPV16 infection regulates RASSF1A transcription mediated by p53. Mol Med Rep 8: 413-418, 2013
APA
Lei, Y., Hu, C., Xu, H., & Tian, Y. (2013). HPV16 infection regulates RASSF1A transcription mediated by p53. Molecular Medicine Reports, 8, 413-418. https://doi.org/10.3892/mmr.2013.1529
MLA
Lei, Y., Hu, C., Xu, H., Tian, Y."HPV16 infection regulates RASSF1A transcription mediated by p53". Molecular Medicine Reports 8.2 (2013): 413-418.
Chicago
Lei, Y., Hu, C., Xu, H., Tian, Y."HPV16 infection regulates RASSF1A transcription mediated by p53". Molecular Medicine Reports 8, no. 2 (2013): 413-418. https://doi.org/10.3892/mmr.2013.1529